Sei sulla pagina 1di 570

Obituary

William C. Koller, MD, PhD


19452005

William C. Koller died unexpectedly on October 3, 2005, in Chapel Hill, North Carolina,
while this volume, which he was co-editing, was in preparation. Bill was born in Milwau-
kee on July 12, 1945, where he graduated with a BS degree from Marquette University
in1968. He went on to Northwestern University in Chicago, where he received a Masters
degree in pharmacology in 1971, a PhD in pharmacology in 1974, and an MD in 1976.
After completing his internship and residency at Rush Presbyterian St. Lukes Medical
Center in Chicago, he held positions at the Rush Medical College, University of Illinois,
Chicago VA, Hines VA, and Loyola University. In 1987, he was appointed Professor
and Chairman of Neurology at the University of Kansas Medical Center, where he
remained until 1999, when he moved to the University of Miami and became the National
Research Director for the National Parkinson Foundation. He subsequently moved on to direct the Movement Dis-
orders clinical program at the Mount Sinai Medical Center in New York, and then to the University of North Car-
olina, where he laid the foundation for yet another superb clinical and academic program.
Bill was a world-renowned neurologist who specialized in Parkinsons disease, essential tremor and related
disorders. He published more than 270 peer-reviewed manuscripts, over 160 review papers and numerous books.
His research interests included the epidemiology and experimental therapeutics of parkinsonism and essential
tremor, and his work contributed enormously to the current treatment of these disorders. His collaborations were
worldwide and many current experts in movement disorders worked with him at one time or another. He was
a Fellow of the American Academy of Neurology, Treasurer of the Movement Disorder Society (19992000),
Executive Board Member of the Parkinson Study Group (19961999), President of WE MOVE (20012002),
a founding member of the Tremor Research Group and founder of the International Tremor Foundation.
Dr. Koller will be especially remembered for his humor, warmth and the youthful vigor and enthusiasm that he
brought to his work. He was the consummate physician, befriending many of his patients who were encouraged to
call him on his cell phone at any time. Whether lecturing in South America, fishing on the boat he shared with
several colleagues, traveling with one of his sons to an international meeting or seeing patients in the clinic, Bills
smile and the sparkle in his eye endeared him to all who knew him. The movement disorders community has lost
a valued colleague, mentor and friend. He is survived by his wife and three sons.

Kelly Lyons
Matthew B. Stern

Photo courtesy of Professor Lindsey and


the European Parkinsons Disease Association.
Foreword

The Handbook of Clinical Neurology was started by Pierre Vinken and George Bruyn in the 1960s and continued
under their stewardship until the second series concluded in 2002. This is the fifth volume in the new (third) ser-
ies, for which we have assumed editorial responsibility. The series covers advances in clinical neurology and the
neurosciences and includes a number of new topics. In order to provide insight to physiological and pathogenic
mechanisms and a basis for new therapeutic strategies for neurological disorders, we have specifically ensured
that the neurobiological aspects of the nervous system in health and disease are covered. During the last quar-
ter-century, dramatic advances in the clinical and basic neurosciences have occurred, and those findings related
to the subject matter of individual volumes are emphasized in them. The series will be available electronically
on Elseviers Science Direct site, as well as in print form. It is our hope that this will make it more accessible
to readers and also facilitate searches for specific information.
The present volume deals with Parkinsons disease and related disorders. This group of disorders constitutes
one of the most common of neurodegenerative disorders and is assuming even greater importance with the aging
of the population in developed countries. The volume has been edited by Professor William Koller (USA) and
Professor Eldad Melamed (Israel). It is with particular sadness that we must record the sudden and untimely death
of Professor Koller while the volume was coming to fruition. An experienced clinician, neuroscientist, author and
editor, he was a friend of many of the contributors to this volume, as well as of the series editors, and we shall
greatly miss him. It is our hope that he would have been proud of this volume, which he did so much to craft.
As series editors, we reviewed all of the chapters in the volume and made suggestions for improvement, but we
were delighted that the volume editors had produced such a scholarly and comprehensive account of the parkin-
sonian disorders, which should appeal to clinicians and neuroscientists alike. When the Handbook series was
initiated in the 1960s, understanding of these disorders was poor, any genetic basis of them was speculative, sev-
eral of the syndromes described here had not even been recognized, the prognosis was bleak and the therapeutic
options were almost unchanged since the late Victorian era. Advances in understanding of the biochemical back-
ground of parkinsonism during the 1960s and early 1970s led to dramatic pharmacological advances in the man-
agement of Parkinsons disease and profoundly altered the approach to other degenerative disorders of the nervous
system. The pace of advances in the field has continued, and the exciting new insights being gained have man-
dated a need for a thorough but critical appraisal of recent developments so that future investigative approaches
and therapeutic strategies are based on a solid foundation, the limits of our knowledge are clearly defined and
an account is provided for practitioners of the clinical features and management of the various neurological dis-
orders that present with parkinsonism.
It has been a source of great satisfaction to us that two such eminent colleagues as the late William Koller and
Professor Eldad Melamed agreed to serve as volume editors and have produced such an important compendium,
and we thank them and the contributing authors for all their efforts. We also thank the editorial staff of the pub-
lisher, Elsevier B.V., and especially Ms Lynn Watt and Mr Michael Parkinson in Edinburgh for overseeing all
stages in the preparation of this volume.

Michael J. Aminoff
Francois Boller
Dick F. Swaab
Preface

James Parkinson described Parkinsons disease in his memorable Essay on the Shaking Palsy in 1817. Since then,
and particularly in recent years, there has been tremendous progress in our understanding of this complex and fas-
cinating neurological disorder. Briefly, we have learned that it is not only manifest by motor symptoms but also
that there is a whole range of non-motor features, including autonomic, psychiatric, cognitive and sensory impair-
ments. We now know how to distinguish better clinically between Parkinsons disease and the various parkinso-
nian syndromes. Likewise, it is now well established that in this disorder not only the substantia nigra but many
other central as well as peripheral neuronal cell populations are involved. Novel diagnostic imaging technologies
have become available. The nature of the Lewy body, the intracytoplasmic inclusion body that is a characteristic
element of Parkinsons disease pathology, is being unraveled.
There are new insights in the etiology and pathogenesis of this illness. Experimental models are now available
to understand better modes of neuronal cell death and help develop new therapeutic approaches. There has been
dramatic progress in discovering the genetic causes of dominant and recessive forms of hereditary Parkinsons dis-
ease with the identification of mutations in several genes. There is new knowledge in the intricate circuitry of the
basal ganglia and the physiology of the connections in the healthy state and in Parkinsons disease. There is more
understanding of the role of dopamine and other neurotransmitters in the control and regulation of movement by
the brain.
All of the above led to the development of many novel pharmacological treatments to improve the motor as
well as non-motor phenomena. There is better understanding of the mechanisms responsible for the complications
caused by long-term levodopa administration. Futuristic approaches using deep brain stimulation with electrodes
implanted in anatomically strategic central nervous system sites are now in common use to improve basic symp-
toms and the side-effects of levodopa therapy. Potentially effective neuroprotective strategies are in development
to modify and slow disease progression. Likewise, cell replacement therapy with stem cells offers great promise.
The best of experts in the field joined in this book and contributed chapters that make up an exciting coverage
of all the exhilarating developments in the many aspects of Parkinsons disease. This volume will certainly expand
the current knowledge of its readers and it is also hoped that it will stimulate further research that will eventually
lead to finding both the cause and the cure of this common and disabling neurological disorder.

William C. Koller
Eldad Melamed

Dr. William Koller died suddenly, unexpectedly and prematurely on October 3, 2005, before this volume went to
press. His loss is painful to all his friends and colleagues. His leadership, wisdom and expertise were the main
driving force behind the creation of this very special book. It is the belief of all involved that Dr. Koller would
have been pleased and proud of this volume in its final form. We hope it will be a tribute to his memory.
List of contributors

L. Alvarez R. Bhidayasiri
Movement Disorders Unit, Centro Internacional The Parkinsons and Movement Disorder Institute,
de Restauracion Neurologica (CIREN), La Habana, Fountain Valley, CA, USA
Cuba
R. E. Breeze
M. Baker Department of Neurosurgery, University of Colorado
European Parkinsons Disease Association (EPDA), School of Medicine, Denver, CO, USA
Sevenoaks, Kent, UK
C. Brefel-Courbon
Y. Balash Department of Clinical Pharmacology, Clinical
Movement Disorders Unit, Department of Investigation Centre and Department of
Neurology, Tel-Aviv Sourasky Medical Center, Neurosciences, University Hospital, Toulouse, France
Tel-Aviv, Israel
D. J. Brooks
E. R. Bauminger MRC Clinical Sciences Centre and Division of
Racah Institute of Physics, Hebrew University, Neuroscience and Mental Health, Imperial College
Jerusalem, Israel London, Hammersmith Hospital, London, UK

M. F. Beal R. E. Burke
Department of Neurology and Neuroscience, Weill Departments of Neurology and Pathology, Columbia
Medical College of Cornell University, New York, University, New York, NY, USA
NY, USA
D. J. Burn
P. J. Bedard Institute of Ageing and Health, University of
Centre de Recherche en Neurosciences, CHUL, Newcastle upon Tyne, Newcastle upon Tyne, UK
Faculte de Medicine, Universite Laval, Quebec,
Canada M. G. Cersosimo
Program of Parkinsons Disease and Other Movement
A. Berardelli Disorders, Hospital de Clnicas, University of Buenos
Department of Neurological Sciences and Aires, Buenos Aires, Argentina
Neuromed Institute, Universita La Sapienza,
Rome, Italy A. Chade
The Parkinsons Institute, Sunnyvale, CA, USA
R. Betarbet
Department of Neurology, Emory University, Atlanta, K. R. Chaudhuri
GA, USA Regional Movement Disorders Unit, Kings College
Hospital, London, UK
K. P. Bhatia
Sobell Department of Motor Neuroscience Y. Chen
and Movement Disorders, Institute of Neurology, Morris K. Udall Parkinsons Disease Research Center
University College London, of Excellence, University of Kentucky College of
London, UK Medicine, Lexington, KY, USA
xii LIST OF CONTRIBUTORS
K. L. Chou C. Fox
Department of Clinical Neurosciences, Brown National Center for Voice and Speech, Denver, CO, USA
University Medical School and NeuroHealth
Parkinsons Disease and Movement Disorders Center, S. H. Fox
Warwick, RI, USA Toronto Western Hospital, Movement Disorders
Clinic, Division of Neurology, University of Toronto,
C. Colosimo Toronto, Ontario, Canada
Dipartimento di Scienze Neurologiche, Universita La
Sapienza, Rome, Italy J. Frank
Department of Neurology, Mount Sinai Medical
Y. Compta Center, New York, NY, USA
Neurology Service, Hospital Clinic, University of
Barcelona, Barcelona, Spain C. R. Freed
University of Colorado School of Medicine, Denver,
E. Cubo CO, USA
Unit of Neuroepidemiology, National Centre for
Epidemiology, Carlos III Institute of Health, Madrid, A. Friedman
Spain Department of Neurology, Medical University,
Warsaw, Poland
B. Dass
Department of Neurological Sciences, Rush University J. H. Friedman
Medical Center, Chicago, IL, USA Department of Clinical Neurosciences, Brown
University Medical School and NeuroHealth
M. R. DeLong Parkinsons Disease and Movement Disorders Center,
Department of Neurology, Emory University, Atlanta, Warwick, RI, USA
GA, USA
V. S. C. Fung
G. Deuschl Department of Neurology, Westmead Hospital,
Department of Neurology, Christian-Albrechts- Sydney, NSW, Australia
University, Kiel, Germany
J. Galazka-Friedman
V. Dhawan Faculty of Physics, Warsaw University of Technology,
Regional Movement Disorders Unit, Kings College Warsaw, Poland
Hospital, London, UK
C. Gallagher
Therese Di Paolo Department of Neurobiology, University of
Centre de Recherche en Endocrinologie Moleculaire Wisconsin School of Medicine and Public Health,
et Oncologique, CHUL, Faculte de Pharmacie, Madison, WI, USA
Universite Laval, Quebec, Canada
D. M. Gash
R. Djaldetti Morris K. Udall Parkinsons Disease Research Center
Department of Neurology, Rabin Medical Center, of Excellence, University of Kentucky College of
Petah Tiqva and Sackler Faculty of Medicine, Medicine, Lexington, KY, USA
Tel Aviv University, Tel Aviv, Israel
G. Gerhardt
M. Emre Morris K. Udall Parkinsons Disease Research Center
Department of Neurology, Behavioral Neurology and of Excellence, University of Kentucky College of
Movement Disorders Unit, Istanbul Faculty of Medicine, Lexington, KY, USA
Medicine, Istanbul University, Istanbul, Turkey
O. S. Gershanik
G. Fabbrini Department of Neurology, Centro Neurologico-Hospital
Dipartimento di Scienze Neurologiche, Universita La Frances, Laboratory of Experimental Parkinsonism,
Sapienza, Rome, Italy ININFA-CONICET, Buenos Aires, Argentina
LIST OF CONTRIBUTORS xiii
C. G. Goetz J. S. Hui
Department of Neurological Sciences, Rush University Department of Clinical Neurology, University of
Medical Center, Chicago, IL, USA Southern California, Los Angeles, CA, USA

J. G. Goldman J. Jankovic
Department of Neurological Sciences, Rush University Parkinsons Disease Center and Movement Disorders
Medical Center, Chicago, IL, USA Clinic, Department of Neurology, Baylor College of
Medicine, Houston, TX, USA
D. S. Goldstein
Clinical Neurocardiology Section, National Institute of P. Jenner
Neurological Disorders and Stroke, National Institutes Neurodegenerative Disease Research Center, School
of Health, Bethesda, MD, USA of Health and Biomedical Sciences, Kings College,
London, UK
J.-M. Gracies
Department of Neurology, Mount Sinai Medical M. Kasten
Center, New York, NY, USA The Parkinsons Institute, Sunnyvale, CA, USA

J. T. Greenamyre H. Kaufmann
Department of Neurology, Emory University, Atlanta, Department of Neurology, Mount Sinai School of
GA, USA Medicine, New York, NY, USA

J. Guridi W. C. Kollery
Department of Neurology and Neurosurgery, Department of Neurology, University of North
University Clinic and Medical School and Carolina, NC, USA
Neuroscience Division, University of Navarra and
CIMA, Pamplona, Spain A. D. Korczyn
Sieratzki Chair of Neurology, Tel-Aviv University
T. D. Halbig Medical School, Ramat-Aviv, Israel
Department of Neurology, Mount Sinai School of
Medicine, New York, NY, USA J. H. Kordower
Department of Neurological Sciences, Rush University
N. Hattori Medical Center, Chicago,
Department of Neurology, Juntendo University School IL, USA
of Medicine, Tokyo, Japan
V. Koukouni
Sobell Department of Motor Neuroscience and
M. A. Hely
Movement Disorders, Institute of Neurology,
Department of Neurology, Westmead Hospital,
University College London, London, UK
Sydney, NSW, Australia
A. E. Lang
C. Henchcliffe
Toronto Western Hospital, Movement Disorders
Department of Neurology and Neuroscience, Weill
Clinic, Division of Neurology, University of Toronto,
Medical College of Cornell University, New York,
Toronto, Ontario, Canada
NY, USA
M. Leehey
B. Hogl Department of Neurology, University of Colorado
Department of Neurology, Medical University of School of Medicine, Denver, CO, USA
Innsbruck, Innsbruck, Austria
A. J. Lees
X. Huang Reta Lila Weston Institute of Neurological Studies,
Departments of Neurology and Medicinal Chemistry, University College London, London, UK
University of North Carolina School of Medicine,
Chapel Hill, NC, USA
y
Deceased.
xiv LIST OF CONTRIBUTORS
F. A. Lenz Y. Mizuno
Department of Neurosurgery, Johns Hopkins Hospital, Department of Neurology, Juntendo University School
Baltimore, MD, USA of Medicine, Tokyo, Japan

N. Lev H. Mochizuki
Laboratory of Neuroscience and Department of Department of Neurology, Juntendo University School
Neurology, Rabin Medical Center, Petah-Tikva, of Medicine, Tokyo, Japan
Tel Aviv University, Tel Aviv, Israel
J. C. Moller
M. F. Lew Department of Neurology, Philipps-Universitat
Department of Neurology, University of Southern Marburg, Marburg, Germany
California, Los Angeles, CA, USA
J.-L. Montastruc
M. Lugassy Department of Clinical Pharmacology, Clinical
Department of Neurology, Mount Sinai Medical Investigation Center, University Hospital, Toulouse,
Center, New York, NY, USA France

R. B. Mailman E. B. Montgomery Jr
Departments of Psychiatry, Pharmacology, Neurology National Primate Research Center, University of
and Medicinal Chemistry, University of North Wisconsin-Madison, Madison, WI, USA
Carolina School of Medicine, Chapel Hill, NC, USA
J. G. L. Morris
C. Marin Department of Neurology, Westmead Hospital,
Laboratori de Neurologia Experimental, Fundacio Sydney, NSW, Australia
Clnic-Hospital Clnic, Institut dInvestigacions
Biomediques August Pi i Sunyer (IDIBAPS), Hospital J. A. Obeso
Clinic, Barcelona, Spain Department of Neurology and Neurosurgery,
University Clinic and Medical School and
P. Martnez-Martn Neuroscience Division, University of Navarra and
Unit of Neuroepidemiology, National Centre for CIMA, Pamplona, Spain
Epidemiology, Carlos III Institute of Health, Madrid,
Spain W. H. Oertel
Department of Neurology, Philipps-Universitat Marburg,
I. McKeith Marburg, Germany
Institute for Ageing and Health, Newcastle University,
Newcastle upon Tyne, UK D. Offen
Laboratory of Neuroscience and Department of
K. St. P. McNaught Neurology, Rabin Medical Center, Petah-Tikva, Tel
Department of Neurology, Mount Sinai School of Aviv, University, Tel Aviv, Israel
Medicine, New York, NY, USA
F. Ory-Magne
E. Melamed Department of Neurosciences, University Hospital,
Department of Neurology, Rabin Medical Center, Toulouse, France
Petah Tiqva and Sackler Faculty of Medicine, Tel
Aviv University, Tel Aviv, Israel B. Owler
Department of Neurosurgery, Westmead Hospital,
M. Merello Sydney, NSW, Australia
Movement Disorders Section, Raul Carrea Institute for
Neurological Research, FLENI, Buenos Aires, Argentina D. P. Perl
Mount Sinai School of Medicine, New York, NY, USA
F. E. Micheli
Program of Parkinsons Disease and Other Movement R. F. Pfeiffer
Disorders, Hospital de Clnicas, University of Buenos Department of Neurology, University of Tennessee
Aires, Buenos Aires, Argentina Health Science Center, Memphis, TN, USA
LIST OF CONTRIBUTORS xv
S. Przedborski P. Samadi
Departments of Neurology, Pathology and Cell Centre de Recherche en Endocrinologie Moleculaire et
Biology, Columbia University, New York, NY, USA Oncologie, CHUL, Faculte de Pharmacie, Universite
Laval, Quebec, Canada
J. M. Rabey
Department of Neurology, Assaf Harofeh Medical S. Sapir
Center, Zerifin, Israel Department of Communication Sciences and Disorder,
Faculty of Social Welfare and Health Studies,
A. Rajput University of Haifa, Haifa, Israel
Division of Neurology, Department of Medicine,
University of Saskatchewan, Saskatoon, SK, Canada A. H. V. Schapira
University Department of Clinical Neurosciences,
A. H. Rajput Royal Free and University College Medical School,
Division of Neurology, Department of Medicine, University College London, London, UK
University of Saskatchewan, Saskatoon, SK, Canada
J. Shahed
L. O. Ramig Parkinsons Disease Center and Movement Disorders
Department of Speech, Language and Hearing Clinic, Baylor College of Medicine, Department of
Sciences, University of Colorado-Boulder Department Neurology, Houston, TX, USA
of Speech, and National Center for Voice and Speech,
Denver, CO, USA T. Slaoui
Department of Neurosciences, University Hospital,
J. Rao Toulouse, France
Department of Neurology, Louisiana State University
Health Sciences Center, New Orleans, LA, USA M. B. Stern
Department of Neurology, University of Pennsylvania
O. Rascol School of Medicine, Philadelphia, PA, USA
Department of Clinical Pharmacology, Clinical
Investigation Centre and Department of F. Stocchi
Neurosciences, University Hospital, Toulouse, France Department of Neurology, IRCCS San Raffaele
Pisana, Rome, Italy
J. Rasmussen
Merstham Clinic, Redhill, Surrey, UK N. P. Stover
Department of Neurology, University of Alabama at
W. Regragui Birmingham, Birmingham, AL, USA
Department of Neurosciences, University Hospital,
Toulouse, France C. M. Tanner
The Parkinsons Institute, Sunnyvale, CA, USA
P. F. Riederer
Clinical Neurochemistry, Department of E. Tolosa
Psychiatry and Psychotherapy, National Parkinson Neurology Service, Hospital Clinic, University of
Foundation (USA) Center of Excellence Research Barcelona, Barcelona, Spain
Laboratories, University of Wurzburg, Wurzburg,
Germany C. Trenkwalder
Paracelsus Elena-Klinik, Center of Parkinsonism and
M. C. Rodrguez-Oroz Movement Disorders, Kassel, and University of
Department of Neurology and Neurosurgery, Gottingen, Gottingen, Germany
University Clinic and Medical School and
Neuroscience Division, University of Navarra and D. D. Truong
CIMA, Pamplona, Spain The Parkinsons and Movement Disorder Institute,
Fountain Valley, CA, USA
C. Rouillard
Centre de Recherche en Neurosciences, CHUL, W. Tse
Faculte de Medicine, Universite Laval, Quebec, Department of Neurology, Mount Sinai Medical
Canada Center, New York, NY, USA
xvi LIST OF CONTRIBUTORS
J. Volkmann T. Wichmann
Department of Neurology, Christian-Albrechts- Department of Neurology and Yerkes National
University, Kiel, Germany Primate Center, Emory University,
Atlanta, GA, USA
H. C. Walker
Department of Neurology, University of Alabama at M. B. H. Youdim
Birmingham, Birmingham, AL, USA Department of Pharmacology, Technion-Bruce
Rappaport Faculty of Medicine, Eve Topf and NPF
R. H. Walker Neurodegenerative Diseases Centers, Rappaport
Movement Disorders Clinic, Department of Family Research Institute, Haifa, Israel
Neurology, James J. Peters Veterans Affairs Medical
Center, Bronx, and Department of Neurology, Mount W. M. Zawada
Sinai School of Medicine, New York, NY, USA Division of Clinical Pharmacology, Department of
Medicine, University of Colorado School of Medicine,
R. L. Watts Denver, CO, USA
Department of Neurology, University of Alabama at
Birmingham, Birmingham, AL, USA W. Zhou
Division of Clinical Pharmacology, Department of
D. Weintraub Medicine, University of Colorado School of Medicine,
Departments of Psychiatry and Neurology, University Denver, CO, USA
of Pennsylvania School of Medicine, Philadelphia, PA,
USA
Contents of Part II

Obituary vi
Foreword vii
Preface ix
List of contributors xi

SECTION 5 Treatment of Parkinsons disease

30. Physical therapy in Parkinsons disease 3


Jean-Michel Gracies, Winona Tse, Mara Lugassy and Judith Frank (New York, NY, USA)

31. Neuroprotection in Parkinsons disease: clinical trials 17


Fabrizio Stocchi (Rome, Italy)

32. Levodopa 31
Thomas D. H
albig and William C. Koller (New York, NY and Chapel Hill, NC, USA)

33. Dopamine agonists 73


Olivier Rascol, Tarik Slaoui, Wafa Regragui, Fabiene Ory-Magne,
Christine Brefel-Courbon and Jean-Louis Montastruc (Toulouse, France)

34. Monoamine oxidase A and B inhibitors in Parkinsons disease 93


Moussa B. H. Youdim and Peter F. Riederer (Haifa, Israel and W
urzburg, Germany)

35. Anticholinergic medications 121


Yaroslau Compta and Eduardo Tolosa (Barcelona, Spain)

36. Antiglutamatergic drugs in the treatment of Parkinsons disease 127


Mara Graciela Cers
osimo and Federico Eduardo Micheli (Buenos Aires, Argentina)

37. Investigational drugs 137


Carlo Colosimo and Giovanni Fabbrini (Rome, Italy)

38. The importance of patient groups and collaboration 151


Mary Baker and Jill Rasmussen (Sevenoaks and Redhill, UK)

SECTION 6 Complications of therapy

39. Motor and non-motor fluctuations 159


Susan H. Fox and Anthony E. Lang (Toronto, ON, Canada)
xviii CONTENTS
40. Levodopa-induced dyskinesias in Parkinsons disease 185
Jose A. Obeso, Marcelo Merello, Maria C. Rodrguez-Oroz, Concepci
o Marin, Jorge Guridi and
Lazaro Alvarez (Pamplona and Barcelona, Spain, Buenos Aires, Argentina and La Habana, Cuba)

41. Treatment-induced mental changes in Parkinsons disease 219


Kelvin L. Chou and Joseph H. Friedman (Warwick, RI, USA)

SECTION 7 Surgical treatment

42. Ablative surgery for the treatment of Parkinsons disease 243


Frederick A. Lenz (Baltimore, MD, USA)

43. Deep brain stimulation 261


J. Volkmann and G. Deuschl (Kiel, Germany)

44. Transplantation 279


Curt R. Freed, W. Michael Zawada, Maureen Leehey,
Wenbo Zhou and Robert E. Breeze (Denver, CO, USA)

45. Gene therapy approaches for the treatment of Parkinsons disease 291
Biplob Dass and Jeffrey H. Kordower (Chicago, IL, USA)

SECTION 8 Other parkinsonian syndromes

46. Multiple system atrophy 307


Ronald F. Pfeiffer (Memphis, TN, USA)

47. Progressive supranuclear palsy 327


David J. Burn and Andrew J. Lees (Newcastle upon Tyne and London, UK)

48. Corticobasal degeneration 351


Natividad P. Stover, Harrison C. Walker and Ray L. Watts (Birmingham, AL, USA)

49. Infectious basis to the pathogenesis of Parkinsons disease 373


V. Dhawan and K. Ray Chaudhuri (London, UK)

50. Toxic causes of parkinsonism 385


Nirit Lev, Eldad Melamed and Daniel Offen (Petah-Tikva and Tel-Aviv, Israel)

51. Drug-induced parkinsonism 399


Federico Eduardo Micheli and Mara Graciela Cers
osimo (Buenos Aires, Argentina)

52. Vascular parkinsonism 417


Yacov Balash and Amos D. Korczyn (Tel-Aviv and Ramat-Aviv, Israel)

53. Old age and Parkinsons disease 427


Alex Rajput and Ali H. Rajput (Saskatoon, SK, Canada)

54. Other degenerative processes 445


J. Carsten Moller and Wolfgang H. Oertel (Marburg, Germany)

55. Hydrocephalus and structural lesions 459


John G. L. Morris, Brian Owler, Mariese A. Hely and
Victor S. C. Fung (Sydney, NSW, Australia)
CONTENTS xix
56. Calcification of the basal ganglia 479
Jennifer S. Hui and Mark F. Lew (Los Angeles, CA, USA)

57. Trauma and Parkinsons disease 487


Oscar S. Gershanik (Buenos Aires, Argentina)

58. Psychogenic parkinsonism 501


Vasiliki Koukouni and Kailash P. Bhatia (London, UK)

59. Parkinsonism and dystonia 507


Ruth H. Walker (Bornx and New York, NY, USA)

60. Dementia with Lewy bodies 531


Ian McKeith (Newcastle upon Tyne, UK)

61. Myoclonus and parkinsonism 549


Daniel D. Truong and Roongroj Bhidayasiri (Fountain Valley and Los Angeles, CA,
USA and Bangkok, Thailand)

Subject index 561


Color plate section 571
Section 5

Treatment of Parkinsons disease


Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 30

Physical therapy in Parkinsons disease

JEAN-MICHEL GRACIES*, WINONA TSE, MARA LUGASSY AND JUDITH FRANK

Department of Neurology, Mount Sinai Medical Center, New York, NY, USA

The movement disturbances characteristic of Parkinsons teaching of compensation strategies allowing preser-
disease (PD), such as hypometria, akinesia, rigidity and vation of as much functional independence as possible.
disturbed postural control, can significantly impact func- These strategies include adaptation of the home envir-
tion and quality of life. Typical disabilities resulting from onment, both to lessen the effects of motor impairment
these motor impairments range from dressing or rising and to optimize safety.
from a chair to maintaining balance and initiating gait
(Morris et al., 1995, Morris and Iansek, 1996). Since 30.1. Physical exercises in mild to moderate
the emergence of levodopa in the late 1960s, pharmaco- stages of Parkinsons disease
logic therapy has been the primary strategy to manage
these symptoms and has been considered the gold-stan- Most studies investigating physical exercises have been
dard therapy. However, medication regimens are unable carried out in subjects with mild to moderate PD, i.e. up
to control the disease satisfactorily in the long term, as to Hoehn and Yahr stages 3 (Dietz et al., 1990, Kuroda
dyskinesias, fluctuations of the medication efficacy and et al., 1992, Comella et al., 1994; Bond and Morris,
cognitive difficulties invariably occur after a number of 2000, Marchese et al., 2000, Hirsch et al., 2003).
years (Olanow, 2004). Over the past decades, there has
been increasing awareness as to the potential role of phy- 30.1.1. Metabolic and neuroprotective effects
sical exercise and investigations have been carried out to of physical exercise in Parkinsons disease
evaluate techniques that may alleviate functional disabil-
ities in patients with PD. Despite this rising interest, sur- Exercise intensity may affect dopaminergic metabolism
veys show that only 329% of PD patients regularly in PD. In unmedicated PD patients, 1 hour of strenuous
consult with a paramedical therapist, such as a physical, walking reduces the dopamine transporter availability in
occupational or speech therapist (Deane et al., 2002). the medial striatum (caudate) and in the mesocortical
A large variety of physical therapy methods have dopaminergic system as measured using positron emis-
been evaluated in PD. The approach to therapy in an sion tomography (PET) scans, which has been considered
individual patient, however, may be governed at the highly suggestive of increased endogenous dopamine
most basic level by the stage of the disease. In indivi- release (Ouchi et al., 2001). In addition, exogenous levo-
duals with mild to moderate disease, who are ambula- dopa seems to be better absorbed during moderate-inten-
tory and have retained a certain degree of physical sity endurance exercise, as measured using maximal
independence, therapy may focus on the teaching of levodopa concentrations in plasma (Reuter et al., 2000;
exercises directly designed to delay or prevent the Poulton and Muir, 2005).
aggravation of the motor impairment in PD, with the The beneficial effect of exercise on the dopamine
goal of maintaining or even increasing functional metabolism in PD patients has recently been supported
capacities. At the other end of the spectrum, in an indi- by a number of compelling studies in animal models.
vidual with compromised ambulation and significant Rats exposed to either 1-methyl-4-phenyl-1,2,3,6-tetrahy-
disability due to advanced PD, the therapeutic focus dropyridine (MPTP) or 6-hydroxydopamine (6-OHDA)
may shift from the teaching of exercises to the to induce behavioral and neurochemical loss analogous

*Correspondence to: Jean-Michel Gracies, Department of Neurology, Mount Sinai Medical Center, One Gustave L Levy Place,
Annenberg 2/Box 1052, New York, NY 10029-6574, USA. E-mail: jean-michel.gracies@mssm.edu, Tel: 1-(212)-241-8569,
Fax: 1-(212)-987-7363.
4 J. M. GRACIES ET AL.
to PD lesions that are then exercised show significant 2005). It has been shown that musculotendinous stiff-
sparing of striatal dopamine compared to lesioned ani- ness decreases following strength training in healthy
mals that remain sedentary (Fisher et al., 2004; Poulton elderly individuals (Ochala et al., 2005). In a recent
and Muir, 2005). Further, one study showed that exercise open-label study, 40 Hoehn and Yahr III PD patients
after MPTP exposure increases dopamine D2 transcript and 20 healthy age-matched controls underwent a
expression and downregulates the striatal dopamine 30-day program comprising a variety of physical
transporter (Fisher et al., 2004). However, behavioral therapies including regular physical activity, aerobic
effects of training were inconsistent in these studies strengthening, muscle positioning and lengthening
(Tillerson et al., 2003; Mabandla et al., 2004; Poulton exercises (Stankovic, 2004). Physical therapy resulted
and Muir, 2005). Rodents with unilateral depletion of in significant improvement in tandem stance, one-leg
striatal dopamine display a marked preferential use of stance, step test and external perturbation all tests
the ipsilateral forelimb. After casting of the unaf- of balance in the PD group.
fected forelimb in unilaterally 6-OHDA-lesioned rats, Important risk factors for falls in PD are the muscle
the forced use of the affected forelimb spares its func- atrophy and the decrease in physical conditioning
tion as well as the dopamine remaining in the lesioned that may result from activity reduction (Scandalis
striatum (Faherty et al., 2005). There was a negative et al., 2001). There is a proven relationship between
correlation in this study between the time from lesion decreased lower-limb muscle strength and impaired
to immobilization, i.e. to forced use and the degree of balance in PD, as muscle weakness in the lower extre-
behavioral and neurochemical sparing. This may sug- mities may limit the ability to mount appropriate pos-
gest the importance of initiating an exercise regimen tural adjustments when balance is challenged (Toole
early in the course of PD. et al., 1996). A controlled study addressed the effects
Furthermore, two recent studies have shown that of lower-limb resistance training in PD, in which
exposing animals to an environment prompting exer- patients were randomized to two groups. The first
cise and activity prior to MPTP lesion, or to unilateral group underwent 30-minute sessions three times a
forced limb use prior to contralateral lesioning with week for 10 weeks of standard balance rehabilitation
6-OHDA, may prevent the emergence of the beha- exercises, including practicing standing on foam and
vioral and neurochemical deficits that normally fol- weight-shifting exercises. The second group under-
low the administration of 6-OHDA (Cohen et al., went the same balance training and, in addition,
2003; Faherty et al., 2005). In one of these studies, received tri-weekly high-intensity resistance training
animals receiving a unilateral cast had an increase in sessions focusing on plantar flexion, as well as knee
glial cell-line derived neurotrophic factor (GDNF) pro- extension and flexion. Subjects who received balance
tein in the striatum corresponding to the contralateral training only increased their lower-extremity strength
overused limb. The prevention of parkinsonian deficits (composite score from knee extensor, flexor and plan-
by prior exercise was suggested partly to involve tar flexor strength) by 9%, whereas subjects who
GDNF changes in the striatum (Cohen et al., 2003). underwent additional resistance training increased
their strength by 52%. Balance training only increased
the subjects ability to maintain balance. This effect
30.1.2. Training techniques
was significantly greater and lasted longer in the group
undergoing additional lower-limb resistance training
Several types of exercise techniques have been evalu-
(Hirsch et al., 2003).
ated with regard to their impact on motor deficits in
Improvement in lower-limb muscle strength in PD
mild to moderate PD. Few studies have been con-
may also improve gait. In an open protocol, 14 PD
trolled and much of the evidence is anecdotal or relies
patients and 6 normal controls underwent an 8-week
on open trials. The strongest line of evidence to date
supports the benefit of lower-limb resistance training course of resistance training, twice a week, with
exercises including leg press, calf raise, leg curl,
in PD patients, particularly for balance and gait.
leg extension and abdominal crunches. Lower-limb
strength and gait were assessed in the practically
30.1.2.1. Resistance training defined levodopa off state (i.e. off medication for
Major goals of physical therapy in PD should be the at least 12 hours) before and after the training period,
reduction of rigidity, the improvement of postural showing gains in strength in the PD patients that were
control and the prevention of falls as most PD patients similar to those of the control subjects and improve-
experience balance disturbances and increased risk ment on quantitative measures of gait such as stride
of falls in the course of their illness (Koller et al., length, gait velocity and postural angles (Scandalis
1989; Pelissier and Perennou, 2000; Ochala et al., et al., 2001).
PHYSICAL THERAPY IN PARKINSONS DISEASE 5
30.1.2.2. Attentional strategies and sensory cueing slightly faster than the baseline walking cadence to be
It is a common clinical observation that increased efficacious. PD patients using such rhythmic cues set
attention or effort may allow improvements of remark- at rates of 107.5 and 115% of their baseline walking
able magnitude in motor tasks performed by PD cadence are able to increase the cadence and mean
patients (Muller et al., 1997). This observation has velocity of their gait correspondingly (Howe et al.,
contributed to the development of cueing, an increas- 2003; Suteerawattananon et al., 2004). In contrast, Cubo
ingly prevalent concept in the field of physical therapy and colleagues (2004) found that the use of the metro-
in PD, in which external visual or auditory cues are nome set at the baseline walking cadence slowed ambu-
used to enhance attention and thus performance. It lation and increased the total walking time without
has been hypothesized that the basal ganglia, which having any significant effect on freezing. However,
normally discharge in bursts during the preparation the latter results were obtained in the on state, which
of well-learned motor sequences, provide phasic cues does not allow any conclusions as to the effects of such
to the supplementary motor area, activating and deac- treatment in the off state, when patients typically
tivating the cortical subunits corresponding to a given experience the most slowness and freezing. Another
motor sequence (Morris and Iansek, 1996). In PD specific situation in which sensory cueing may not
however, the internal cues provided by the basal gang- be associated with functional improvements is that in
lia are no longer appropriately supplied. Thus, restora- which patients initiate walking at their maximal speed.
tion of phasic activation of the premotor cortex might Sensory cueing then interferes with movement speed
be facilitated by external means. and performance, which suggests competition between
the external and internal signals of movement command
in situations in which strong internal signals may be
30.1.2.2.1. Auditory cueing
adequate to achieve optimal movement performance
Use of auditory cueing has gained popularity over the (Dibble et al., 2004). In the clinical setting, auditory
past decade (Rubinstein et al., 2002). In the upper cueing is most often used in the form of rhythmic audi-
limb, auditory cues for button-pressing tasks have tory stimulation during gait, in which patients pace their
shown that added external auditory information dra- walking to either a metronome beat or a rhythmic beat
matically reduces initiation and execution time and embedded in music.
improves motor sequencing (Georgiou et al., 1993;
Kritikos et al., 1995). Another study showed that a sin-
gle external auditory cue to start a movement was 30.1.2.2.2. Combination of auditory cueing with
associated with a more forceful, more efficient and attentional strategies
more stable movement than if the movement was Auditory cueing may also be involved in attentional
accomplished only when the patients were ready strategies such as the use of verbal instruction sets.
(internal cue) (Ma et al., 2004). In one controlled study in PD, gait was analyzed dur-
Musical beats, metronomes and rhythmic clapping ing trials of natural walking interspersed with rando-
have been used as cueing techniques to improve gait mized conditions in which subjects were verbally
in PD patients (Thaut et al., 1996; McIntosh et al., instructed to increase arm swing, or step size or walk-
1997). The use of metronome stimulation has been ing speed. In addition to being able to improve any of
shown to reduce acutely the number of steps and the these variables in response to specific instructions,
time to complete a walking course, compared to uncued hearing only one of these instructions was associated
walking in PD patients (Enzensberger et al., 1997). with an improvement in the other gait variables as well
A study of a 3-week home-based gait-training program (Behrman et al., 1998). Conversely, giving an addi-
revealed that PD patients trained with rhythmic auditory tional concurrent task (cognitive or an upper-limb
stimulation in the form of metronome pulsed patterns motor task) to a walking patient worsens gait in PD
embedded into the beat structure of music improved as it may distract attention directed towards gait. This
gait velocity, stride length and step cadence compared is the situation of dual task, which is a classical cause
to subjects receiving gait training without rhythmic of movement deterioration in PD (Brown et al., 1993)
auditory stimulation or no gait training at all (Thaut and more specifically of gait deterioration (Bond and
et al., 1996). Additional research further indicated that Morris, 2000; Hausdorff et al., 2003). Experimental
these gait improvements occur regardless of whether situations combining such dual tasks (reducing atten-
the patient is on or off medication at the time of training tion) with verbal instructions to focus on walking
(McIntosh et al., 1997). The effects of auditory cueing (increasing attention) or on auditory cues tended to
by metronome on gait may depend on the frequency reverse the deterioration and restore better walking
used for the metronome beat, which may have to be (Canning, 2005; Rochester et al., 2005).
6 J. M. GRACIES ET AL.
30.1.2.2.3. Visual cueing improved cadence whereas the visual cue improved
Since classic experiments by Martin (Martin and stride length. However, the simultaneous use of visual
Hurwitz, 1962), a number of studies have shown that and auditory cues did not improve gait significantly
stride length can be improved by visual cueing, in the more than each cue alone.
form of horizontal lines marked on the floor, over which
the patient is encouraged to step. In a series of experi- 30.1.2.3. Active appendicular and axial
ments, Morris et al. (1996) demonstrated that PD patients mobilization, stretch
using such horizontal floor markers as visual cues were Axial mobility may affect function in PD. There is a
able to normalize their stride length, velocity and significant association between reduced axial rotation
cadence, an effect that persisted for 2 hours after the and functional reach (maximal reach without taking a
intervention. It was noted that although transverse lines step forward) independent of disease state (Schenkman
of a color contrasting with the floor and separated by et al., 2000). A controlled study confirmed that physical
an appropriate width are effective for this purpose, zig- intervention targeted on improving spinal flexibility
zag lines or lines parallel to the walking direction are improves functional reach in PD (Schenkman et al.,
not. These visual cues might function by supplying a 1998). An open study suggested that active mobilization
now deficient well-learned motor program with external exercises of the trunk and lower limbs improved trans-
visual information on the appropriate stride length fers (from supine to sitting and sitting to supine), supine
(Rubinstein et al., 2002). Similar improvement in stride rolling and rising from a chair (Viliani et al., 1999).
length and gait velocity is possible using light devices It has been hypothesized that muscle stiffness alone
attached to the chest to provide a visual stimulus on the may be a factor of functional impairment in PD, particu-
floor over which the patient must step (Lewis et al., larly in the lower limb with respect to shortened stride
2000). However, such light devices increase attentional length and altered gait pattern (Lewis et al., 2000).
demand and perceived effort of walking, which suggests Aggressive stretch programs might decrease muscle
that static cues are more effective in improving gait stiffness. However, stretch alone as a therapy techni-
while minimizing effort. Finally, there are suggestions que has not been systematically evaluated as a method
that benefit from cueing techniques might be optimal in to improve motor function in PD. In one controlled
earlier stages of the disease (Lewis et al., 2000). study an improvement in rigidity was observed as
compared to baseline after a therapy program invol-
30.1.2.2.4. Combination of visual cueing with ving passive stretch as well as other motor tasks and
attentional strategies balance training. This effect had disappeared by 2
Gait improvement also occurs when, instead of using months after study completion, which suggests that
horizontal markers on the floor, an attentional strategy standard therapy programs should probably be contin-
is used with instructions to visualize the length of stride ued in the long term, or at least repeated frequently
that subjects should take while walking (Morris et al., (Pacchetti et al., 2000).
1996). Whether gait is assisted by direct visual cueing
or by such attentional visualization strategy, the benefit 30.1.2.4. Treadmill training
is reversed when patients are given additional tasks to Treadmill training has been increasingly used in the
do while walking, distracting attention from the gait rehabilitation of patients with spinal cord injury, hemi-
(Morris et al., 1996). Thus, direct visual cues and visuali- paresis and other gait disorders (Hesse et al., 2003).
zation exercises may both function by focusing attention However, treadmill training may be expensive for some
on the gait, such that walking ceases to be a primarily patients and the specific literature on treadmill training
automatic task delegated to the deficient basal ganglia in PD must be analyzed with particular caution. Some
(Morris et al., 1996). studies have recently suggested that treadmill training
might increase walking speed and stride length in PD
30.1.2.2.5. Combination of visual and auditory cues (Miyai et al., 2000, 2002; Pohl et al., 2003). However,
Suteerawattananon et al. (2004) have studied the effect although these studies compared treadmill with non-
of combining visual and auditory cues to determine the treadmill training, they were not controlled for the walk-
effect of such a combination on gait pattern in PD. In ing speed used in the training. In particular the non-tread-
this study the auditory cue consisted of a metronome mill training did not involve specific requirements of gait
beat 25% faster than the subjects fastest gait cadence. velocity, step cadence or stride length. Under these cir-
Brightly colored parallel lines placed along a walkway cumstances, the positive effects seen after treadmill use
at intervals equal to 40% of the subjects height served may have been the effects of higher energy demands,
as the visual cue. The auditory cueing significantly or a higher walking speed used on the treadmill training
PHYSICAL THERAPY IN PARKINSONS DISEASE 7
(Miyai et al., 2000, 2002; Pohl et al., 2003). In a study In addition to having greater effects on motor function
which did control for walking speed by having subjects than physical therapy without cueing (Thaut et al., 1996;
walk first on the ground and then on a treadmill set at McIntosh et al., 1997), the use of cueing may extend
the same speed, both PD patients and age-matched con- the duration of the effects of therapy (Rubinstein
trols showed shorter stride lengths and higher stride fre- et al., 2002). In a recent single-blind, prospective study,
quency in the treadmill condition (Zijlstra et al., 20 PD patients were randomized to two physical ther-
1998). These effects were more pronounced in the apy groups (Marchese et al., 2000). The first group
PD patients. This is consistent with previous findings underwent a 6-week program of posture control exer-
that, in healthy subjects, walking on a treadmill results cises, passive and active stretch and walking exercises,
in smaller steps than when walking on the ground at whereas the second group combined the same regimen
the same speed (Murray et al., 1985). This is particu- with a variety of visual, auditory and proprioceptive
larly relevant to the PD patient population, in which cueing techniques. Although both groups showed
decreased stride length and impaired stride length regu- improvement on activities of daily living and motor
lation are fundamental characteristics (Morris et al., ability (UPDRS) at the end of the program, the group
1996). Thus, the typical shortening of stride length in without sensory cue training had returned to baseline
PD may in fact be accentuated when walking on a at a 6-week follow-up, whereas the group trained with
treadmill (Zijlstra et al., 1998). cueing techniques was still improved at that visit.
Although it remains uncertain whether all the involved
30.1.3. Long-term effects of physical therapy types of sensory cueing or only specific types were
associated with this benefit, the learning of new motor
The exact duration of the effects of programs of physi- strategies associated with cueing may have caused the
cal exercise in PD remains unknown. Most studies on lasting improvement (Marchese et al., 2000).
physical therapy in PD have been open and had fol-
low-up periods of less than 8 weeks, making the long-
term persistence of beneficial effects difficult to deter- 30.1.4. Emotional arousal, group therapy and use of
mine (Deane et al., 2001). However, in one recent motivational processes
open-label trial 20 PD patients followed a comprehen-
sive rehabilitation program three times a week for Attempts at increasing cortical excitability in PD have
20 weeks. Following the program, there was a signifi- involved, in addition to external sensory inputs, the
cant improvement in Unified Parkinsons Disease use of emotional arousal. A recent open-label study sug-
Rating Scale (UPDRS) activities of daily living and gested improved precision of arm movements as a con-
motor sections scores, self-assessment Parkinsons Dis- sequence of exposure to stimulating music (Bernatzky
ease Disability scale, 10-meter walk test and Zung scale et al., 2004). Pacchetti and colleagues (2000) compared
for depression, which was still seen at the 3-month fol- standard physical therapy (passive stretching exercises,
low-up, suggesting that a sustained motor improvement motor tasks, gait and balance training) with music
can be achieved with a long-term rehabilitation program therapy, involving choral singing, voice exercises and
in PD (Pellecchia et al., 2004). A few previous open rhythmic and free body expression, both administered
studies have similarly suggested motor improvements in weekly group sessions in a randomized, prospective
lasting 6 weeks to 6 months after physical therapy controlled study. The improvement of bradykinesia,
was discontinued (Comella et al., 1994; Reuter et al., emotional well-being, activities of daily living and
1999; Pellecchia et al., 2004). This might underscore quality of life scores was greater in the music therapy
the importance of physical therapy not as one event lim- group, whereas rigidity was the only measure that was
ited in time, but as a continuous or repetitive effort, so more improved in the standard physical therapy group.
that its benefits might be maintained and perhaps These effects dissipated at a 2-month follow-up. The
strengthened over time. improvement in bradykinesia associated with music
Other long-term benefits from exercise in PD have therapy may have resulted from external rhythmic cues
been suggested, involving in particular an increased or from the affective arousal induced by the music,
sense of well-being and an improved quality of life influencing motivational processes (Pacchetti et al.,
(Reuter et al., 1999). One observational study reported 2000).
that mortality was higher by a hazard ratio of 1.8 It has been theorized that physical therapy in group
amongst PD patients who did not exercise regularly com- sessions might enhance socialization and motivation,
pared with patients who did. However, the odds ratios but group therapy has not been systematically compared
were not adjusted for major health factors, such as cardi- with individual therapy and group therapy studies have
ovascular disease, lung disease, smoking or obesity. not been controlled. In their study, Pacchetti and
8 J. M. GRACIES ET AL.
colleagues (2000) evaluated training in a group setting, indeed demonstrated that PD patients can improve per-
both in the physical therapy and music therapy arms. formance on complex motor tasks with intense repeated
The authors did not observe any increase in emotional practice an effect that persists days after the practice
function or quality of life in the physical therapy trials have ended (Soliveri et al., 1992; Behrman
group. In open label studies of group physical therapy et al., 2000). Similar principles of rapid repetition can
in PD, subjects have reported subjective impressions be applied to daily physical exercise.
of benefit in motor symptoms and quality of life, but It has been suggested that such exercises should be
there was no improvement in quantitative measures performed during the levodopa on state, in order to
of motor function (Pedersen et al., 1990; Lokk, 2000; optimize their execution (Koller et al., 1989). However,
Deane et al., 2002). this is not supported by evidence and the opposite strat-
egy may also be suggested, i.e. the performance of phy-
sical exercises during the early morning off phase for
30.2. Recommendations for physical exercise in
example, which might improve dopamine availability
mild to moderate Parkinsons disease
and possibly delay the need for the first daily pill
30.2.1. Schedule (Reuter et al., 2000; Ouchi et al., 2001). Finally, for
exercises to be effectively replicated at home, it is prob-
Although most protocols of the literature have involved ably optimal to teach them as early as possible in the
supervised exercise sessions one to three times per course of the disease.
week (Deane et al., 2002), we recommend that the exer-
cise schedule be intensified and expanded from orga- 30.2.2. Suggested exercises
nized physical therapy sessions into a daily event,
with a time window (11.5 hours) consistently devoted We recommend alternating between two types of exer-
to this activity every day. Controlled studies have cises in a practice session (Figs. 30.1 and 30.2). Active

STRETCHES & EXERCISES UPPER BODY

Lift the weight (bag) forward up & down


Repeat with each arm until fatigued

Stretch shoulder with hand behind wall (e.g. in a doorway)


5 mins. Each side.
Stretch shoulder with elbow leaning against wall as high as
possible.
5 mins. Each side

Lift the weight (bag) to the side, up & down


Repeat with each arm until fatigued
Fig. 30.1. Stretches and exercises for the upper body.
PHYSICAL THERAPY IN PARKINSONS DISEASE 9
exercises should consist of vigorous series of light- elderly patients. Therefore, the weight recommended
resistance rapid alternating movements focused on should cause fatigue optimally after 1020 repeats as
strengthening muscles that open the body (extensors, opposed to fewer than 10 repeats. On the contrary,
abductors, external rotators, supinators and shoulder stretching postures should focus on stretching the mus-
flexors). Passive exercises should involve limb and cle groups that tend to close the body: horizontal
trunk posturing focused on lengthening muscles that and vertical adductors, internal rotators at the shoulder,
close the body (flexors, adductors, internal rotators, flexors and pronators at the elbow, flexors at the wrist
pronators). Following a series of active exercises with and fingers. Ideally, each active exercise should be pur-
passive posturing for a few minutes also allows cardi- sued for about a minute whereas each posture of passive
orespiratory rest. stretch should be maintained for about 5 minutes on
In the upper body, the active resistance exercises each side.
may involve light weight-lifting, particularly focusing In the lower body, stretching postures (passive
on active shoulder abductions and shoulder flexions, exercises) should again focus on muscles such as
as these movements are associated with spinal extensor the hamstrings and hip adductors that tend to adopt a
recruitment (Moseley et al., 2002), which should contri- shortened position in PD because of hypoactivity in
bute to strengthening these muscles (Fig. 30.1). The their antagonists. The active exercises should primarily
spinal extensors are typically hypoactive in PD and focus on sit-to-stand (training trunk and lower-limb
strengthening them should improve trunk posture. extensors) and walking practice (Fig. 30.2).
These exercises should be vigorous so as to provoke a
clear sense of fatigue at the end of the series (Rooney
et al., 1994). In this population we recommend choos- 30.2.2.1. Sit-to-stand
ing weights so as to avoid using maximal or near max- Patients should repeat a series of sit-to-stand exercises
imal intensity training (Khouw and Herbert, 1998) in every day, if possible with arms crossed, ideally as many
order to limit the risk of muscle and tendon strain in times as possible in a continuous series so as to achieve

STRETCHES & EXERCISES LOWER BODY

Stretch by bending forward


5 mins. on each side
Walk the same distance in as few steps as possible.

Stand from sitting position without using hands. Stand with support and stretch legs for 5 mins.
Repeat as many times as possible until fatigued.
Fig. 30.2. Stretches and exercises for the lower body.
10 J. M. GRACIES ET AL.
a sense of fatigue in the primary muscles involved (hip, possibly even to rehearse them mentally as a new task
knee and spinal extensors). This should lead to strengthen- each time, as opposed to just doing them. This corre-
ing of these muscles, which should improve sit-to-stand sponds to a major change in the approach to daily
ability and walking balance. activities. Such change can be facilitated through
a clear understanding by the therapist and/or the
30.2.2.2. Walking care-giver of: (1) the fundamental difference between
Patients should not focus on the speed achieved while automatic and consciously controlled movements;
walking but on their stride length. Ideally the patient and (2) the need to switch to conscious movement
selects a specific distance that should be covered every control for virtually all daily motor activities, particu-
day and counts the steps taken to walk that distance. larly those that we tend to view as the most natural,
Each day, the patient should try to walk the same dis- such as talking, writing, standing up and walking.
tance using as few steps as possible (Fig. 30.2). When We provide examples of these changes in daily strate-
stride length improvement over that distance is maxi- gies below.
mized (i.e. the number of steps taken cannot be further However, teaching and maintaining such discipline
decreased), the same process should be repeated on a can become a highly challenging proposition in advanced
longer distance. In terms of walking in conditions PD, depending on the presence of depression and par-
other than a flat ground, treadmill walking has not ticularly mental deterioration (impairment in executive
been compared to walking in a swimming pool in functions) and on the degree of patient motivation to
controlled studies. However, we would hypothesize improve quality of life (Gracies and Olanow, 2003).
that walking against the viscous resistance of water Depression is a common feature of PD, particularly
should maximize hip flexion and ankle push-off train- in advanced stages (Gracies and Olanow, 2003;
ing and thus better improve stride length than treadmill McDonald et al., 2003) and is characterized by hope-
walking, which might lead to opposite results lessness and pessimism, as well as decreased motiva-
(see above). tion and drive (Brown et al., 1988). This may
undermine the motivation to practice or to change
daily living strategies. Apathy and abulia (lack of
30.3. Compensation strategies in advanced
drive and initiative) can also be prominent symptoms
stages of Parkinsons disease
in PD, independent of depression, and occur with
30.3.1. Role of discipline greater frequency than in patients of similar disability
level from other causes (Pluck and Brown, 2002).
As PD progresses and motor function deteriorates, Most importantly, the gradual emergence of demen-
patients become significantly less mobile and therefore tia, in particular frontal dysexecutive features (perse-
less able to perform daily self-initiated exercises such verations, impulsivity), may hamper the ability to
as active movements against resistance and walking. pursue a strict but slower routine of conscious rehear-
Although the goal of physical therapy remains to sal when performing daily activities that used to be
optimize functional independence, the method gradu- automatic. In these cases, the care-giver often
ally shifts towards teaching strategies to patients becomes the primary focus of the teaching and the
and care-givers to compensate for worsening motor person effectively responsible for implementing the
impairments. Omitting some of these strategies may discipline of the compensation strategies.
jeopardize activities such as walking or swallowing,
with potentially serious consequences. The emphasis 30.3.2. Strategies for freezing episodes
on a strict patient discipline and on the importance of
its enforcement by the care-giver thus becomes even In advanced PD, episodes of freezing, characterized by
more important than in the early stages, as the patient an interruption of motor activity especially when
must now consistently apply the compensation strate- encountering obstacles or constricted spaces, can con-
gies learned in therapy. stitute a significant problem, occurring in up to one-
One fundamental compensatory strategy in advanced third of patients (Giladi et al., 1992). Management
PD involves increasing the amount of attention and may consist primarily of behavioral strategies. As
effort the patient directs toward any given motor mentioned in the previous section, one can teach the
activity. Tasks such as walking, talking, writing and patient how to substitute external auditory, visual, or
standing up are no longer automatic and should proprioceptive cues to replace the deficient internal
no longer be taken for granted. The individual with motor cues normally provided by the basal ganglia
advanced PD must learn to want to perform each of (Morris et al., 1995). Specific attentional strategies
these activities and actively concentrate on them, may also be useful (see below).
PHYSICAL THERAPY IN PARKINSONS DISEASE 11
30.3.2.1. Sensory cues for freezing a narrow exit, entering the doctors office), the
Several techniques have been used to alleviate freezing more difficult the task becomes, particularly as
episodes through external visual input. Visual markers others look on. The emotional stress associated
can be placed in the home in areas where freezing epi- with the social function of walking in these situa-
sodes are common such as doorways and narrow hall- tions makes the freezing episode worse. At the
ways. These may include horizontal markers on the Mount Sinai Movement Disorders Clinic we
floor over which the individual is instructed to step, or attempt to have the patient disconnect for a short
a dot on the wall on which the patient is instructed to while from the social and functional implications
focus in the event of freezing. If the patient is accompa- of walking forward again, and instead to focus
nied, the other person may place a foot in front of the analytically on the walking technique. Walking is
patient, which the patient then steps over (Morris normally a smooth succession of steps. The patient
et al., 1995). Inverted walking sticks, with the handle is asked to focus only on achieving one elemen-
used as a horizontal visual cue at the level of the foot, tary unit of walking, i.e. one step only. One single
have also been investigated as a potential means of step should have minimal social role (since one
improving freezing (Kompoliti et al., 2000). Results step is usually not sufficient to enter or exit a
have been inconsistent, with some subjects showing crowded place) and thus minimal emotional
worsening of freezing while using such a walking stick charge. In practice, the first stage is to stop trying
and others showing improvement (Dietz et al., 1990; to walk. The patient may then take a deep breath
Kompoliti et al., 2000). Additionally, caution must be to mark a pause in the effort and achieve better
used with inverted sticks in PD patients, as these sticks relaxation and spread the feet. Then the patient
may cause tripping and increase the risk of fall. Acous- should concentrate on taking one big step only
tic cues can also be used to decrease freezing. PD and specifically on the power of the hip, knee
patients may carry a metronome, which can be switched and foot flexor movements required to achieve this
on during a freezing episode emitting an auditory one step. Clinical experience shows that when a
beat. Such external cues may be sufficient to initiate strong first step is achieved the second and third
movement. steps naturally follow.
 Movement planning and attention switching back
30.3.2.2. Attentional strategies for freezing from automatic movements to consciously con-
trolled movements. To enhance movement in
A number of adjustments of motor behavior have been
advanced PD, patients can be taught or repeatedly
suggested to alleviate freezing episodes when they occur.
reminded to rehearse mentally each movement
These include:
before it is executed and to pay close attention to
 Focusing on swaying from side to side, transfer- the movement while it is being performed. For
ring body weight from one leg to the other (Morris example, in crowded environments where the risk
et al., 1995) of freezing episodes or tripping increases, the
 Singing, whistling, loudly saying go or left, patient should think ahead and plan the most direct
right, left, right, clapping or saying a rhyme and route through the obstacles. Turning around is
stepping off at the last word are behavioral strate- another difficult task in advanced PD, which may
gies that have the additional advantage of generat- be the most common circumstance causing falls
ing acoustic cues (Morris et al., 1995) in the home setting. Before turning, the patient
 Cue cards posted on the walls of freezing-prone should rehearse the individual leg movements that
areas, with instructions such as go or large step are required to turn the body around effectively.
(Morris et al., 1995) Also, it is recommended to accomplish the turn
 The one-step-only technique strategy of distrac- over a wide arc instead of swiveling (Morris et al.,
tion from the functional or social meaning of the 1995).
action to be accomplished. Success with a method
using a deep-breathing relaxation technique has
been noted in a patient who had failed several 30.3.3. Strategies to minimize postural instability
other techniques to reduce freezing episodes
(Macht and Ellgring, 1999). It is often noted that In general, advanced PD patients have difficulty main-
during an episode of freezing, the more the patient taining balance secondary to slow righting reactions
worries about the functional end-goal of walking (recruitment of the appropriate axial muscles) in response
(freeing the elevator entry for other people to to a challenge to equilibrium. Therefore, patients should
come out, moving out of a crowded store through try to apply the above principle of conscious control of
12 J. M. GRACIES ET AL.
previously automatic tasks to postural balance. Patients defective automatic basal ganglia activation (Morris
may be taught actively to focus their attention on balance et al., 2000). When conscious attention is diverted
whenever they perform an activity involving standing, from the task of maintaining equilibrium, the balan-
similar to what healthy subjects must do when they stand cing deficits may be accentuated. The set-shifting dif-
on a small boat in a turbulent sea. The purpose is to be ficulties commonly seen in advanced PD prevent
in a state of alertness and readiness to respond to threats efficient and rapid switches in concentration between
of balance and thus promptly implement the necessary two motor tasks that must be achieved simultaneously
actions to restore equilibrium (Morris and Iansek, (Gracies and Olanow, 2003).
1996). A recent open-label study suggested that 14-day Not surprisingly, studies have shown that it is ben-
repetitive training of compensatory steps might enhance eficial in PD to avoid performing two tasks simulta-
protective postural responses and shorten the period of neously. In a study comparing PD patients to age-
double support during gait in PD. In addition, significant matched healthy controls, subjects performed two
increases in cadence, step length and gait velocity were walking trials, one freely and one while carrying a tray
observed after such training. These effects were stable with four glasses on it. Whereas the gait performance
for 2 months without additional training (Jobges et al., changed only minimally across conditions in controls,
2004). the PD patients showed decreased walking speed and
stride length while carrying the tray with glasses
30.3.4. Motor subunits (Bond and Morris, 2000). In another study, single set
instructions to increase walking speed, arm swing or
It is beneficial for the PD patient to treat long move- stride length (all contributors to efficient walking)
ment sequences not as a whole, but to break them resulted in improvement not only of the specific vari-
down as a series of component parts or subunits. With able upon which the patient had focused, but in the
this strategy, each subunit is considered and performed other gait variables as well. However, when subjects
as if it were itself a whole movement. This strategy were instructed to count aloud while walking (an
is partly used in the one-step-only technique to allevi- activity that is not a direct component of the walking
ate freezing. Focusing on each subunit of a motor movement), these gait improvements did not occur
sequence may be particularly effective for multijoint (Behrman et al., 1998). In this particular paradigm,
actions such as reaching and grasping, thus facilitating the acoustic cue provided by the loud counting that
activities such as feeding and dressing, or whole-body could have been expected to help walking may have
activities such as standing up from a bed or a chair been counteracted by the distraction from the walking
(Morris and Iansek, 1996). For example, to stand up movements caused by the additional cognitive task.
from a bed, the patient should first mentally rehearse A more recent study confirms that dual-task perfor-
the entire movement and then break the motor mance worsens gait in PD with an equal impairment
sequence down into a series of subunits, including whether the secondary task is motor or cognitive in
bending the knees, turning the head, reaching both nature (OShea et al., 2002).
arms in the desired direction, turning the body, swing- Dual-task performance appears to affect standing
ing the legs over the bed and then finally sitting up balance as well, particularly in PD patients with a pre-
(Morris et al., 1995). vious history of falls (Morris et al., 2000; Marchese
A similar strategy can be used for rising from a et al., 2003). In a study comparing PD patients and
chair. The patient is encouraged to rehearse the age-matched controls there were no differences in pos-
sequence mentally, then to wriggle forward to the front tural stability between groups when subjects simply
of the seat, make sure the feet are placed back under- stood on a platform, but PD patients showed signifi-
neath the chair, lean forward, push on the legs and cantly greater postural instability compared to controls
straighten up the back to stand up. In an open study, when given additional tasks, either cognitive or motor
PD patients trained with these techniques as part of a (Marchese et al., 2003).
6-week physical therapy home regimen showed signif- Therefore PD patients should be instructed to avoid
icant improvement in their ability to transfer in and out carrying out dual tasks and focus on one task at a time.
of chairs and beds (Nieuwboer et al., 2001). For example, while walking, patients should be encour-
aged to avoid carrying objects (the use of backpacks
30.3.5. Avoidance of dual-task performance may be recommended), talking or thinking about other
matters and instead, focus attention towards each
It has been hypothesized that PD patients may mini- individual step and on increasing the stride length
mize their balance or walking difficulties by using (Morris et al., 1995). To prevent loss of balance and falls,
conscious cortically mediated control to overcome PD patients should avoid standing while performing
PHYSICAL THERAPY IN PARKINSONS DISEASE 13
complex motor or cognitive tasks such as showering, during inadvertent backward sways (i.e. appropriately
dressing or conversing (Morris, 2000). timed rectus femoris and tibialis anterior contractions)
and aggravate or even generate a clinical syndrome of
30.3.6. Modification of the home environment retropulsion. This risk has not been measured in a pro-
spective study, but anecdotal evidence has been suffi-
In advanced PD, attention should be paid to the home ciently prevalent in our center and others (Morris
environment, with the goal of maintaining indepen- et al., 1995), that leads us to limit the chronic use of
dence and ensuring safety from falls. The ability to walkers in PD to a minimum in our clinics.
transfer self from bed to chair, chair to toilet and to The use of a cane without objectively verifying
stand up is of primary importance in remaining inde- a positive effect on gait parameters and without provid-
pendent. To assist with difficulties in transferring from ing specific training to the patient is also questionable.
a lying or sitting to a standing position, higher chairs, Patients with PD often handle canes improperly, carry-
toilet seats and beds can be beneficial as they reduce ing them around instead of using them as a support.
the energy requirements to raise the center of gravity. This is particularly problematic in this condition, as
In addition, because narrow constricted spaces and the use of a cane becomes a form of dual task perfor-
obstacles can induce freezing episodes and place indi- mance, involving the simultaneous activities of walking
viduals at risk for tripping and falling, care should be and carrying an object. As described above, performing
taken to create clear, wide spaces with a minimum of additional tasks while walking can result in gait dete-
low-lying obstacles (such as carpets and stools) in rioration. However, it should be recognized that patients
the home setting. Finally, to assist with difficulties may sometimes feel more comfortable using a cane for
with turning in bed, sheets made of satin in the upper walking outdoors or in public places, not for the sup-
part (to allow the body to slide) and of cotton in the posed increase in stability that the cane may provide,
lower part (to allow the heels to grip on it and initiate but as a social signal helping to be recognized by others
the turning movement) may be used. as someone walking slowly or with a handicap.
Whether a cane or a walker is considered, the
30.3.7. Ambulation assistive devices indication should be determined objectively and
accurately: psychological reassurance, social signal,
Although walkers are meant to improve walking sta- objective improvement in stability, reduction in
bility and prevent falls in general and particularly in energy consumption during gait. Whichever indica-
orthopedic conditions, the impact of chronic walker tions are assumed, patients should be tested with
use in PD needs to be critically examined. The argu- and without the assistive device at the clinic to obtain
ments for or against a walker should be weighed on a a rigorous assessment of the acute impact of the device
case-by-case basis, as the use of walkers may worsen on freezing episodes, walking speed and stride length.
gait and increase the risk of tripping or falling Finally, regardless of the acute effects observed at the
(Morris et al., 1995; Kompoliti et al., 2000). A recent clinic, the potential effects of chronic use of these
study evaluated the acute effects of standard walkers devices should also be considered, particularly with
and wheeled walkers as compared to unassisted walk- the use of walkers. An assistive device must not neces-
ing in PD (Cubo et al., 2003). Both wheeled and stan- sarily be used indefinitely. One may consider the
dard walkers significantly slowed gait compared to temporary use of an assistive device in acute periods
unassisted walking, and the standard walker also such as after deep brain stimulation surgery, during a
increased freezing. In addition to potentially exacer- period of intensive medication adjustments with risks
bating posture and balance difficulties, walkers may of walking instability due to excess levodopa and dys-
also become deleterious in individuals whose steps kinesias, or after an orthopedic injury such as a hip
have become faster and shorter: when the frame fracture.
advances too far in front of the feet, the person may
bend over too far and possibly fall (Morris et al., 30.4. Conclusions
1995).
However, the main issue with walkers may not be Interest in physical therapy for patients with PD has
their acute effects on freezing, gait-slowing or the grown over recent years. It should intensify further
possibility of forward falls, but the possibility of with the recent evidence of neuroprotective effects of
long-standing posture and balance impairment caused physical exercises in animal models of PD. Although
by chronic use of these devices. By chronically a number of studies have explored specific treatment
providing passive forward support, walkers may options, they are complicated by heterogeneous treat-
decondition the forward-righting reactions required ment methods, different outcome measures and varying
14 J. M. GRACIES ET AL.
timeframes of analysis (Deane et al., 2002). Many studies
Cohen AD, Tillerson JL, Smith AD et al. (2003). Neuropro-
have also been limited by inadequate randomization tective effects of prior limb use in 6-hydroxydopamine-
methods and lack of convincing sham treatments. The treated rats: possible role of GDNF. J Neurochem 85:
latter two are a particular problem in physical therapy 299305.
research, since neither the therapist nor the patient can Comella CL, Stebbins GT, Brown-Toms N et al. (1994). Phy-
be blinded to the arm of the trial (Deane et al., 2002). sical therapy and Parkinsons disease: a controlled clinical
In the future, larger, randomized, sham-controlled studies trial. Neurology 44: 376378.
with staged follow-up will be necessary to determine for Cubo E, Moore CG, Leurgans S et al. (2003). Wheeled and
each program the benefits, the duration and the appropri- standard walkers in Parkinsons disease patients with gait
ate frequency of training. freezing. Parkinsonism Relat Disord 10: 914.
However, the limitations of the current literature Cubo E, Leurgans S, Goetz CG (2004). Short-term and prac-
should not lead neurologists to underestimate the fun- tice effects of metronome pacing in Parkinsons disease
damental role of daily physical exercise in PD. To patients with gait freezing while in the on state: rando-
mized single blind evaluation. Parkinsonism Relat Disord
optimize motor function, we provide personal recom-
10: 507510.
mendations consisting of strict programs of daily
Deane KH, Jones D, Ellis-Hill C et al. (2001). A comparison
home exercises in the mild to moderate stages of of physiotherapy techniques for patients with Parkinsons
the disease and the teaching to the patient and then disease. Cochrane Database Syst Rev CD002815.
to the care-giver of compensation strategies in the Deane KH, Ellis-Hill C, Jones D et al. (2002). Systematic
late stages. review of paramedical therapies for Parkinsons disease.
Mov Disord 17: 984991.
Acknowledgments Dibble LE, Nicholson DE, Shultz B et al. (2004). Sensory
cueing effects on maximal speed gait initiation in persons
We are grateful to Jerri Chen and Jonathan Alis for with Parkinsons disease and healthy elders. Gait Posture
their excellent work in illustrating some of the exer- 19: 215225.
cises recommended in this chapter. We also thank Dietz MA, Goetz CG, Stebbins GT (1990). Evaluation of a
Sheree Loftus Fader, MSN, APRN, BC, CRRN for her modified inverted walking stick as a treatment for parkin-
expert comments. sonian freezing episodes. Mov Disord 5: 243247.
Enzensberger W, Oberlander U, Stecker K (1997). [Metro-
References nome therapy in patients with Parkinson disease.] Nerve-
narzt 68: 972977.
Faherty CJ, Raviie Shepherd K, Herasimtschuk A et al.
Behrman AL, Teitelbaum P, Cauraugh JH (1998). Verbal
(2005). Environmental enrichment in adulthood eliminates
instructional sets to normalise the temporal and spatial gait
neuronal death in experimental Parkinsonism. Brain Res
variables in Parkinsons disease. J Neurol Neurosurg Psy-
Mol Brain Res 134: 170179.
chiatry 65: 580582.
Fisher BE, Petzinger GM, Nixon K et al. (2004). Exercise-
Behrman AL, Cauraugh JH, Light KE (2000). Practice as an
induced behavioral recovery and neuroplasticity in the
intervention to improve speeded motor performance and
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned
motor learning in Parkinsons disease. J Neurol Sci 174:
mouse basal ganglia. J Neurosci Res 77: 378390.
127136.
Georgiou N, Iansek R, Bradshaw JL et al. (1993). An evalua-
Bernatzky G, Bernatzky P, Hesse HP et al. (2004). Stimulat-
tion of the role of internal cues in the pathogenesis of par-
ing music increases motor coordination in patients
kinsonian hypokinesia. Brain 116 (Pt 6): 15751587.
afflicted with Morbus Parkinson. Neurosci Lett 361:
48. Giladi N, McMahon D, Przedborski S et al. (1992). Motor
blocks in Parkinsons disease. Neurology 42: 333339.
Bond JM, Morris M (2000). Goal-directed secondary motor
tasks: their effects on gait in subjects with Parkinson dis- Gracies JM, Olanow C (2003). Dementia in Parkinsons dis-
ease. Arch Phys Med Rehabil 81: 110116. ease. In: DS Charney, EJ Nestler (Eds.), The Neurobiology
of Mental Illness, 2nd edn. Oxford University Press, Lon-
Brown RG, MacCarthy B, Gotham AM et al. (1988). Depres-
don, pp. 896916.
sion and disability in Parkinsons disease: a follow-up of
132 cases. Psychol Med 18: 4955. Hausdorff JM, Balash J, Giladi N (2003). Effects of cognitive
challenge on gait variability in patients with Parkinsons
Brown RG, Jahanshahi M, Marsden CD (1993). The execu-
disease. J Geriatr Psychiatry Neurol 16: 5358.
tion of bimanual movements in patients with Parkinsons,
Huntingtons and cerebellar disease. J Neurol Neurosurg Hesse S, Werner C, Seibel H et al. (2003). Treadmill training
Psychiatry 56: 295297. with partial body-weight support after total hip arthro-
plasty: a randomized controlled trial. Arch Phys Med
Canning CG (2005). The effect of directing attention during
Rehabil 84: 17671773.
walking under dual-task conditions in Parkinsons disease.
Parkinsonism Relat Disord 11: 9599. Hirsch MA, Toole T, Maitland CG et al. (2003). The effects
of balance training and high-intensity resistance training
PHYSICAL THERAPY IN PARKINSONS DISEASE 15
on persons with idiopathic Parkinsons disease. Arch Phys
Miyai I, Fujimoto Y, Ueda Y et al. (2000). Treadmill train-
Med Rehabil 84: 11091117.
ing with body weight support: its effect on Parkinsons
Howe TE, Lovgreen B, Cody FW et al. (2003). Auditory cues
disease. Arch Phys Med Rehabil 81: 849852.
can modify the gait of persons with early-stage Parkinsons
Miyai I, Fujimoto Y, Yamamoto H et al. (2002). Long-term
disease: a method for enhancing parkinsonian walking
effect of body weight-supported treadmill training in
performance? Clin Rehabil 17: 363367.
Parkinsons disease: a randomized controlled trial. Arch
Jobges M, Heuschkel G, Pretzel C et al. (2004). Repetitive
Phys Med Rehabil 83: 13701373.
training of compensatory steps: a therapeutic approach
Morris M, Iansek R (1996). Characteristics of motor distur-
for postural instability in Parkinsons disease. J Neurol
bance in Parkinsons disease and strategies for movement
Neurosurg Psychiatry 75: 16821687.
rehabilitation. Hum Mov Sci 15: 649669.
Khouw W, Herbert R (1998). Optimisation of isometric
Morris M, Iansek R, Kirkwood B (1995). Moving Ahead
strength training intensity. Aust J Physiother 44 (1): 4346.
with Parkinsons: A Guide to Improving Mobility in Peo-
Koller WC, Glatt S, Vetere-Overfield B et al. (1989). Falls
ple with Parkinsons Kingston Centre, Australia.
and Parkinsons disease. Clin Neuropharmacol 12: 98105.
Morris M, Iansek R, Smithson F et al. (2000). Postural
Kompoliti K, Goetz CG, Leurgans S et al. (2000). On
instability in Parkinsons disease: a comparison with and
freezing in Parkinsons disease: resistance to visual cue
without a concurrent task. Gait Posture 12: 205216.
walking devices. Mov Disord 15: 309312.
Morris ME (2000). Movement disorders in people with
Kritikos A, Leahy C, Bradshaw JL et al. (1995). Contingent
Parkinson disease: a model for physical therapy. Phys
and non-contingent auditory cueing in Parkinsons dis-
Ther 80: 578597.
ease. Neuropsychologia 33: 11931203.
Morris ME, Iansek R, Matyas TA et al. (1996). Stride length
Kuroda K, Tatara K, Takatorige T et al. (1992). Effect of
regulation in Parkinsons disease. Normalization strategies
physical exercise on mortality in patients with Parkinsons
and underlying mechanisms. Brain 119 (Pt 2): 551568.
disease. Acta Neurol Scand 86: 5559.
Moseley GL, Hodges PW, Gandevia SC (2002). Deep and
Lewis GN, Byblow WD, Walt SE (2000). Stride length reg-
superficial fibers of the lumbar multifidus muscle are dif-
ulation in Parkinsons disease: the use of extrinsic, visual
ferentially active during voluntary arm movements. Spine
cues. Brain 123 (Pt 10): 20772090.
27 (2): E29E36.
Lokk J (2000). The effects of mountain exercise in
Muller V, Mohr B, Rosin R et al. (1997). Short-term effects
Parkinsonian personsa preliminary study. Arch Gerontol
of behavioral treatment on movement initiation and pos-
Geriatr 31: 1925.
tural control in Parkinsons disease: a controlled clinical
Ma HI, Trombly CA, Tickle-Degnen L et al. (2004). Effect
study. Mov Disord 12: 306314.
of one single auditory cue on movement kinematics in
Murray MP, Spurr GB, Sepic SB et al. (1985). Treadmill vs.
patients with Parkinsons disease. Am J Phys Med Rehabil
floor walking: kinematics, electromyogram, and heart rate.
83: 530536.
J Appl Physiol 59: 8791.
Mabandla M, Kellaway L, St Clair Gibson A et al. (2004).
Nieuwboer A, De Weerdt W, Dom R et al. (2001). The effect
Voluntary running provides neuroprotection in rats after
of a home physiotherapy program for persons with
6-hydroxydopamine injection into the medial forebrain
Parkinsons disease. J Rehabil Med 33: 266272.
bundle. Metab Brain Dis 19: 4350.
Ochala J, Lambertz D, Van Hoecke J et al. (2005). Effect of
Macht M, Ellgring H (1999). Behavioral analysis of the
strength training on musculotendinous stiffness in elderly
freezing phenomenon in Parkinsons disease: a case study.
individuals. Eur J Appl Physiol 94: 126133.
J Behav Ther Exp Psychiatry 30: 241247.
Olanow CW (2004). The scientific basis for the current treat-
Marchese R, Diverio M, Zucchi F et al. (2000). The role of
ment of Parkinsons disease. Annu Rev Med 55: 4160.
sensory cues in the rehabilitation of parkinsonian patients:
OShea S, Morris ME, Iansek R (2002). Dual task interference
a comparison of two physical therapy protocols. Mov Dis-
during gait in people with Parkinson disease: effects of motor
ord 15: 879883.
versus cognitive secondary tasks. Phys Ther 82: 888897.
Marchese R, Bove M, Abbruzzese G (2003). Effect of cog-
Ouchi Y, Kanno T, Okada H et al. (2001). Changes in dopa-
nitive and motor tasks on postural stability in Parkin-
mine availability in the nigrostriatal and mesocortical
sons disease: a posturographic study. Mov Disord 18:
dopaminergic systems by gait in Parkinsons disease.
652658.
Brain 124: 784792.
Martin JP, Hurwitz LJ (1962). Locomotion and the basal
Pacchetti C, Mancini F, Aglieri R et al. (2000). Active music ther-
ganglia. Brain 85: 261276.
apy in Parkinsons disease: an integrative method for motor
McDonald WM, Richard IH, DeLong MR (2003). Preva-
and emotional rehabilitation. Psychosom Med 62: 386393.
lence, etiology, and treatment of depression in Parkinsons
Pedersen SW, Oberg B, Insulander A et al. (1990). Group
disease. Biol Psychiatry 54: 363375.
training in parkinsonism: quantitative measurements of
McIntosh GC, Brown SH, Rice RR et al. (1997). Rhythmic
treatment. Scand J Rehabil Med 22: 207211.
auditory-motor facilitation of gait patterns in patients with
Pelissier J, Perennou D (2000). [Exercises program and reha-
Parkinsons disease. J Neurol Neurosurg Psychiatry 62:
bilitation of motor disorders in Parkinsons disease]. Rev
2226.
Neurol (Paris) 156 (Suppl 2 Pt 2): 190200.
16 J. M. GRACIES ET AL.
people with Parkinsons disease: a randomized, controlled
Pellecchia MT, Grasso A, Biancardi LG et al. (2004). Physi-
trial. J Am Geriatr Soc 46: 12071216.
cal therapy in Parkinsons disease: an open long-term
Schenkman M, Morey M, Kuchibhatla M (2000). Spinal flex-
rehabilitation trial. J Neurol 251: 595598.
ibility and balance control among community-dwelling
Pluck GC, Brown RG (2002). Apathy in Parkinsons disease.
adults with and without Parkinsons disease. J Gerontol A
J Neurol Neurosurg Psychiatry 73: 636642.
Biol Sci Med Sci 55: M441M445.
Pohl M, Rockstroh G, Ruckriem S et al. (2003). Immediate
Soliveri P, Brown RG, Jahanshahi M et al. (1992). Effect of
effects of speed-dependent treadmill training on gait para-
practice on performance of a skilled motor task in patients
meters in early Parkinsons disease. Arch Phys Med Reha-
with Parkinsons disease. J Neurol Neurosurg Psychiatry
bil 84: 17601766.
55: 454460.
Poulton NP, Muir GD (2005). Treadmill training ameliorates
Stankovic I (2004). The effect of physical therapy on balance
dopamine loss but not behavioral deficits in hemi-parkin-
of patients with Parkinsons disease. Int J Rehabil Res 27:
sonian rats. Exp Neurol 193: 181197.
5357.
Reuter I, Engelhardt M, Stecker K et al. (1999). Therapeutic
Suteerawattananon M, Morris GS, Etnyre BR et al. (2004).
value of exercise training in Parkinsons disease. Med Sci
Effects of visual and auditory cues on gait in individuals
Sports Exerc 31: 15441549.
with Parkinsons disease. J Neurol Sci 219: 6369.
Reuter I, Harder S, Engelhardt M et al. (2000). The effect of
Thaut MH, McIntosh GC, Rice RR et al. (1996). Rhythmic
exercise on pharmacokinetics and pharmacodynamics of
auditory stimulation in gait training for Parkinsons dis-
levodopa. Mov Disord 15: 862868.
ease patients. Mov Disord 11: 193200.
Rochester L, Hetherington V, Jones D et al. (2005). The effect
Tillerson JL, Caudle WM, Reveron ME et al. (2003). Exer-
of external rhythmic cues (auditory and visual) on walking
cise induces behavioral recovery and attenuates neuro-
during a functional task in homes of people with Parkinsons
chemical deficits in rodent models of Parkinsons
disease. Arch Phys Med Rehabil 86: 9991006.
disease. Neuroscience 119: 899911.
Rooney KJ, Herbert RD, Balnave RJ (1994). Fatigue contri-
Toole T, Park S, Hirsch MA et al. (1996). The multicom-
butes to the strength training stimulus. Med Sci Sports
ponent nature of equilibrium in persons with parkinson-
Exerc 26 (9): 11601164.
ism: a regression approach. J Neural Transm 103:
Rubinstein TC, Giladi N, Hausdorff JM (2002). The power of
56180.
cueing to circumvent dopamine deficits: a review of physical
Viliani T, Pasquetti P, Magnolfi S et al. (1999). Effects
therapy treatment of gait disturbances in Parkinsons disease.
of physical training on straightening-up processes in
Mov Disord 17: 11481160.
patients with Parkinsons disease. Disabil Rehabil 21:
Scandalis TA, Bosak A, Berliner JC et al. (2001). Resistance
6873.
training and gait function in patients with Parkinsons dis-
Zijlstra W, Rutgers AW, Van Weerden TW (1998). Volun-
ease. Am J Phys Med Rehabil 80: 3843: quiz 446.
tary and involuntary adaptation of gait in Parkinsons
Schenkman M, Cutson TM, Kuchibhatla M et al. (1998).
disease. Gait Posture 7: 5363.
Exercise to improve spinal flexibility and function for
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 31

Neuroprotection in Parkinsons disease: clinical trials

FABRIZIO STOCCHI*

Department of Neurology, IRCCS San Raffaele Pisana, Roma, Italy

31.1. Introduction PD, as suggested by the association of parkinsonian


syndromes with exposure to neurotoxins such as 1-
Parkinsons disease (PD) is an age-related neurodegen- methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)
erative disorder characterized clinically by resting or hydrocarbons (Langston et al., 1983; Pezzoli et al.,
tremor, rigidity, bradykinesia and a gait disorder. 1996). Further, epidemiological studies indicate that
Pathologically, there is degeneration of nigrostriatal rural living, well-water consumption and exposure to
neurons in the substantia nigra pars compacta (SNpc) pesticides increase the risk of developing PD, whereas
and the presence of intracytoplasmic inclusions known there is a decreased risk of PD associated with smok-
as Lewy bodies. Biochemically, degeneration of ing and coffee consumption (Koller et al., 1990). None
nigrostriatal neurons is associated with a decline in of these factors, however, explain the large majority of
striatal dopamine and this finding is the basis for the cases who appear to have a sporadic form of the dis-
symptomatic treatment of the disorder with the dopa- ease. Indeed, twin studies suggest that genetic factors
mine precursor levodopa. However, in the majority probably play a dominant role in young-onset cases
of patients, levodopa treatment is complicated by where environmental factors are likely to be the more
motor fluctuations, dyskinesia and the development important in most older sporadic cases (Tanner et al.,
of features that do not respond to the drug (e.g. freez- 1999). It is likely that sporadic PD is related to a com-
ing, dementia, autonomic disturbances and postural plex interaction between a variety of genetic and
instability). Further, levodopa does not stop disease environmental factors that may be different in differ-
progression. Consequently, despite the major benefits ent individuals. This implies that there are many dif-
associated with levodopa therapy, the majority of ferent causes of PD and makes it unlikely that a
patients with advanced disease suffer unacceptable single neuroprotective treatment aimed at interfering
levels of functional disability that cannot be controlled with a single etiologic factor will be effective in the
with existing therapies. majority of PD patients.
Because of these limitations, there has been a con- Other opportunities for neuroprotection in PD
certed effort aimed at designing a neuroprotective derive from studies on the pathogenesis and mechan-
therapy for PD (Marsden and Olanow, 1998). Such a ism of cell death. Current information suggests that
treatment can be defined as a treatment or intervention neurodegeneration in PD is associated with a cascade
that slows or stops disease progression by protecting, of events including oxidant stress, mitochondrial
rescuing or restoring the nerve cells that degenerate abnormalities, excitotoxicity and inflammation (Jenner
in PD. Toward this end, an understanding of the etiolo- and Olanow, 1998). Based on this evidence, a number
gic and pathogenetic factors responsible for cell death of theoretical neuroprotective strategies can be
is of critical importance. Both autosomal-dominant designed. What is not clear, however, is whether these
and -recessive gene mutations have been reported to processes are primary or secondary, which if any is the
cause PD (see Ch. 9), but these account for only a driving force that initiates neurodegeneration and what
small number of cases. There is also evidence that role each plays in the neurodegenerative process that
environmental factors contribute to the etiology of occurs in an individual patient. In addition, recent

*Correspondence to: Dr Fabrizio Stocchi, Department of Neurology, IRCCS San Raffaele, Via della Pisana 285, 00165 Rome,
Italy. E-mail: fabrizio.stocchi@fastwebnet.it, Tel: 39-33-690-9255, Fax: 39-06-678-9158.
18 F. STOCCHI
studies suggest that protein aggregation due to failure (MPP) which is responsible for MPTP toxicity (Chiba
of the ubiquitin-proteasome system to clear unwanted et al., 1984). It was reasoned that if PD were related to
proteins may be a common theme in PD neurodegen- an MPTP-like protoxin, MAO-B inhibition with deprenyl
eration, but how to prevent or deal with this problem might similarly prevent the oxidation of other toxins that
is presently not known (McNaught et al., 2001; see contribute to neuronal degeneration. In addition, it was
Ch. 28). Further, considerable evidence supports the also postulated that deprenyl could block the MAO-B-
notion that cell death in PD, regardless of etiology, dependent oxidative metabolism of dopamine and
occurs by way of signal-mediated apoptosis (Hirsch thereby limit free radical damage to SNpc neurons
et al., 1999). The importance of defining a common (Cohen and Spina, 1989).
factor that contributes to neurodegeneration in PD is A prospective, randomized, double-blind, pla-
that it provides an opportunity to develop a single neu- cebo-controlled study involving 800 patients known
roprotective therapy that might interfere with the neu- as the DATATOP study (Deprenyl And Tocopherol
rodegenerative process that ensues as a result of many Antioxidant Therapy Of PD) was performed to
different etiologies and therefore may be of value to a evaluate deprenyl 10 mg/day and tocopherol 2000
large population of PD patients. This chapter addresses IU (Parkinson Study Group, 1989a, b, 1993).
a clinical relevant question regarding the management Untreated PD patients were randomly assigned in a
of PD: are there any therapies that can slow down the 2  2 factorial design to treatment with deprenyl
progression of PD? (10 mg/day), tocopherol (2000 IU), the combination
of both, or placebo. Patients were followed with no
31.2. Trials with antioxidants and monoamine other treatment until they deteriorated to a point that
oxidase-B inhibitors blinded investigators felt that symptomatic therapy
in the form of levodopa needed to be introduced
Considerable evidence supports the notion that oxida- (the primary endpoint). In this study, no effect on
tive stress plays a role in the pathogenesis of cell death the primary endpoint was detected with a-tocopherol
in PD (see Ch. 24). Postmortem studies in the substan- treatment. In contrast, treatment with selegiline sig-
tia nigra of PD patients demonstrate increased levels nificantly delayed the emergence of disability neces-
of iron (which promotes oxidative stress) and sitating treatment with levodopa (P < 0.0001).
decreased levels of glutathione (the major brain anti- After 12 months, levodopa treatment was required
oxidant) (Jenner and Olanow, 1998). Further, there is in 26% of selegiline recipients compared with 47%
evidence of oxidative damage to carbohydrates, lipids, of those who received placebo. Onset of disability
proteins and DNA in the SNpc of PD patients. Addi- necessitating levodopa therapy occurred after a
tionally, the oxidative metabolism of levodopa and/or mean of 26 months in the 375 selegiline recipients
dopamine can generate reactive oxygen species that compared to 15 months in 377 placebo recipients
can induce or aggravate underlying oxidative stress. (with or without tocopherol). Similar results were
These considerations formed the basis for initiating observed with selegiline in a smaller study involving
clinical trials aimed at obtaining a neuroprotective 54 patients that served as a pilot for the DATATOP
effect in PD with antioxidant agents. The initial drugs study (Tetrud and Langston, 1989). Although these
chosen for study were a-tocopherol (vitamin E) and results were impressive and suggested that deprenyl
deprenyl (selegiline, eldepryl). a-Tocopherol was might have a neuroprotective effect, a careful post-
selected because it is the most potent lipid-soluble anti- hoc analysis demonstrated that selegiline was asso-
oxidant in plasma and plasma levels can be increased ciated with both wash-in and wash-out effects indi-
by dietary augmentation (Ingold et al., 1987). Deprenyl cative of a symptomatic effect in addition to any
was selected because in doses of 10 mg/day it is a rela- putative neuroprotective effect. Thus, although there
tively selective inhibitor of monoamine oxidase type B was no doubt that selegiline delayed the emergence
(MAO-B) that avoids the risk of a sympathomimetic cri- of disability in otherwise untreated PD patients, it
sis (the cheese effect) associated with non-selective was unclear as to the responsible mechanism
MAO inhibition (Elsworth et al., 1978). Interest in depre- (Olanow and Calne, 1991). Namely, was the delay
nyl as a possible neuroprotective agent in PD was fos- in requiring levodopa in selegiline-treated patients
tered by the observation that MPTP can induce due to the drug having a neuroprotective effect on
parkinsonism in humans (Langston, 1983) and that remaining dopamine neurons or was it due to the
MPTP-induced parkinsonism in monkeys can be pre- symptomatic effect of the drug masking ongoing
vented by deprenyl (Cohen et al., 1985). In this model, neurodegeneration?
deprenyl inhibits the MAO-B oxidation of MPTP to the This confound remains unresolved, although sub-
pyridinium ion 1-methyl-4-phenylpyridinium ion sequent clinical trials provide some support for the
NEUROPROTECTION IN PARKINSONS DISEASE: CLINICAL TRIALS 19
possibility that the drug may have neuroprotective agents employed. Indeed, Hauser et al. (2000) suggest
effects in addition to its established symptomatic that even 2 weeks of withdrawal from levodopa or
effects. The SELEDO (selegiline-levodopa) study bromocriptine may not be sufficient to eliminate the
evaluated 120 early-stage PD patients who received symptomatic effects associated with their use.
treatment with either levodopa monotherapy or levo- Although the debate continues as to whether or not
dopa plus selegiline (Przuntek et al., 1999). The pri- the drug has protective effects, it is clear that selegiline
mary endpoint was the need for a 50% increase in treatment does not stop disease progression (Elizan
baseline levodopa dosage. Selegiline-treated patients et al., 1989) and several reports suggest that after
were less likely to require this additional increase in many years of treatment patients receiving deprenyl
levodopa, suggesting the possibility that the disease are no less impaired than patients who have not
was progressing at a slower rate. Further, the levo- received the drug (Parkinson Disease Research Group
dopa plus selegiline group had a lower incidence of in the United Kingdom, 1993; Brannan and Yahr,
motor fluctuations despite comparable clinical con- 1995; Parkinson Study Group, 1996a, b). A long-term
trol, again suggesting that selegiline influences the follow-up of the DATATOP patients similarly noted no
natural course of levodopa-treated PD. major difference between those originally on selegiline
The SINDEPAR (Sinemet-Deprenyl-Parlodel) was or placebo, but it is perhaps noteworthy that selegi-
a prospective double-blind study that attempted to line-treated patients had a reduced frequency of freez-
define the putative neuroprotective effects of selegiline ing and off episodes which are thought to be related
in a trial that controlled for the drugs symptomatic to degeneration of non-dopaminergic systems (Shoulson
effects (Olanow et al., 1995). Patients with untreated et al., 2002). The problem has been further confused by
PD (n 100) were randomized to receive treatment the report of an open-label long-term study suggesting
with selegiline versus placebo in addition to treatment that selegiline increases the risk of early death in levo-
with either Sinemet or bromocriptine. Thus, patients dopa-treated PD patients (Lees et al., 1995). There
were randomized to one of four treatment groups; were, however, statistical concerns about how this study
Sinemet plus selegiline, Sinemet plus placebo, bromo- was performed and analyzed (Olanow et al., 1996a) and
criptine plus selegiline, or bromocriptine plus placebo. similar findings were not observed in an assessment of
The primary endpoint of the study was the change in mortality in the DATATOP cohort (Parkinson Study
Unified Parkinsons Disease Rating Scale (UPDRS) Group, 1998) or in a large meta-analysis that included
motor score between original untreated baseline all prospective long-term double-blind controlled trials
and final visit performed 2 months after washout of comparing selegiline to placebo (Olanow et al., 1998b).
selegiline and 7 days after washout of either the Based on the results of the DATATOP study, trials
bromocriptine or the Sinemet. This study showed that of other MAO-B inhibitors were initiated as putative
deterioration in UPDRS score over the course of the neuroprotective agents. Lazabemide is a relatively
study was significantly less in patients randomized to short-acting, reversible and selective MAO-B inhibitor
receive selegiline than in those receiving placebo that is not metabolized to amphetamines or active
regardless of whether they received symptomatic compounds (Cesura et al., 1989). The ability of lazabe-
treatment with levodopa/carbidopa (Sinemet) or mide to influence the progression of disability in
bromocriptine (P < 0.0001). To insure adequate drug untreated PD was assessed in a randomized, multicen-
washout, a subgroup of 23 patients underwent a ter, placebo-controlled, double-blind clinical trial (Par-
14-day washout of their Sinemet or bromocriptine. kinson Study Group, 1996c). A group of 321 untreated
Even with this small sample size, deprenyl-treated PD patients were assigned to one of five treatment
patients had significantly less deterioration from origi- groups (placebo or lazabemide at a dose of 25, 50,
nal baseline than did placebo controls. The authors 100 or 200 mg/day) and followed for up to 1 year.
interpreted these findings as being consistent with the The risk of reaching the primary endpoint (the onset
hypothesis that selegiline has a neuroprotective effect of disability sufficient to require levodopa therapy)
that could not be accounted for by the symptomatic was reduced by 51% for the patients who received
effect of the drug in view of the fact that patients were lazabemide compared with placebo-treated subjects
washed out of selegiline for a relatively long period of (P<0.001). This effect was consistent among all
time while receiving powerful symptomatic agents in dosages and the magnitude and pattern of benefits seen
addition to the study drugs. However, even in this with lazabemide were similar to those observed with
circumstance some doubt remains as it is not possible selegiline treatment in the DATATOP clinical trial.
to state with any certainty that washout was sufficient However, as with selegiline, lazabemide also has
to rid patients of a long-duration symptomatic effect symptomatic effects and there were similar concerns
associated with either selegiline or the symptomatic as to whether or not benefits seen with this drug
20 F. STOCCHI
Randomize
N = 404* N = 380/382 completers N = 306/360 completers
intent-to-treat started next phase elected to continue
(n = 134) (n = 124)
Rasagiline Open-label
1 mg/d 9 terminated 4 terminated Rasagiline 1 mg/d

(n = 132) (n = 124)
Rasagiline
2 mg/d 8 terminated 6 terminated

(n = 138) (n = 132)
Delayed
start Placebo Rasagiline 2 mg/d
5 terminated 10 terminated
Up to 6.5 y from
6 mo 6 mo TEMPO start

Double-blind, Double-blind, Ongoing extension


placebo-controlled rasagiline early vs delayed
Fig. 31.1. TEMPO trial study design.

were due to neuroprotective or symptomatic effects benefit over patients who had their treatment delayed
(Le Witt et al., 1993). This uncertainty caused the by 6 months, as measured by UPDRS-Total score
sponsor to discontinue further trials in PD with this (2.29 unit difference, P 0.01) and UPDRS-ADL
agent (Youdim and Weinstock, 2001). (activities of daily living) score (0.96 unit difference,
P < 0.01). Once again, this was supported by a super-
31.2.1. Rasagiline ior responder rate in the 12-month treatment group
(P < 0.05). Comparisons of other subscales (UPDRS-
Rasagiline is an irreversible and selective MAO-B Motor and -Mental) were not significant. Patients
inhibitor which does not generate amphetamine meta- who received 1 mg/day rasagiline for 12 months also
bolites. The drug proved to be effective in the sympto- showed significant benefits over the 2 mg/day delayed-
matic treatment of early and advanced parkinsonian treatment group in UPDRS-Total score (1.82 unit
patients. However, there are some indications that the difference; P 0.05).
drug may have disease modification effects. A total of One potential disadvantage of this method of mea-
380 patients from the TEMPO trial (Tvp-1012 (rasagi- suring disease modification is that symptomatic effects
line) in early monotherapy for Parkinsons disease out- may be enhanced if treatment is started earlier in the dis-
patients a randomized, double-blind study versus ease course. However, taken as a preliminary indication,
placebo including 404 patients with early PD placebo- this delayed-start study provides promising indications
controlled phase) entered a 6-month phase of active of disease modification that certainly warrant further
treatment, where patients previously receiving placebo investigation. To this end, a new controlled study of
were switched to rasagiline 2 mg/day (Fig. 31.1). The rasagiline, with disease modification as a primary
aim of this phase of the study was a comparison between endpoint, is ongoing ADAGIO-study (Attenuation of
those patients who received 12 months treatment with disease progression with azilect given once-daily).
rasagiline and those patients, previously on placebo,
who had a delayed start and received only 6 months 31.2.2. Antiexcitotoxic agents
treatment with rasagiline. This delayed-start technique
is one of several study designs that have been employed There is substantial evidence suggesting that excess glu-
to evaluate the disease-modifying potential of antipar- tamatergic stimulation with consequent excitotoxicity
kinsonian agents. The theory behind this particular contributes to the neurodegenerative process in PD, rais-
design is that by the end of the full 12 months of study, ing the possibility that antiexcitotoxic drugs might be
the symptomatic effects of treatment will be balanced in neuroprotective (Lipton and Rosenberg, 1994).
all groups, leaving any observed differences attributed Physiologic and metabolic studies demonstrate that
to disease-modifying effects. neuronal activity in the subthalamic nucleus (STN)
Results showed that patients receiving 2 mg/day is increased in parkinsonian animals and humans
rasagiline for a 12-month period had a significant (Obeso et al., 2000). As the STN uses the excitatory
NEUROPROTECTION IN PARKINSONS DISEASE: CLINICAL TRIALS 21
neurotransmitter glutamate as a neurotransmitter and (see Ch. 22). Decreased levels of complex I activity
projects to the internal segment of globus pallidus, the and reduced staining are observed in the substantia
pedunculopontine nucleus and the SNpc, it is reasonable nigra of PD patients and the selective complex I inhi-
to consider that these targets might be subject bitor MPTP induces a levodopa-responsive PD syn-
to excitotoxic damage (Rodriguez et al., 1998). N- drome in both animal models and human patients.
methyl-D-aspartate (NMDA) receptor antagonists have More recently chronic administration of rotenone, a
been reported to protect dopamine neurons from gluta- widely available pesticide which selectively inhibits
mate-mediated toxicity in tissue culture and in rodent complex I, has been shown to be capable of inducing
and primate models of PD (Turski et al., 1991; Greena- a parkinsonian syndrome with selective degeneration
myre et al., 1994; Doble, 1999). Further, there is a retro- of nigral dopaminergic neurons, a reduction in striatal
spective report suggesting that early use of the NMDA dopamine and intracellular inclusions resembling
receptor antagonist amantadine might slow the rate of Lewy bodies (Betarbet et al., 2000). The precise
PD progression (Uitti et al., 1996). A double-blind pla- mechanism whereby inhibition of complex I activity
cebo-controlled dosing study was performed testing the leads to a parkinsonian syndrome is not known and
safety and tolerability of remacemide hydrochloride, a reduced adenosine triphosphate formation, free radical
low-affinity NMDA channel blocker (Parkinson Study generation and reduction in mitochondrial membrane
Group, 2000a). Total daily doses of 150, 300 and 600 potential have all been implicated. These findings do,
mg were not as well tolerated as placebo, but were not however, raise the possibility that bioenergetic agents
associated with serious side-effects and, importantly, such as creatine, coenzyme Q10, ginkgo biloba, nicoti-
symptomatic effects were not detected, although only namide, riboflavin, acetyl-carnitine or lipoic acid
small numbers of patients were studied. However, the might be neuroprotective in PD. Indeed, creatine and
drug failed to provide evidence of a neuroprotective coenzyme Q have been shown to protect dopamine
benefit in patients with Huntingtons disease even when neurons in MPTP-treated rodents (Beal et al., 1998;
combined with coenzyme Q10 and formal tests of neuro- Matthews et al., 1999).
protection in PD have not been carried out (Huntingtons The Parkinson Study Group recently completed a
Disease Study Group, 2000). phase II clinical study of coenzyme Q10 in de novo
Riluzole is another antiglutamate agent that acts PD patients. The study was double-blind and the
by inhibiting sodium channels and thereby prevents patients were randomized to treatment with placebo,
the release of glutamate in overactive glutamatergic 300, 600 or 1200 mg of coenzyme Q10 for 16 months
neurons. The drug is approved for use in the treat- or until disability required levodopa. The primary out-
ment of amyotrophic lateral sclerosis (Bensimon come measure was the change from baseline in the
et al., 1994; Lacomblez et al., 1996) and preclinical UPDRS. In addition, levels of complex I activity
studies have demonstrated the capacity of riluzole to of the mitochondrial electron transport chain and
protect dopamine neurons in rodent and primate mod- coenzyme Q10 levels were obtained. The study demon-
els of PD (Araki et al., 2001; Obinu et al., 2002). A strated a dose-dependent reduction in disease progres-
small open-label pilot study in PD patients demon- sion as assessed by the UPDRS which correlated with
strated that riluzole is well tolerated at doses of 100 increases in plasma coenzyme Q10 levels. Although
mg/day and does not provide symptomatic benefits the results were not statistically significant, a positive
(Jankovic and Hunter, 2002). Morover, despite the trend was observed. The study was designed to deter-
short duration of the trial and the small sample size mine safety and tolerability and only a relatively small
(20 patients), there was a trend toward benefit for number of patients were included; therefore a larger
patients in the riluzole group with riluzole patients phase III study is required. Nonetheless, this study
remaining unchanged over the course of the study supports the notion that bioenergetic agents such as
whereas those randomized to placebo seemed to coenzyme Q10 might offer promise as neuroprotective
deteriorate. A randomized, double-blind, placebo- agents for PD and studies of other such agents are
controlled trial evaluated riluzole 50 mg b.i.d. com- likely to be performed in the near future.
pared to placebo with a primary outcome of change
in UPDRS. No significant difference was found 31.2.4. Antiapoptotic agents
(Jankovic and Hunter, 2002).
Although the issue of neuroprotection with deprenyl
31.2.3. Bioenergetics and other MAO-B inhibitors remains unresolved at the
clinical level, in the laboratory the situation has become
A body of information suggests that mitochondrial much clearer. An increasing body of evidence has now
dysfunction plays a role in the pathogenesis of PD accumulated supporting the capacity of deprenyl to
22 F. STOCCHI
provide neuroprotection for dopaminergic and other There are several different signaling pathways that
motoneurons in a variety of in vitro and in vivo studies can lead to apoptosis depending on the model and
(Mytilineou and Cohen, 1985; Tatton and Greenwood, toxin that are tested, providing different opportunities
1991; Ansari et al., 1993; Roy and Bedard, 1993; Wu for introducing antiapoptotic strategies (Reed, 2002),
et al., 1993, and reviewed in Olanow et al., 1998c). but none has yet been adequately tested in clinical
Further, it has now become clear that MAO-B inhibi- trials or established to be neuroprotective in PD.
tion is not a prerequisite for these benefits (Mytilineou
and Cohen, 1985; Mytilineou et al., 1997a). Indeed, 31.3. Restorative therapies
there is evidence indicating that the neuroprotective
effects seen with deprenyl are due to its metabolite, Whereas most studies have focused on attempts to
desmethyl deprenyl (Mytilineou et al., 1997b, 1998). alter the natural course of PD through manipulation
These benefits are attributed to the propargyl com- of factors thought to be involved in the pathogenesis
ponent of the deprenyl molecule and indeed some of cell death, other approaches have attempted to
other drugs that are propargylamines show a similar restore dopaminergic neuronal function. This has been
effect on glyceraldehyde-3-phosphate dehydrogenase attempted with transplantation strategies designed to
(GAPDH) and similar neuroprotective effects provide new cells to replace those that have under-
(Boulton, 1999; Waldmeir et al., 2000; Youdim and gone neurodegeneration or trophic factors that might
Weinstock, 2001). Data now indicate that neuroprotec- restore or enhance function in remaining dopamine
tion associated with deprenyl is due to an antiapoptotic neurons.
effect related to drug-induced transcriptional events Transplanted fetal nigral cells implanted into the
with the synthesis of new proteins (Tatton et al., 1994, denervated striatum have been demonstrated to sur-
2002). Specifically, deprenyl has been shown to induce vive, manufacture dopamine and provide behavioral
altered expression of a number of genes involved in effects in rodent and primate models of PD (reviewed
apoptosis, including SOD1 and SOD2, glutathione by Olanow et al., 1996b). Open-label studies in
peroxidase, c-JUN, GAPDH, BCL-2 and BAX. These advanced PD patients have reported clinical benefits,
findings suggest that the antiapoptotic properties of increased fluorodopa uptake on positron emission
deprenyl relate to their potential to protect the mito- tomography (PET) and robust survival of implanted
chondrial membrane potential and to exert antioxidant dopamine neurons with organotypic reinnervation of
effects. More recent findings indicate that GAPDH the striatum (reviewed by Olanow et al., 1996b). How-
may be central to these effects. GAPDH is an intermedi- ever, these benefits have not yet been reproduced in
ate in glycogen metabolism and plays an important role double-blind, placebo-controlled trials. Freed et al.
in protein translation and synthesis. However, under (2001) randomized 40 advanced PD patients to receive
certain adverse conditions, GAPDH in its tetrameric transplantation with embryonic ventromesencephalic
form translocates to the nucleus where it interferes with cells or sham surgery. The study failed to meet its
BCL-2 upregulation and promotes apoptosis. Desmethyl primary endpoint (a quality-of-life measure). Improve-
deprenyl binds to GAPDH and maintains it as a dimmer, ment was observed in UPDRS motor scores, particu-
in which form it does not translocate to the nucleus and larly in patients younger than 60 years, but benefits
allows the normal antiapoptotic neuroprotective pro- were not of the magnitude reported in open-label
grams to be initiated (Carlile et al., 2000). Other propar- studies. Further, some of the patients developed severe
gylamines can also provide neuroprotective effects in off-medication dyskinesia which persisted for days
model systems of parkinsonism (Tatton et al., 2000; after withdrawal of levodopa. A similar type of dyski-
Waldmeir et al., 2000; Youdim and Weinstock, 2001). nesia has now been reported by the Swedish group
Two currently undergoing testing for putative neuro- (Hagell et al., 2002). The mechanism responsible for
protective effects in PD are rasagiline (see above) and this unanticipated side-effect is not known, but it is
CGP3466 or TCH346. TCH346 is a more potent neuro- noteworthy that this type of dyskinesia has not been
protectant than deprenyl in laboratory models and does detected in patients with advanced PD who have not
not inhibit MAO-B, which may eliminate a sympto- undergone transplantation (Cubo et al., 2001). This
matic confound and make detection of a neuroprotective study illustrates the importance of placebo-controlled
effect easier to detect. In MPTP monkeys, TCH346 pre- trials (Freeman et al., 1999) and the need to address
vented neuronal degeneration and led to functional clinically relevant issues in the laboratory before
improvement (Waldmeir et al., 2000). Prospective dou- beginning trials in PD patients in order to minimize
ble-blind trials comparing TCH346 with placebo were the risk of running into unanticipated side-effects. In
stopped after an interim analysis for lack of efficacy this regard it should be noted that transplantation has
(Olanow, 2006). not yet been tested in levodopa-treated parkinsonian
NEUROPROTECTION IN PARKINSONS DISEASE: CLINICAL TRIALS 23
monkeys to see if they have been primed to develop transplantation, they can survive and induce behavioral
off-medication dyskinesia. A second double-blind pla- effects in the dopamine-lesioned rodents (Bjorklund
cebo-controlled trial of fetal nigral transplantation has et al., 2002). However, these experiments are not without
just been completed in which different concentrations their own problems and, in one study, 5 of 19 transplanted
of donor tissue were tested, immunosuppression was animals developed tumors (Kim et al., 2002). Stem cells
employed and patients were followed for 2 years have not yet been implanted into PD patients and,
(Freeman et al., 1999), but final results are not yet although they offer considerable promise, many clinical
published. issues remain to be resolved at the preclinical level before
The use of human embryonic neural tissue creates clinical trials in PD patients can be considered.
logistical and societal problems such as acquiring A second restorative approach involves the use of
enough tissue for grafting in a predictable and large- trophic factors. Numerous studies have demonstrated
scale manner and ethical problems inherent in the use the capacity of a variety of trophic factors to protect
of aborted human fetal tissue. For these reason alterna- dopamine neurons in tissue culture and to restore
tive sources of fetal dopaminergic neurons have been nigrostriatal function in dopamine-lesioned animals
sought. One such approach involves the use of porcine (Collier and Sortwell, 1999). Among these, glial-
fetal nigral cells. Implanted porcine fetal nigral cells derived neurotrophic factor (GDNF) appears to be
have been shown to survive and to provide some beha- the most effective in laboratory models (see Ch. 45).
vioral effects. However, open-label trials showed no GDNF treatment has been shown to protect cultured
benefit for patients with Huntingtons disease and only dopamine neurons from a variety of toxins (Lin
minimal benefit in PD patients with no increase in et al., 1993) and to restore function to the nigrostriatal
striatal fluorodopa uptake on PET and only minimal system of the MPTP-lesioned monkey even when
survival at postmortem (Deacon et al., 1997; Fink treatment is delayed for up to several weeks (reviewed
et al., 2000; Schumacher et al., 2000). A double- by Hebert et al., 1999). One of the major limitations of
blind, placebo-controlled study has been performed trophic factors relates to delivery of the protein to the
in patients with advanced PD and demonstrated no target site. An open-label trial of intraventricular
benefit in comparison to patients receiving a placebo GDNF did not provide meaningful benefit to advanced
procedure (unpublished information). In this study PD patients, probably because the protein was not able
the magnitude of the placebo effect was extensive to gain entry from the cerebrospinal fluid into the brain
and sustained, emphasizing again the importance of (Kordower et al., 1999). Side-effects of GDNF deliv-
placebo-controlled trials in evaluating new interven- ered in this way were troublesome and included consi-
tions (Freeman et al., 1999). A small open-label trial titutional complaints, pain and Lhermittes sign,
reported some clinical benefits following transplanta- probably reflecting meningeal irritation. Another
tion of retinal pigmented epithelial cells (Watts, open-label study was subsequently performed in
2002) and a prospective double-blind placebo-con- advanced PD patients in which GDNF was directly
trolled trial has been initiated. injected into the striatum (Barker, 2006). These
Although these studies are not encouraging, it is researchers claimed to observe antiparkinsonian bene-
possible that better results can be obtained with differ- fits and direct infusion of GDNF was better tolerated
ent transplant variables such as increased numbers of than with the intraventricular approach, although 4 of
cells, continued use of immunosuppression, combined 5 patients still experienced Lhermittes sign. A pro-
use of antiapoptotic factors and different transplant tar- spective double-blind placebo-controlled trial testing
gets (Barker, 2002). It is also possible that better continuous infusion of GDNF into the striatum of
results can be obtained with stem cells (see Ch. 44). parkinsonian patients, recently conducted, failed to
Stem cells are capable of self-renewal and expand demonstrate efficacy versus placebo (Lang, 2006). An
after implantation. Neural stem cells are found in the alternate approach utilizes gene therapy to deliver
developing brain (embryonic neural stem cells), but GDNF. Using a modified lentivirus vector to deliver
also in certain sites within the adult brain (Armstrong GDNF to the striatum and nigra of MPTP-lesioned mon-
and Svendsen, 2000; Gage, 2000). Embryonic neural keys, dramatic histologic and behavioral improvement
stem cells can be isolated and grown in culture and was observed (Kordower et al., 2000). Further, the pro-
made to differentiate into all of the major cell types tein was well tolerated. Long-term safety studies in non-
of the central nervous system, including dopaminergic human primates are currently underway and a clinical
neurons (Kawasaki et al., 2002). Indeed, initial studies trial of lentivirus GDNF in PD patients is ongoing.
demonstrate that embryonic stem cells can sponta- Numerous other preclinical studies have been per-
neously differentiate into neurons, with a small percen- formed testing different vectors (e.g. adeno-associated
tage being dopaminergic neurons. Further, following virus), different proteins (e.g. AADC or GAD) and
24 F. STOCCHI
different targets (e.g. SNpc or STN) and it is anticipated function as a primary endpoint. The REAL-PET study
that favorable behavioral results and safety profiles will used striatal fluorodopa uptake on PET whereas the
lead to clinical trials in PD in the not too distant future. CALM-PD study used striatal beta-carbomethoxy-3
Based on the unanticipated off-medication dyskinesia beta-(4-iodophenyl)tropane (b-CIT) uptake on single-
observed in some patients following transplantation, it photon emission computed tomography (SPECT). The
remains necessary to demonstrate that GDNF does not results of two trials have recently been released.
induce dyskinesias and debate continues with regard to In the CALM PD-SPECT study, an estimate of the
whether or not to use a regulatable gene therapy system. rate of dopamine neuronal degeneration was assessed
Nevertheless, based on laboratory studies available to by measuring the rate of decline in b-CIT, a marker
date and the preliminary results with direct infusion in of the dopamine transporter. This study was performed
PD patients, GDNF therapy appears to be very promising in a subsection of patients participating in the CALM-
for PD patients. PD trial (Olanow et al., 1998). A total of 82 patients
Immunophilins are partial ligands for the ciclos- with early PD requiring dopaminergic therapy to treat
porin-binding site that, independent of their immuno- emerging disability were enrolled in the study. Patients
suppressant properties, provide trophic-like effects were randomly assigned to receive pramipexole in a
for dopamine neurons in tissue culture and animal dose of 0.5 mg t.i.d. (n 42), or carbidopa/levodopa
models (Steiner et al., 1997). Two immunophilin 25/100 mg t.i.d. (n 40). The primary outcome vari-
agents have been tested in PD patients, but the results able was the percentage change from baseline in stria-
have been disappointing and no evidence of a neuro- tal b-CIT uptake after 46 months. The mean ( SD)
protective or restorative effect has been observed percentage loss in striatal b-CIT uptake between base-
(Gold and Nutt, 2002). line and 46 months was significantly reduced in the
pramipexole group compared with the levodopa group
31.4. Dopamine agonists (16.0  13.3% versus 25.5  14.1%; P < 0.05). The
authors concluded that patients initially treated with
There has been considerable interest for more than a pramipexole demonstrated a reduced rate of PD pro-
decade in the potential of dopamine agonists to provide gression, as determined by rate of decline in striatal
neuroprotective effects and to alter the natural course b-CIT uptake if treatment was initiated with the dopa-
of the levodopa-treated PD patient (Olanow, 1992; mine agonist. As there was no placebo control group, no
Stocchi, 2000). In the laboratory, dopamine agonists comment could be made as to whether the agonist group
have been shown to protect dopamine neurons from a deteriorated at a slower rate or the levodopa group at a
variety of toxins in both in vitro and in vivo models faster rate. Interestingly, there was no clinical correlate
(Olanow et al., 1998; see Ch. 22). Various theories have of this finding, with patients randomized to initial treat-
emerged to account for these effects, including levo- ment with levodopa doing as well as or better than ago-
dopa-sparing, stimulation of dopamine autoreceptors, nist-treated patients on the UPDRS scale.
direct free radical scavenging, inhibition of STN- The REAL-PET study was designed to compare the
mediated excitotoxicity and activation of dopamine rate of PD progression using striatal fluorodopa uptake
receptors with signal-mediated antiapoptotic effects. on PET as a marker of nigrostriatal function. A total
Two prospective randomized clinical trials have of 186 patients with early untreated PD were rando-
recently been performed in early PD patients to test mized in a 1:1 ratio to initiate treatment with either
the putative neuroprotective effects of dopamine ago- the dopamine agonist ropinirole or levodopa. The pri-
nists. In one study (the Comparison of the agonist pra- mary outcome variable was the percentage reduction
mipexole with levodopa on motor complications of in putamenal 18F-dopa influx constant (Ki) over the
Parkinsons disease, CALM-PD, study), patients with 2-year study in patients with both clinical and PET evi-
untreated PD were randomized to initiate therapy with dence of PD. Data were collected from six PET centers
either the dopamine agonist pramipexole or levodopa and centrally transformed at the Hammersmith Hospital
(Marek et al., 2002). In the second (the Requip as an (London, UK) so as to standardize brain position and
early therapy versus levodopa PET, REAL-PET, study), shape and the placement of the regions of interest
patients were randomized to initiate therapy with the (ROIs) for calculation of putamen Ki. Parametric
agonist ropinirole or levodopa (Whone et al., 2002). images were also interrogated with statistical para-
Open-label levodopa could be added to the blinded ther- metric mapping (SPM) to localize regions of significant
apy if deemed necessary for patients in both studies. In an within-group and between-group differences in rates of
attempt to circumvent the problems of detecting a neuro- loss of dopaminergic function. Secondary endpoints
protective effect with symptomatic agents, both trials uti- included the incidence of dyskinesias and clinical effi-
lized a neuroimaging surrogate marker of nigrostriatal cacy evaluations. The central ROI analysis of putamen
NEUROPROTECTION IN PARKINSONS DISEASE: CLINICAL TRIALS 25
Ki showed that, over the course of the 2-year study, benefit so slight at this early time point that it is
there was a significantly slower rate of deterioration in masked by the symptomatic effects of the drugs and
the ropinirole group as compared to the levodopa group is longer follow-up required?
(13% versus 20%; P < 0.05). Using the SPM analy- These results are most exciting and, independent of
tic technique, a significantly slower rate of progression whether or not dopamine agonists prove to be neuro-
for agonist-treated patients was detected in both puta- protective in the long run, neuroimaging offers the
men and nigra (P < 0.05 for both). The authors con- potential of establishing a surrogate marker of nigros-
cluded that the progression of PD, as reflected by loss triatal function that will enable the assessment of the
of putamenal 18F-dopa uptake, was slower in PD many putative neuroprotective drugs that are currently
patients when treatment was initiated with the dopa- or will eventually become available. Toward this end,
mine agonist ropinirole as compared to levodopa. Inter- it is important to perform careful studies to establish
estingly, this study also found no clinical correlate for that these imaging surrogates do in fact represent accu-
the imaging finding, with UPDRS scores being compar- rate markers of the number of dopamine terminals and/
able or superior in the levodopa group. or neurons and to determine the influence on these
Both studies clearly demonstrate a reduced rate of loss markers of time, drug therapy and disease progression.
of nigrostriatal function, as determined by imaging surro- A delayed start design trial with pramipexole in early
gate markers in patients treated with dopamine agonists PD patients is ongoing.
compared to levodopa. As previously (Parkinson Study
Group, 2000b; Rascol et al., 2000), patients initiated on 31.5. Levodopa
agonists also had a significantly reduced risk of develop-
ing motor complications than did those started on levo- A double-blind controlled randomized trial levodopa
dopa. Interestingly, neither study demonstrated a (150, 300, 600 mg/day) versus placebo including 361
clinical correlate to go along with the improvement patients was recently conducted. The primary outcome
detected in the imaging studies. In both studies clinical was masked assignment of change in UPDRS from
efficacy, as measured by the UPDRS rating scale, favored baseline after 40 weeks of treatment and a 2-week wash-
patients in the levodopa group. This, too, is similar to out. In addition SPECT -CIT was performed at base-
what was described in other trials comparing initial ther- line and week 40 in a subgroup of 116 patients.
apy with dopamine agonists versus levodopa (Parkinson Patients randomized to all levodopa doses had signifi-
Study Group, 2000b; Rascol et al., 2000). Although the cantly better UPDRS scores than patients on placebo,
clinical significance of this difference can be debated, with the greatest improvement seen on the highest dose,
as patients in the agonist groups could receive levodopa showing a clear doseresponse relationship. After the
supplement whenever either the patient or physician washout period none of the three treated groups matched
deemed it was necessary, it is evident that there is no clear the disease severity scored in the placebo group. These
clinical indication of slower disease progression in the data suggested that patients on levodopa had a sustained
agonist groups. functional improvement and this was more evident in
These represent the first studies in PD to demon- the higher-dose group. However, it is possible that the
strate positive results utilizing imaging techniques to washout period was not sufficient to exclude a persistent
seek evidence of neuroprotection. There remain sev- symptomatic effect (long-duration response). Patients
eral issues. Firstly, if benefits seen on imaging reflect on the highest dose of levodopa developed more dyski-
a change in the rate of disease progression, is this a nesias and motor complications. This may be due to
function of a neuroprotective effect of the agonist or the dose of levodopa but also to the way levodopa was
accelerated disease progression due to levodopa? administered (t.i.d.). There was no significant difference
Further studies with a placebo control will be required in -CIT uptake across the groups. However a greater
to address this issue. Secondly, are there pharmacolo- reduction in -CIT uptake was observed in patients
gic effects that interfere with the comparison of levo- taking the higher levodopa dose.
dopa versus a dopamine agonist that give the false
impression that agonists have a slower rate of reduc- 31.6. Conclusion
tion in imaging markers of nigrostriatal function than
does levodopa? Here too, further studies should clarify In establishing that an intervention has a neuroprotective
this issue. Preliminary studies have not shown a speci- effect, it is necessary to choose outcome measures of dis-
fic levodopa or dopamine agonist effect on fluorodopa ease progression that are satisfactory to both clinicians
uptake on PET or -CIT uptake on SPECT. Finally, if and regulatory authorities. To date, no drug has been
the imaging studies do reflect a neuroprotective effect established to be neuroprotective in PD (Morrish, 2002)
of agonists, why is there no clinical correlate? Is the and regulatory agencies have not yet confirmed that any
26 F. STOCCHI
of the outcome measures that have been used are in the treatment of Parkinsons disease. Ann Neurol 37:
acceptable for purposes of drug approval and labeling. 9598.
Several clinical trials aimed at detecting a neuropro- Carlile GW, Chalmers-Redman RME, Tatton NA et al.
tective effect have shown positive results, but interpre- (2000). Reduced apoptosis after nerve growth factor and
tation has been confounded by the symptomatic effects serum withdrawal: conversion of tetrameric glyceralde-
hydes-3-phosphate dehydrogenase to a dimmer. J Pharma-
of the drugs being tested. As a consequence, positive
col Exp Ther 57: 212.
results cannot be interpreted as representing neuropro- Cesura AM, Galva MD, Imhof R et al. (1989). [3H]Ro
tection. More recently, clinical trials have attempted to 19-6327: a reversible ligand and affinity labelling
detect neuroprotection by utilizing neuroimaging probe for monoamine oxidase-B. Eur J Pharmacol 162:
surrogate markers of nigrostriatal function. However, 457465.
even though results have been positive, it is not estab- Chiba K, Trevor A, Castagnoli N Jr (1984). Metabolism of
lished that these imaging surrogate markers do in fact the neurotoxic tertiary amine, MPTP, by brain monoamine
accurately represent the number of dopamine neurons or oxidase. Biochem Biophys Res Commun 120: 457478.
terminals and no clinical correlate has been observed in Cohen G, Spina MB (1989). L-deprenyl supresses the oxi-
these studies. In the final analysis, it will probably be dant stress associated with increased dopamine turnover.
Ann Neurol 26: 689690.
necessary to provide evidence of improvement in both
Cohen G, Pasik P, Cohen B et al. (1985). Pargyline and
imaging and clinical markers of disease progression
deprenyl prevent the neurotoxicity of 1-methyl-4-phenyl-
before it can be declared that an intervention is neuropro- 1,2,3,6-tetrahydropyridine (MPTP) in monkeys. Eur
tective and has a disease-modifying effect. J Pharmacol 106: 209210.
Collier T, Sortwell CE (1999). Therapeutic potential of nerve
References growth factors in Parkinsons disease. Drugs Aging 14:
261287.
Ansari KS, Yu PU, Kruck TPA et al. (1993). Rescue of axo- Cubo E, Gracies JM, Benabou R et al. (2001). Early morning
tomized immature rat facial motoneurons by L-deprenyl: off-medication dyskinesias, dystonia, and choreic sub-
stereospecificity and independence from monoamine oxi- types. Arch Neurol 58: 13791382.
dase inhibition. J Neurosci 13: 40424053. Deacon T, Schumacher J, Dinsmore J et al. (1997). Histologi-
Araki T, Kumagai T, Tanaka K et al. (2001). Neuroprotective cal evidence of fetal pig neural cell survival after trans-
effect of riluzole in MPTP-treated mice. Brain Res 918: plantation into a patient with Parkinsons disease. Nat
176181. Med 3: 350353.
Armstrong RJE, Svendsen CN (2000). Neural stem cells: Doble A (1999). The role of excitotoxicity in neurodegenera-
from the cell biology to cell replacement. Cell Transplant tive disease: implications for therapy. Pharmacol Ther 81:
9: 132145. 163221.
Barker RA (2002). Repairing the brain in Parkinsons dis- Elizan TS, Yahr MD, Moros DA et al. (1989). Selegiline use
ease: where next? Mov Disord 17: 233241. to prevent progression of Parkinsons disease. Experience
Barker RA (2006). Continuing trials of GDNF in Parkinsons in 22 de novo patients. Arch Neurol 46: 12751279.
disease. Lancet Neurol 5 (4): 285286. Elsworth JD, Glover V, Reynolds GP et al. (1978). Deprenyl
Beal MF, Matthews RT, Tieleman A et al. (1998). Coenzyme administration in man: a selective monoamine oxidase B
Q10 attenuates the 1-methyl-4-phenyl-1,2,3,6-tetrahydro- inhibitor without the cheese effect. Psychopharmacol-
pyridine (MPTP) induced loss of striatal dopamine and ogy 57: 3338.
dopaminergic axons in aged mice. Brain Res 783 (1): Fink JS, Schumacher JM, Ellias SL et al. (2000). Porcine
109114. xenografts in Parkinsons disease and Huntingtons dis-
Bensimon G, Lacomblez L, Meininger V (1994). A con- ease patients: preliminary results. Cell Transplant 9:
trolled trial of riluzole in amyotrophic lateral sclerosis. 273278.
ALS/Riluzole Study Group. N Engl J Med 330: 585591. Freed CR, Greene PE, Breeze RE et al. (2001). Embryonic
Betarbet R, Sherer TB, MacKenzie G et al. (2000). Chronic dopaminergic tissue transplants in advanced Parkinsons
systemic pesticide exposure reproduces features of disease. N Engl J Med 344: 710719.
Parkinsons disease. Nat Neurosci 3: 13011306. Freeman TB, Vawter DE, Leaverton PE et al. (1999). The use
Bjorklund LM, Sanchez-Pernaute R, Chung S et al. (2002). of a surgical placebo-control in evaluating cellular-based
Embryonic stem cells develop into functional dopaminer- therapy for Parkinsons disease. N Eng J Med 341: 988992.
gic neurons after transplantation in a Parkinson rat model. Gage FH (2000). Mammalian neural stem cells. Science 287:
Proc Natl Acad Sci USA 99: 23442349. 14331438.
Boulton AA (1999). Symptomatic and neuroprotective Gold BG, Nutt JG (2002). Neuroimmunophilin ligands in the
properties of the aliphatic propargylamines. Mech Ageing treatment of Parkinsons disease. Curr Opin Pharmacol 2:
Dev 111: 201209. 8286.
Brannan T, Yahr MD (1995). Comparative study of selegi- Greenamyre JT, Eller RV, Zhang Z et al. (1994). Antiparkin-
line plus L-dopa-carbidopa versus L-dopa-carbidopa alone sonian effects of remacemide hydrochloride, a glutamate
NEUROPROTECTION IN PARKINSONS DISEASE: CLINICAL TRIALS 27

antagonist, in rodent and primate models of Parkinsons Langston JW, Ballard PA, Tetrud JW et al. (1983). Chronic
disease. Ann Neurol 35: 635661. parkinsonism in humans due to a product of meperidine
Hagell P, Piccini P, Bjorklund A et al. (2002). Dyskinesias analog synthesis. Science 219: 979980.
following neural transplantation in Parkinsons disease. Lees AJ (1995). on behalf of the Parkinsons disease
Nat Neurosci 5: 627628. Research Group of the United Kingdom. Comparison of
Hauser RA, Koller WC, Amin J et al. (2000). Time course of therapeutic effects and mortality data of levodopa and
loss of clinical benefit following withdrawal of levodopa/ levodopa combimed with selegeline in patients with
carbidopa and bromocriptine in early Parkinsons disease. early, mild Parkinsons disease. BMJ 311 (7020):
Mov Disord 15: 485489. 16021607.
Hebert MA, Hoffer BJ, Zhang Z et al. (1999). Functional Le Witt PA, Segel SA, Mistura KL et al. (1993). Sympto-
effects of GDNF in normal and parkinsonian rats and matic anti-parkinsonian effects of monoamine oxidase-B
monkeys. In: M Tuszynski, JH Kordower (Eds.), CNS inhibition: comparison of selegiline and lazabemide. Clin
Regeneration: Basic Science and Clinical Advances. Aca- Neuropharmacol 16: 332337.
demic Press, San Diego, pp. 419436. Lin LF, Doherty DH, Lile JD et al. (1993). GDNF: a Glial
Hirsch EC, Hunot S, Faucheux B et al. (1999). Dopaminergic cell line-derived neurotrophic factor for midbrain dopami-
neurons degenerate by apoptosis in Parkinsons disease. nergic neurons. Science 260: 11301132.
Mov Disord 14: 383385. Lipton SA, Rosenberg PA (1994). Excitatory amino acids as
Huntingtons Disease Study Group (2000). A randomized, a final common pathway for neurologic disorders. N Engl
placebo-controlled trial of coenzyme Q10 and remacemide J Med 330: 613622.
in Huntingtons disease. Neurology 57: 397404. Marek K, Seibyl J, Shoulson I et al. (2002). Dopamine trans-
Ingold KU, Webb AC, Witter D et al. (1987). Vitamin E porter brain imaging to assess the effects of pramipexole
remains the major lipid-soluble, chain-breaking antioxi- vs levodopa on Parkinson disease progression. JAMA
dant in human plasma even in individuals suffering severe 287: 16531661.
vitamin E deficiency. Arch Biochem Biophys 259: Marsden CD, Olanow CW (1998). Neuroprotection in Par-
224225. kinsons disease: the causes of Parkinsons disease are
Jankovic J, Hunter C. (2002). A double-blind, placebo- being unravelled and rational neuroprotective therapy is
controlled and longitudinal study of riluzole in early close to reality. Ann Neurol 44 (3): 189196.
Parkinsons disease. Parkinsonism Relat Disord 8: Matthews RT, Ferrante RJ, Klivenyi P et al. (1999). Creatine
271276. and cyclocreatine attenuate MPTP neurotoxicity. Exp
Jenner P, Olanow CW (1998). Understanding cell death in Neurol 157 (1): 142149.
Parkinsons disease. Ann Neurol 44 (3): 7284. McNaught KS, Olanow CW, Halliwell B et al. (2001). Fail-
Kawasaki H, Suemori H, Mizuseki K et al. (2002). Genera- ure of the ubiquitin-proteasome system in Parkinsons dis-
tion of dopaminergic neurons and pigmented epithelia ease. Nat Rev/Neurosci 2 (8): 589594.
from primate ES cells by stromal cell-derived inducing Morrish P (2002). Is it time to abandon functional imaging in
activity. Proc Natl Acad Sci USA 99: 15801585. the study of neuroprotection? Mov Disord 17: 229232.
Kim JH, Auerbach JM, Rodriguez-Gomez JA et al. (2002). Mytilineou C, Cohen G (1985). Deprenyl protects dopamine
Dopamine neurons derived from embryonic stem cells neurons from the neurotoxic effect of 1-methyl-4-phenyl-
function in an animal model of Parkinsons disease. pyridinium ion. J Neurochem 45: 19511953.
Nature 418: 5056. Mytilineou C, Radcliffe P, Leonardi EK et al. (1997a). L-
Koller W, Vetere-Overfield B, Gray C et al. (1990). Environ- deprenyl protects mesencephalic dopamine neurons from
mental risk factors in Parkinsons disease. Neurology 40: glutamate receptor-mediated toxicity. J Neurochem 68:
12181221. 3339.
Kordower JH, Palfi S, Chen E et al. (1999). Clinicopathologi- Mytilineou C, Radcliffe PM, Olanow CW (1997b). L-(-)-
cal findings following intraventricular glial-derived neuro- Desmethylselegiline, a metabolite of L-(-)-selegiline, pro-
trophic factor treatment in a patient with Parkinsons tects mesencephalic dopamine neurons from excitotoxicity
Disease. Ann Neurol 46: 419424. in vitro. J Neurochem 68: 434436.
Kordower JH, Emborg ME, Bloch J et al. (2000). Neuro- Mytilineou C, Leonardi EK, Radcliffe P et al. (1998). Depre-
degeneration prevented by lentiviral vector delivery of nyl and desmethylselegiline protect mesencephalic
GDNF in primate models of Parkinsons disease. Science neurons from toxicity induced by glutathione depletion.
290: 767773. J Pharmacol Exp Ther 284: 700706.
Lacomblez L, Bensimon G, Leigh PN et al. (1996). Dose Obeso JA, Rodriguez-Oroz MC, Rodriguez M et al. (2000).
ranging study of riluzole in amyotrophic lateral sclerosis. Pathophysiology of the basal ganglia in Parkinsons
Lancet 347: 14251431. disease. TINS 81: S8S19.
Lang AE, Gill S, Patel NK et al. (2006). Randomized con- Obinu MC, Reibaud M, Blanchard V et al. (2002). Neuropro-
trolled trial of intraputamenal glial cell line-derived neuro- tective effect of riluzole in a primate model of Parkinsons
trophic factor infusion in Parkinson disease. Ann Neurol disease: behavioral and histological evidence. Mov Disord
59 (3): 459466. 17: 1319.
28 F. STOCCHI

Olanow CW (1992). A rationale for dopamine agonists as Parkinson Study Group (2000b). Pramipexole vs levodopa as
primary therapy for Parkinsons disease. Can J Neurol initial treatment for Parkinson disease: a randomized con-
Sci 19: 108112. trolled trial. JAMA 284: 231238.
Olanow CW, Calne D (1991). Does selegiline monotherapy Pezzoli G, Strada O, Silani V et al. (1996). Clinical and
in Parkinsons disease act by symptomatic or protective pathological features in hydrocarbon-induced parkinson-
mechanisms? Neurology 42: 1326. ism. Ann Neurol 40: 922925.
Olanow CW, Hauser RA, Gauger L et al. (1995). The effect Przuntek H, Conrad B, Dichgans J et al. (1999). SELEDO: a
of deprenyl and levodopa on the progression of signs 5-year long-term trial on the effect of selegiline in early
and symptoms in Parkinsons disease. Ann Neurol 38: Parkinsonian patients treated with levodopa. Eur J Neurol
771777. 6: 141150.
Olanow CW, Fahn S, Langston JW et al. (1996a). Selegiline Rascol O, Brooks DJ, Korczyn AD et al. (2000). A five-year
and mortality: a point of view. Ann Neurol 40: 841845. study of the incidence of dyskinesia in patients with early
Olanow CW, Freeman TB, Kordower JH (1996b). Fetal Parkinsons disease who were treated with ropinirole or levo-
nigral transplantation as a therapy for Parkinsons disease. dopa. 056 Study Group. N Engl J Med 342: 14841491.
TINS 19: 102109. Reed JC (2002). Apoptosis based therapies. Nat Rev Drug
Olanow CW, Jenner P, Brooks D (1998a). Dopamine ago- Discov 1 (2): 111121.
nists and neuroprotection in Parkinsons Disease. Ann Rodriguez MC, Obeso JA, Olanow CW (1998). Subthalamic
Neurol 44 (3): 167174. nucleus-mediated excitotoxicity in Parkinsons Disease: a
Olanow CW, Myllyla VV, Sotaniemi KA et al. (1998b). target for neuroprotection. Ann Neurol 44 (3): 175188.
Effect of selegiline on mortality in patients with Roy E, Bedard PJ (1993). L-deprenyl increases survival of
Parkinsons disease: a meta-analysis. Neurology 51: rat foetal nigral neurones in culture. Neuroreport 4:
825830. 11831186.
Olanow CW, Mytilineou C, Tatton WH (1998c). Status of Schumacher JM, Ellias SA, Palmer EP et al. (2000). Trans-
selegiline as a neuroprotective agent in Parkinsons Dis- plantation of embryonic porcine mesencephalic tissue in
ease. Mov Disord 13 (S): 5558. patients with Parkinsons disease. Neurology 54:
Olanow CW, Schapira AH, Le Witt PA et al. (2006). 10421050.
TCH346 as a neuroprotective drug in Parkinsons disease: Shoulson I, Oakes D, Fahn S et al. (2002), and the Parkinson
a double-blind, randomised, controlled trial. Lancet Neurol Study Group (2002). The impact of sustained deprenyl
5 (12): 10131020. (selegiline) in levodopa-treated Parkinsons disease: a ran-
Parkinson Disease Research Group in the United Kingdom domized placebo-controlled extension. Ann Neurol 51:
(1993). Comparisons of therapeutic effects of levodopa, 604612.
levodopa and selegiline, and bromocriptine in patients Steiner JP, Hamilton GS, Ross DT et al. (1997). Neurotrophic
with early, mild Parkinsons disease: three year interim immunophilin ligands stimulate structural and functional
report. BMJ 307: 469472. recovery in neurodegenerative animal models. Proc Natl
Parkinson Study Group (1989a). DATATOP: a multicenter Acad Sci USA 94: 20192024.
controlled clinical trial in early Parkinsons disease. Arch Stocchi F (2000). Dopamine agonists in Parkinsons disease:
Neurol 46: 10521060. what is their role in early treatment? In: KJ Palmer (Ed.),
Parkinson Study Group (1989b). Effect of deprenyl on the Drug Treatment Issues in Parkinsons Disease. Adis Inter-
progression of disability in early Parkinsons disease. national, Hong Kong, pp. 2740.
N Engl J Med 321 (20): 13641371. Tanner CM, Ottman R, Goldman SM et al. (1999). Parkinson
Parkinson Study Group (1993). Effects of tocopherol and disease in twins: an etiologic study. JAMA 281: 341346.
deprenyl on the progression of disability in early Parkin- Tatton WG, Greenwood CE (1991). Rescue of dying neu-
sons disease. N Engl J Med 328 (3): 176183. rons: a new action for deprenyl in MPTP parkinsonism.
Parkinson Study Group (1996a). The impact of extended J Neurosci Res 30: 666677.
deprenyl and tocopherol treatment in Parkinsons disease Tatton WG, Ju WYL, Holland DP et al. (1994). ()-Deprenyl
patients requiring levodopa. Ann Neurol 39: 2936. reduces PC12 cell apoptosis by inducing new protein
Parkinson Study Group (1996b). The impact of extended synthesis. J Neurochem 63: 15721575.
deprenyl and tocopherol treatment in Parkinsons disease Tatton WG, Chalmers-Redman RME, Ju WJH et al. (2002).
patients not requiring levodopa. Ann Neurol 39: 3745. Propargylamines induce antiapoptotic new protein synth-
Parkinson Study Group (1996c). Effect of lazabemide on the esis in serum- and nerve growth factor (NGF)-withdrawn,
progression of disability in early Parkinsons disease. Ann NGF-differentiated PC-12 cells. J Pharmacol Exp Ther
Neurol 40: 99107. 301: 753764.
Parkinson Study Group (1998). Mortality in DATATOP: a Tetrud JW, Langston JW (1989). The effect of deprenyl
multicenter trial in early Parkinsons disease. Ann Neurol (selegiline) on the natural history of Parkinsons disease.
43: 318325. Science 245: 519522.
Parkinson Study Group (2000a). A multicenter randomized Turski L, Bressler K, Rottig KJ et al. (1991). Protection of
controlled trial of remacemide hydrochloride as monother- substantia nigra from MPP neurotoxicity by NMDA
apy for PD. Neurology 54: 15831588. antagonists. Nature 349: 414418.
NEUROPROTECTION IN PARKINSONS DISEASE: CLINICAL TRIALS 29

Uitti RJ, Rajput AH, Ahlskog JE et al. (1996). Amantadine Wu RM, Chieuh CC, Pert A et al. (1993). Apparent antioxi-
treatment is an independent predictor of improved survival dant effect of I-deprenyl on hydroxyl radical formation
in Parkinsons disease. Neurology 46: 15511556. and nigral injury elicited by MPP in vivo. Eur J Pharma-
Waldmeir PC, Boulton AA, Cools AR et al. (2000). Neuro- col 243: 241247.
rescuing effects of the GAPDH ligand CGP 3466B. Youdim MB, Weinstock M (2001). Molecular basis of neu-
J Neural Transm 60: 197214. roprotective activities of rasagiline and the anti-Alzheimer
Watts R. (2002). Neurology abst. drug TV3326 [(N-propargyl-(3R)aminoindan-5-YL)-ethyl
Whone AL, Remy P, Davis MR et al. (2002). Neurology methyl carbamate]. Cell Mol Neurobiol 21: 555573.
abst.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 32

Levodopa

THOMAS D. HALBIG1* AND WILLIAM C. KOLLER2

1
Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA
2
Department of Neurology, University of North Carolina, NC, USA

32.1. History Hornykiewicz, 1960). Based on these findings, only 1 year


later, Birkmayer and Hornykiewicz administered levo-
The English physician James Parkinson from London dopa in an open trial intravenously in 20 patients and
first described Parkinsons disease (PD) in 1817. Since reported marked improvements of all motor symptoms
then several different drugs have been employed to treat (Birkmayer and Hornykiewicz, 1961). However, several
PD symptomatically. The first therapeutic agents that other trials provided conflicting results on the efficacy of
were proven to be clinically effective were anticholiner- levodopa and a double-blind study published in 1966
gics. Charcot and Ordendstein in the early 1860s at the reported a negative result (Fehling, 1966). The clinical
Salpetriere in Paris treated patients with belladonna breakthrough in levodopa treatment of PD is grounded
compounds. However, therapeutic efficacy of treatments on the seminal work of the American Cotzias (Cotzias
with potato plant extracts such as belladonna and atro- et al., 1967). In a placebo-controlled trial, he escalated
pine was disappointing. This changed with the introduc- doses of externally administered levodopa up to 16 g and
tion of synthetic anticholinergics in the 1940s, which observed marked effects on all cardinal symptoms of PD.
had fewer side-effects and provided better symptomatic Based on several other confirmatory controlled trials, the
control, in particular of tremor and rigidity. US Food and Drug Administration (FDA) finally
It was only in the mid-1960s that replacement therapy approved levodopa for use in PD in 1970. Since then
with levodopa was introduced and marked the turning it has become the gold standard in the symptomatic
point, eventually leading to a revolution in symptomatic treatment of PD.
therapy of PD (for review, see Tolosa et al., 1998;
Hornykiewicz, 2002). Levodopa treatment became the 32.2. Dopamine pharmacology
so-far only true success story in the symptomatic treatment 32.2.1. Synthesis and storage
of a neurodegenerative disorder and PD in particular. The
key for the establishment of dopaminergic replacement Dopamine (3,4-dihydroxyphenylethylamine) is a mono-
therapy was the hypothesis that dopamine deficiency is amine belonging to the family of catecholamines. In
the pathophysiological basis of PD. The work of Arvid mammalians, it is synthesized in nerve terminals via
Carlson in Lund in Sweden paved the ground for the estab- intermediate products in several steps (Webster, 2001).
lishment of this assumption. In the late 1950s Carlson L-Tyrosine is considered the initial substrate for the
observed that reserpine-treated rabbits present a clinical synthesis of levodopa (Fig. 32.1). It is synthesized from
picture with parkinsonian features and that these symp- phenylalanine. L-Tyrosine is hydroxylated by tyrosine
toms could be reversed by injection of the norepinephrine hydroxylase (TH) at position 3 of the phenyl ring to
precursor DL-dopa. Carlson furthermore provided experi- L-3,4-dihydroxyphenylalanine (levodopa). Tyrosine is
mental evidence for the presence of dopamine in the stria- present in all catecholamine-containing neurons and
tum. In 1960, Ehringer and Hornykiewicz in Vienna requires tetrahydrobiopterin and oxygen as cofactors.
finally demonstrated neuropathological evidence for Tyrosine hydroxylase is a rate-limiting enzyme in
striatal dopamine deficits of PD patients (Ehringer and the synthesis of dopamine and higher concentrations

*Correspondence to: Thomas D. Halbig, MD, Centre dInvestigation Clinique et Federation des Maladies du Systeme Nerveux,
Centre Hospitalier Universitaire Pitie-Salpetriere, 47-83 Boulevard de lHopital, 75651 Paris cedex 13, France. E-mail: thomas.
halbig@psl.aphp.fr & halbig.psl@gmail.com, Tel: +33-1-42-16-19-50, Fax: +33-1-42-16-19-58.
32 T. D. HALBIG AND W. C. KOLLER
NH2 specific vesicular transport system and stored in
presynaptic vesicles.
CH2 CH COOH
Calcium-dependent mechanisms mediate the release
of dopamine into the synaptic cleft (von Bohlen und
Halbach and Dermietzel, 2002). More specifically,
Phenylalanine
action potentials induce calcium influx via voltage-
dependent channels, eliciting fusion of the storage vesi-
Phenylalanine hydroxylase cles with the presynaptic membrane and release of
dopamine into the synaptic cleft. After diffusing in the
NH2
synaptic cleft, the dopamine molecule binds at postsy-
naptic receptors, triggering a complex cascade of post-
CH2 CH COOH synaptic intracellular events and eventually leading to
either activation or inhibition of the postsynaptic neu-
HO ron. Nigrostriatal dopaminergic neurons implicated
Tyrosine in motor functions are characterized by continuous
firing (DeLong et al., 1983) with intermittent-burst firing
Tyrosine hydroxylase (Grace, 1991; Schultz, 1986). However, striatal intra-
synaptic dopamine levels are relatively constant and
NH2 ensure continuous dopaminergic receptor stimulation
HO CH2 CH COOH (DeLong et al., 1983; Moore et al., 1998).
The duration of action of dopamine at the postsynap-
tic receptors is a function of several factors, including
HO the amount of dopamine in the synaptic cleft, activation
DOPA
of presynaptic autoreceptors and active removal from
the synaptic cleft. The latter is in part determined
DOPA decarboxylase
by presynaptic reuptake of dopamine by dopamine
transporters (DAT). DAT is a glycoprotein of 619
amino acids (70 kDa). This rapid presynaptic reuptake
HO CH2CH2NH2
system recaptures about 80% of the released dopamine
(Giros and Caron, 1993). Intraneurally, the dopamine
HO that is reuptaken is transported through a specific vesi-
Dopamine
cular transport system and again stored in vesicles until
Fig. 32.1. Synthesis of dopamine from its precursors. Repro- upcoming release into the synaptic cleft. Another
duced from von Bohlen und Halbach and Dermietzel (2002)
important factor determining the amount of dopamine
with permission from Wiley-VCH Verlag Gmbh & Co. KG
in the synaptic cleft is the activation of dopaminergic
and the authors.
autoreceptors located at the presynaptic membrane.
These autoreceptors monitor the intrasynaptical dopa-
of tyrosine do not increase the amount of hydroxylated mine concentration and their activation inhibits the
levodopa. Levodopa is rapidly decarboxylated to 3,4- synthesis and release of dopamine, whereas their block-
dihydroxyphenethylamine (dopamine) by the cytosolic age enhances dopaminergic synthesis and release (von
aromatic amino acid decarboxylase (AADC) which splits Bohlen und Halbach and Dermietzel, 2002).
the carbon acid group of the amino acid. AADC is not
rate-limiting and higher concentrations of levodopa 32.2.2. Metabolization
increase the production of dopamine.
Although the highest concentrations of AADC are Dopamine is metabolized via two pathways to homova-
found in striatal nigrostriatal dopaminergic nerve nillic acid (HVA) (Webster, 2001). Both pathways
terminals (Lloyd et al., 1975; Melamed et al., involve the action of monoamine oxidase (MAO) and
1980; Trugman et al., 1991), there are other, alterna- catechol-O-methyltransferase (COMT). MAO is located
tive sites of levodopa decarboxylation. For example, in, or attached to, the intraneural mitochondria. There
there is also striatal decarboxylase activity in the are two MAO subtypes, A and B. MAO-A is found
terminals of serotonergic and noradrenergic neurons primarily in the gastrointestinal tract and has stronger affi-
and in capillary endothelial or glial cells (Trugman nity to serotonin (5-HT) and norepinephrine. MAO-B, in
et al., 1991). After dopamine is synthesized in dopa- contrast, has a higher affinity for dopamine. In the brain
minergic nerve terminals, it is transported through a 80% of MAO is formed by MAO-B (Squires, 1972).
LEVODOPA 33
COMT is a cytoplasmic enzyme present throughout levodopa is actively resorbed at the level of the small
the body (Guldberg and Marsden, 1975; Schultz et al., bowel. Resorption is mediated by an active transport
1989; Schultz and Nissinen, 1989; Mannisto et al., system, the energy-dependent large neutral amino acid
1992). Intracerebrally, there is no significant COMT (LNAA) transport system (Nutt, 1992). Several factors
activity in presynaptic dopaminergic terminals, but limit the amount of resorbed levodopa. Firstly, the
some activity is present postsynaptically and substan- speed of gastric emptying can be decreased by auto-
tial activity is located in glial cells (Guldberg and nomic dysfunctions which have been found to be fre-
Marsden, 1975; Mannisto and Kaakkola, 1999). quent in PD (Edwards et al., 1992; Hardoff et al.,
On the first pathway, MAO intraneurally oxidatively 2001; Pfeiffer, 2003). Transfer of orally administered
deaminates dopamine to 3,4-dihydroxyphenylacetalde- levodopa can also be delayed by concurrent food
hyde, which in turn is dehydrogenated by aldehyde intake (Djaldetti et al., 1996), with dietary LNAAs
dehydrogenase to dihydroxyphenylacetic acid (DOPAC) such as lysine or phenylalanine in protein-rich food
(Fig. 32.2). DOPAC is methylated by COMT to HVA. competing with levodopa for transport across the gut
On the second pathway, COMT O-methylates dopa- wall via the LNAA transport system.
mine to 3-methoxytyramine (3-MT). 3-MT in turn is Once absorbed, levodopa is metabolized peri-
deaminated by MAO to 3-methoxy-4-hydroxyphenyla- pherally by AADC and COMT to dopamine and
cetaldehyde, which is dehydrogenated by aldehyde 3-O-methyldopa (3-OMD), reducing the bioavailabil-
dehydrogenase to HVA (Webster, 2001). ity of externally administered levodopa. Peripheral
levodopa half-life is thought to be 6090 minutes
32.3. Pharmacology of orally administered (Sasahara et al., 1980; Nutt and Fellman, 1984; Hardie
levodopa et al., 1986; Fabbrini et al., 1988).
Levodopa crosses the bloodbrain barrier, again via
Dopamine does not penetrate the bloodbrain barrier an LNAA transport system. Since simultaneous intra-
and therefore cannot be used for symptomatic dopami- venous administration of levodopa and intake of pro-
nergic replacement therapy. However, levodopa does tein-rich meals lead to diminished clinical efficacy, it
cross the bloodbrain barrier. Orally administered has to be assumed that levodopa transport via the

HO CH2CH2NH2

HO
Dopamine COMT

MAO

HO CH2CHO HO CH2CH2NH2

HO CH3O
3,4-Dihydroxyphenylacetaldehyde 3-Methoxytyramine (3-MT)

Aldehyde-
MAO
dehydrogenase

HO CH2COOH HO CH2CHO

HO CH3O

3,4-Dihydroxyphenylacetic acid 3-Methoxy-4-hydroxyphenylacetaldehyde


COMT
(DOPAC) Aldehyde-
dehydrogenase
CH3O CH2COOH

HO
HVA
Fig. 32.2. Degradation of dopamine. Reproduced from von Bohlen und Halbach and Dermietzel (2002) with permission from
Wiley-VCH Verlag GmbH & Co. KG and the authors.
34 T. D. HALBIG AND W. C. KOLLER
bloodbrain barrier again competes with dietary decarboxylation of levodopa to dopamine, patients
LNAAs (Nutt et al., 1985). Due to peripheral absorp- experienced intolerable side-effects, such as nausea
tion concurrence, peripheral degradation and competi- or hypotension, resulting from stimulation of periph-
tion with LNAA at the bloodbrain barrier, only 1% eral dopamine receptors (see section 32.6). Thus,
of orally administered levodopa reaches the central major problems related to levodopa administration in
nervous system (CNS) (Mannisto et al., 1990). initial trials were insufficient efficacy related to
After transfer via the bloodbrain barrier, levodopa marked side-effects. In consequence, levodopa-only
is decarboxylated by AADC to dopamine. The exact therapy was not a practical therapeutical option to be
site of decarboxylation of exogenous levodopa to used in most patients.
dopamine in the brain is unknown (Lloyd et al., This changed dramatically when evidence for the
1975; Melamed et al., 1980; Trugman et al., 1991). pharmacological and clinical effects of peripheral inhi-
Although probably most striatal AADC is located in bition of decarboxylase was obtained. For example,
nigrostriatal dopaminergic nerve terminals, decarboxy- Bartholini et al. (1967) demonstrated that the inhibi-
lase activity in the terminals of serotonergic and nora- tion of peripheral decarboxylase enhanced the concen-
drenergic neurons and in capillary endothelial cells tration of cerebral catecholamines. More importantly,
(Trugman et al., 1991) is likely involved in the decar- different peripherally acting decarboxylase inhibitors
boxylation of exogenous levodopa. This is important, when coadministered with levodopa were shown to
since nigrostriatal dopaminergic AADC activity dimi- allow administration of levodopa doses which were
nishes in the course of the progressive dopaminergic about four times lower than the levodopa-only admin-
nigrostriatal cell degeneration. istration. Compatible with this, combined administra-
Based on research using animal models, it has tion of levodopa and AADC led to diminished
to be assumed that the exogenous dopamine com- dopaminergic side-effects and more efficient and fas-
plements remaining endogenous dopamine and is ter symptom control (Birkmayer, 1969; Cotzias et al.,
taken up by nigrostriatal nerve terminals, increasing 1969; Calne et al., 1971; Yahr et al., 1971; Sweet
striatal dopamine levels (Hornykiewicz, 1974; Horne et al., 1972; Howse and Matthews, 1973; Papavasiliou
et al., 1984). Both the endogenous and exogenous et al., 1973; Cedarbaum et al., 1986). In 1975, com-
dopamine are presumably stored in storage vesicles binations of the AADCs a-methyldopa hydrazine
and subsequently released into the synaptic cleft in (carbidopa) and RO44602 (benserazide) plus levo-
order to mediate its effects via stimulation of post- dopa were commercialized as Sinemet and Madopar
synaptic dopaminergic receptors as well as presy- and since then have been used for symptomatic
naptic autoreceptors. Further metabolization underlies dopaminergic replacement therapy (Fig. 32.3).
the same mechanisms as described for endogenous The pharmacokinetics of carbidopa and beneserazide
dopamine. differ. Benserazide in conjunction with levodopa leads
to earlier and higher peak dopa levels which decline ear-
32.4. Dopa-decarboxylase inhibition lier compared to carbidopa. However, pharmacological
differences between both AADCs in general are not
Although initial attempts to treat PD using oral levo- reflected clinically (Greenacre et al., 1976; Rinne and
dopa permitted the reduction of symptoms to some Molsa, 1979; Lieberman et al., 1984; Troconiz et al.,
extent, the overall outcome was frustrating. In their 1998). To ensure therapeutic levodopa efficacy without
seminal paper published in the New England Journal peripheral side-effects, complete peripheral inhibition
of Medicine, Cotzias and colleagues (1967) reported of dopa-decarboxylase is desirable. Although it is gener-
symptom reduction by oral administration of levodopa ally agreed that a daily dose of at least 75 mg is needed
but also marked side-effects. Since only 1% of ad- to achieve clinically satisfactory inhibition of decar-
ministered levodopa eventually reached the CNS, initi- boxylase activity, higher doses of up to 290 mg/day
ally used doses had to be very high. Due to peripheral of carbidopa have been shown to increase levodopa

O OH NH2
COOH
HO
HO HO NH OH
OH NH NH
H3C
NH2 HO NH2 O
HO HO
Levodopa Carbidopa Benserazide
Fig. 32.3. Chemical structures of levodopa, carbidopa and benserazide.
LEVODOPA 35
bioavailability and decrease time to peak plasma con- randomized double-blind placebo-controlled trial test-
centration (Cedarbaum et al., 1986). It is conceivable ing the efficacy of levodopa was initiated in 1999
that further increase in carbidopa doses may lead to (Parkinson Study Group, 2004a). The Early Versus
penetration of carbidopa through the bloodbrain bar- Late L-Dopa (ELLDOPA) study tested the effects of
rier and consecutive inhibition of levodopa conversion levodopa on disease progression in de novo PD
to dopamine. Yet, there are no data at that point to patients with a mean age of 64 years and a disease
support this conclusion. duration of 2 years. Baseline modified HoehnYahr
The administration of AADCs doubles the bioavail- scores (Hoehn and Yahr, 1967; Fahn et al., 1987) were
ability of orally administered levodopa and halves its less than 3, total Unified Parkinsons Disease Rating
plasma clearance (Nutt et al., 1985). Administration of Scale (UPDRS) score (Fahn et al., 1987) was about
standard oral levodopa preparations, including AADC, 27 and UPDRS III (motor score) was about 19. Sub-
in general leads to Cmax after 1545 minutes (Nutt and jects were randomly assigned to receive placebo or
Fellman, 1984). Plasma half-life is mainly determined levodopa/carbidopa at a dose of 50/12.5 mg three
by distribution in tissues and hepatic first-pass effects times daily, 100/25 mg three times daily or 200/50 mg
and varies between 1 and 1.5 hours (Nutt and Fellman, three times daily, respectively. Doses were increased
1984; Hardie et al., 1986; Fabbrini et al., 1988). to the full amount over a period of 9 weeks in a blinded
fashion. For clinical evaluation, the UPDRS was used
32.5. Clinical effects of levodopa on at the screening, baseline and interim visits (at the
Parkinsons disease symptoms end of weeks 3, 9, 24 and 40) and during each of the 2
weeks of a subsequent washout phase. During the
After initial trials (Birkmayer and Hornykiewicz, four interim visits, the evaluations were performed
1961; Barbeau et al., 1962) with encouraging yet before the administration of the first dose of the study
controversial results, the first study reporting rather drug. Of a total of 361 subjects enrolled in the study, 317
consistent beneficial effects of levodopa on PD motor (88%) took the study medication for 40 weeks. The
symptoms was published in 1967 (Cotzias et al., patients in the placebo group had mild improvement
1967). In this ground-breaking report of an open-label at the week 3 visit, but after that their symptoms
study, Cotzias and colleagues were able to show the worsened steadily throughout the study period, including
efficacy of levodopa in 16 PD patients with a mean the 2-week washout phase (Fig. 32.4). In contrast, in
disease duration of 7.8 years (range 132 years). patients treated with levodopa, a strong doseresponse
Patients received levodopa (DL-dopa) at a dose of benefit that persisted through week 40 was detected.
316 g (without AADC) or placebo. The clinical
assessment included handwriting, number of steps
required to walk 10 meters, grasping objects, drawing, 12
sit-to-stand maneuver, rigidity, tremor, festination, 10
Change in Total Score (units)

dysarthria and muscle strength. Eight patients showed 8


Placebo
either complete or marked sustained improvements of 6
symptoms, including tremor, rigidity, festination and 4
hypomimia, and 2 patients showed mild improvements. 2 150 mg
Cotzias et al. already noted that rigidity decreased 0
300 mg
or disappeared at low doses whereas improvements in 2
tremor were observed on higher doses. However, it took 4 600 mg
5 more years until again Cotzias demonstrated that 6
coadministration of a peripheral dopa-decarboxylase 8
inhibitor reduced the side-effects of levodopa therapy 2 6 10 14 18 22 26 30 34 38 42 46
significantly by allowing the use of markedly lower Baseline Week Withdrawal
of study drug
doses of levodopa (Papavasiliou et al., 1972). Since
then, levodopa therapy (in the following, levodopa Fig. 32.4. Early Versus Late L-Dopa (ELLDOPA) study
Unified Parkinsons Disease Rating Scale (UPDRS) changes.
denominates levodopa AADC) was extensively used
Changes in total scores on the UPDRS from baseline through
and soon became established as the gold standard of evaluation at week 42. The points on the curves indicate mean
symptomatic antiparkinsonian therapy. changes from baseline in the total scores at each visit. Nega-
Interestingly, even though worldwide clinical use tive scores on the curves indicate improvement from baseline.
supported the view that levodopa is a highly effective The bars indicate the standard error. From Parkinson Study
agent in the treatment of motor symptoms in PD, Group (2004a), reproduced with permission. # 2004 Massa-
it took about 30 years until the first prospective chusetts Medical Society. All rights reserved.
36 T. D. HALBIG AND W. C. KOLLER
Improvements in the levodopa-treated patients reached a during treatment initiation (Rinne et al., 1998b;
peak at week 24 in patients receiving the highest dose of Parkinson Study Group, 2000, 2004a). Nausea devel-
600 mg. At week 24, improvements were found to be oped in a dose-dependent way and in the ELLDOPA
dose-dependent with a decrease of 6 points in the 600 study occurred on daily doses as low as 150 mg in
mg group, 4 points (300 mg), no change (150 mg) and 16%, on 300 mg in 26% and on 600 mg in 31% of
worsening by 4 points in the placebo group. The scores patients (Parkinson Study Group, 2004a). Nausea and
on the UPDRS in the three levodopa groups worsened vomiting are considered peripheral side-effects of levo-
during the 2-week washout period, but these groups did dopa treatment. They are mediated via dopaminergic sti-
not deteriorate to the level observed in the placebo group mulation of the area postrema which is located at the
and the group receiving the highest dose of levodopa had bottom of the fourth ventricle outside the bloodbrain
the best result. barrier and involved in the regulation of nausea and
Compatible with these results are findings from stu- emesis.
dies assessing the effects of other antiparkinsonian In most patients, nausea resolves within days or
agents such as dopamine agonists (Rinne et al., 1998b; weeks of continued levodopa treatment without any
Parkinson Study Group, 2000, 2004b; Rascol et al., additional action being taken. When nausea persists,
2000) or levodopa slow-release preparations in de novo temporary dose reduction and slower levodopa reti-
patients (Dupont et al., 1996; Block et al., 1997; Koller tration may be tried. Furthermore, several agents
et al., 1999) against levodopa as comparator. These are available to prevent or reduce gastrointestinal
trials assessed de novo patients with a disease duration side-effects. For example, peripherally acting dopa-
of approximately 2 years and patients were followed mine receptor blockers may be employed in order
for up to 5 years. In the agonist vs levodopa trials, to avoid dopaminergic receptor stimulation of the
patients of both treatment conditions usually could area postrema. Domperidone is a peripherally acting
receive additional levodopa when study medication dopamine-2 (D2) receptor antagonist that does not
did not allow sufficient symptom control. Results penetrate the bloodbrain barrier and has a half-life
obtained in levodopa-only-treated patients consistently of more than 10 hours. Its time to maximum peak
showed improvements of motor functions of up to level is about 3060 minutes (Huang et al., 1986).
30% as assessed by the UPDRS at the end of the study Domperidone has no significant effect on dopamine
periods (Table 32.1). Compared to dopamine agonist- pharmacokinetics (Bradbrook et al., 1986). It is
treated patients, studies show significantly greater administered in a dose of 1020 mg per dose of
improvement in patients treated with levodopa only and levodopa. Domperidone has been shown to be as
support the notion that levodopa is the most effective efficient as carbidopa (see below) in preventing nau-
agent in the symptomatic treatment of PD. sea and vomiting as side-effects of levodopa
(Langdon et al., 1986; Barone, 1999). Domperidone
32.6. Tolerability and acute non-motor is approved in Europe (Motilium) and Canada but is
side-effects unavailable in the USA.
Another option is the add-on of trimethobenzamide
Apart from the nigrostriatal dopaminergic system hydrochloride. Although its mechanism of action is
which is the target of dopaminergic replacement not entirely understood, it may also involve the
therapy in PD, there are several other different central chemoreceptor trigger zone of the area postrema.
and peripheral dopaminergic systems (Webster, 2001). There are no controlled studies on the effects of
External administration of dopaminergic agents may trimethobenzamide hydrochloride in PD; however,
therefore exert peripheral, central and combined per- the administration of 200 mg three times daily has
ipheral and central effects and side-effects. As for the been recommended for the treatment or prevention
motor effects, controlled data on side-effects are of dopaminergically induced nausea and vomiting
mainly obtained from the ELLDOPA study and trials (Olanow et al., 2001). Trimethobenzamide is mar-
comparing the effects of different dopaminergic agents keted in the USA as Tigan and not available in most
with levodopa as comparator. The most frequent non- European countries.
motor side-effects include nausea, sedation, hypoten- Furthermore, additional carbidopa may be added
sion and neuropsychiatric symptoms (Table 32.2). to the levodopa regimen (Markham et al., 1974).
Carbidopa alone is available as Lodosyn in the USA.
32.6.1. Nausea Other agents which may be employed are the 5-HT
receptor antagonist (5-HT3) ondansetron (Wilde and
The most frequent side-effect in levodopa-treated Markham, 1996) or the antiemetic diphenidol (Duvoisin,
patients is nausea, occurring in up to 30% of patients 1972).
Table 32.1
Symptomatic motor effects of long-term levodopa (LD) treatment (25 years) reported in phase III studies assessing the effects of dopamine agonists (1, 3, 4, 5) or
controlled release LD (2) against LD as comparator in drug-naive Parkinsons disease (PD) patients

Change in Change in
n at Age H&Y PD duration UPDRS lll UPDRS ll Study duration Levodopa UPDRS lll UPDRS ll
Reference Agents baseline (years) stage months baseline baseline months mg/day at end at end

1 LD 204 62.6 13 24 29.1 NR 48 500 9 2.7*


(cabergoline) (208) (60.5) (22.8) (27.5) (6) (2.4*)
2 LD 306 Range 13 Overall 27.6 15.1 8.7 60 426 0.4 0.1

LEVODOPA
(controlled (312) 3075 (15.4) (9.4) (1.2) (1.0)
release LD)
3 LD 150 60.9 13 21.6 22 8.3 23.5 509 7.3 2.2
(pramipexole) (151) (61.5) (18) (22.3) (9.1) (3.4) (1.1)
4 LD 150 60.9 13 21.6 22 8.3 48 702 3.4 0.5
(pramipexole) (151) (61.5) (18) (22.3) (9.1) (1.3) (1.7)
5 LD 89 63.0 13 30 21.7 8 60 753 4.8 0
(ropinirole) (179) (63.0) (29) (21.5) (8) (0.8) (1.6)

1, Rinne et al. (1998b); 2, Koller et al. (1999); 3, Parkinson Study Group (2000); 4, Parkinson Study Group (2004b); 5, Rascol et al. (2000).
H&Y, Hoehn and Yahr; UPDRS, Unified Parkinsons Disease Rating Scale; *, % value; NR, not reported.

37
38
Table 32.2
Side-effects of levodopa within up to 5 years of treatment in controlled studies assessing the effects of LD against dopamine agonists (1, 3, 4), controlled release LD (2)
and placebo (5) as comparator

Withdrawal
Study Hallucinations Orthostatic rate for

T. D. HALBIG AND W. C. KOLLER


Refer- duration Mean LD Nausea/ Sleep (14)/abnormal hypoten- adverse
ences Agents n (months) dose (mg) vomiting* Somnolence* disturbances* dreaming (5)* sion* events*

1 LD (cabergoline) 204 48 500 32 NR 28 NR 24 13


(208) (37) (27) (32) (16)
4 LD 306 60 426 28 6 2 5 2 9
(LD controlled (312) (26) (6) (4) (4) (3) (7)
release)
2 LD (pramipexole) 150 23.5 509 36.7 17.3 22.0 3.3 10 8.0
(151) (36.4) (32.4) (25.8) (9.3) (6.0) (10.6)
3 LD 89 60 753 49.4 19.1 23.6 5.6 12.4 33
(ropinirole) (179) (48.6) (27.4) (25.1) (17.3) (11.7) (27)
5 LD 92 10 150 16.3 0 8.7 3.3 2
(placebo) (90) (13.3) (2.2) (10.0) (0) (3)
5 LD 88 10 300 26.1 5.7 4.5 0 2
(placebo)
5 LD 91 10 600 31.9 5.5 5.5 5.5 1
(placebo)

1, Rinne et al. (1998b); 2, Koller et al. (1999); 3, Parkinson Study Group (2000); 4, Rascol et al. (2000); 5, Parkinson Study Group (2004a).
* indicates % of patients; NR, not reported.
LEVODOPA 39
32.6.2. Hypotension levodopa to dopamine by AADC does not entirely sup-
press the development of levodopa-induced hypoten-
Since the introduction of levodopa as symptomatic PD sion. It thus is likely that central mechanisms are also
therapy, orthostatic hypotension has been reported as a involved (see Ch. 14).
side-effect (Calne, 1970; Calne et al., 1970). However, Symptomatic orthostatic hypotension may be trea-
the data on the specific acute and long-term effects of ted by non-pharmacologic measures such as increased
levodopa on hypotension are controversial and vary salt and fluid intake, compressive stockings and sleep-
depending on methodological differences. Orthostatic ing with head-up tilt of bed. Pharmacologic treatment
hypotension occurs in 030% of PD patients without includes fludrocortisone acetate 0.050.2 mg/day (Hoehn,
major comorbidity during the first 35 years of treat- 1975; Hakamaki et al., 1998), indometacin 50 mg three
ment in the early stages of the disease (Rinne et al., times daily (Abate et al., 1979), or midodrine hydro-
1998b; Parkinson Study Group, 2000, 2004a). Interest- chloride, an alpha-adrenergic agonist at a dose of up
ingly, the ELLDOPA trial did not report any incidence to 10 mg three times a day (Jankovic et al., 1993;
of orthostatic hypotension. Compatible with this Low et al., 1997). Finally, domperidone may be tried.
result, several other studies did not find acute effects Yet, although there is a pathophysiologically based
of levodopa administration (Sachs et al., 1985; Meco rationale for the employment of domperidone, clini-
et al., 1991; Senard et al., 1995). On the other hand, cal evidence for its beneficial effects on orthostatic
higher doses of stable levodopa or dopamine agonist hypotension is controversial (Merello et al., 1992;
regimes or long-term treatment with levodopa may Lang, 2001). Indeed, a recent study, though demon-
be related to orthostatic hypotension (Camerlingo strating an increase of blood pressure in apomor-
et al., 1990; Senard et al., 1997). The conflicting data phine-treated PD patients, failed to show effects on
may be explained by the fact that, at least in some blood pressure drops in the supine position (Sigurdar-
PD patients, disease-associated pre-existing cardiovas- dottir et al., 2001).
cular dysfunction (Brevetti et al., 1990; Hakamaki
et al., 1998), disease duration and levodopa doses are 32.6.3. Neuropsychiatric side-effects
confounded. Compatible with this, a recent study pro-
vided evidence that levodopa aggravated pre-existent 32.6.3.1. Hallucinations
impairment of the autonomic control of blood pressure Levodopa treatment in PD may cause a number of
and heart rate in de novo patients (Bouhaddi et al., neuropsychiatric side-effects. These include hallucina-
2004). tions, paranoid psychosis and, more rarely, impulsive
The mechanisms of orthostatic hypotension induced and compulsive behavior. Based on the reported
by levodopa administration are complex and incom- controlled studies (Table 32.2), the incidence of hallu-
pletely understood. Lewy body degeneration has been cinations is rather low: 35% during the first 35 years
shown to be present in the sympathetic and parasym- of treatment. However, patients in these trials were a
pathetic autonomic nervous system on various levels selected population early in the disease. In later stages
(Wakabayashi et al., 1993; Kaufmann et al., 2004). of the disease with higher daily levodopa doses and
Peripherally, the sympathetic ganglions and parasym- in patients with additional risk factors (see below),
pathetic cardiac, myenteric and submucosal plexi have levodopa-induced hallucinations and other neuropsy-
been shown to be involved. Centrally, structures such chiatric side-effects develop in 3050% of patients
as the locus ceruleus, the hypothalamus and the dorsal (Sanchez-Ramos et al., 1996; Fenelon et al., 2000; de
vagal nucleus are affected (Wakabayashi and Takaha- Maindreville et al., 2005). Hallucinations are sensory
shi, 1997). Compatible with these findings, evidence perceptions without external stimulation of the rele-
suggests that orthostatic hypotension in PD reflects vant sensory organ (APA, 2000). They are distin-
neurocirculatory failure from generalized sympathetic guished from illusions in which objectively present
denervation (Goldstein et al., 2002; Li et al., 2002). stimuli are perceived and then misinterpreted. Dopa-
Based on these abnormalities, orthostatic hypotension minergically induced hallucinations are most often
induction by dopaminergic agents might be subserved visual; however, they may also present in other sen-
peripherally by activation of postsynaptic D1-receptors sory modalities (Holroyd et al., 2001). One study
mediating vasodilatation and by activation of sym- detected auditory hallucinations in 8% of patients
pathetic presynaptic D2-receptors (Langer, 1980; who mostly also experienced visual hallucinations
Goldberg and Rajfer, 1985). The latter mechanism (Inzelberg et al., 1998). Olfactory or tactile hallucina-
induces reduced vasoconstrictor responses via inhibi- tions have also been reported to be present; however,
tion of norepinephrine release (Bouhaddi et al., 2004). data are sparse and prevalence rates have to be
However, peripheral inhibition of decarboxylation of assumed to be rather low.
40 T. D. HALBIG AND W. C. KOLLER
The development of hallucinations is frequently mesocortical dopaminergic pathways (Agid et al.,
preceded by vivid dreaming which may further 1987). Dopaminergic denervation and chronic levo-
develop into illusions (Pappert et al., 1999; Fenelon dopa treatment may lead to supersensitization of dopa-
et al., 2000). Vivid dreaming may lead to sleep frag- minergic limbic D3- and D4-receptors. Based on
mentation and daytime sleepiness. Illusions are initi- diminished presynaptic buffering capacity, the thera-
ally often present during evenings and at night and peutic non-physiological administration of levodopa
predominantly in twilight and darkness. Sometimes may lead to overstimulation of postsynaptic receptors
patients report hallucinations of presence the physi- and induction of hallucinations (Moskovitz et al.,
cal feeling of the presence of persons or objects near 1978). Compatible with the role of levodopa for the
them, usually behind or beside them. Visual pheno- induction of hallucinations in PD, dopamine receptor-
mena have been described as being simple or complex. blocking agents have been found to alleviate psychotic
Simple elementary hallucinations may be color symptoms in PD patients (Friedman, 1991; Rabey
patches, lightnings or geometrical forms. Most fre- et al., 1995; Fernandez et al., 1999; French Clozapine
quently patients visualize figural appearances of per- Parkinson Study Group, 1999; Parkinson Study Group,
sons or animals and less frequently unanimated 1999). Even though treatment with levodopa and other
objects (Barnes and David, 2001). Figural appearances dopaminergic agents is a contributing factor for the
usually appear normal; however, they may also be development of hallucinations in PD, not all patients
present as distorted or demonic faces or grimaces. treated with dopaminergic agents develop hallucina-
Hallucinations in PD vary on several other characteris- tions. A recent study identified endogen factors
tics. They may be stationary or moving and may rather than the amount of levodopa doses as relevant
develop at any time of the day but preferentially and in the genesis of PD hallucinations (Merims et al.,
with longer episodes in the morning and evening 2004). Compatible with this, there is evidence that
(Barnes and David, 2001). Their duration varies the overall dose of dopaminergic drugs does not
between seconds and hours. The degree of incapacita- significantly differ between patients with and without
tion due to hallucinations varies considerably between hallucinations (Sanchez-Ramos et al., 1996; Klein
patients. In many patients, hallucinations are benign et al., 1997; Aarsland and Karlsen, 1999; Merims
and patients have insight into the nature of their visual et al., 2004). Furthermore, in one study, high doses
experiences. In these patients, hallucinations may of intravenously administered levodopa did not induce
occur sporadically and are often non-threatening. hallucinations in predisposed patients (Goetz et al.,
Hallucinations may even be experienced as entertain- 1998). Several cofactors have been discussed that
ing and be integrated in artwork production. Although may increase the risk for hallucinations. These include
exact data are lacking, patients suffering from benign age, cognitive impairment, depression, sleep disorders,
and infrequent hallucinations may present with a stable PD severity, predominance of axial symptoms, ocular
clinical manifestation over years. However, in many disorders and genetic factors such as the apolipoprotein
patients hallucinatory episodes are experienced as E e 4 allele (Sanchez-Ramos et al., 1996; Inzelberg
frightening. Furthermore, frequently patients demon- et al., 1998; Aarsland and Karlsen, 1999; de la Fuente-
strate lack of insight into the nature of their visual Fernandez et al., 1999; Arnulf et al., 2000; Fenelon
experiences. In many patients, hallucinations increase et al., 2000; de Maindreville et al., 2005). Furthermore,
in frequency of occurrence and severity and may even- treatment with other drugs used in PD therapy, including
tually develop into full-blown psychosis (Sharf et al., dopamine agonists, dopamine-enhancing agents such as
1978). The presence of hallucinations is an important MAO-B and COMT inhibitors, anticholinergic medica-
disease complication and is the most frequent reason tions and amantadine have been shown to contribute
for institutionalization of PD patients (Goetz and (de Smet et al., 1982; Perry et al., 1990; Steckler
Stebbins, 1993; Aarsland and Karlsen, 1999; Holroyd and Sahgal, 1995). Furthermore, there is evidence
et al., 2001). for an involvement of other than dopaminergic trans-
The pathophysiological basis for levodopa-induced mitter systems in the pathogenesis of hallucinations
hallucinations in PD is complex and likely involves in levodopa-treated PD patients. It has been sugges-
widespread disease-associated neuropathological ted that chronic levodopa treatment in conjunction
changes, including Lewy body degeneration and dopa- with serotonergic (5-HT) dysregulation contributes
minergic, cholinergic, noradrenergic and serotonergic to the induction of hallucinations. Neurodegeneration
denervation in association with the effects of levodopa in PD involves brainstem 5-HT neurons associated
supplementation. More specifically, in PD, it is not with diminished central 5-HT levels. Chronic levodopa
only nigrostriatal dopaminergic neurons that degener- administration may further lower 5-HT levels via
ate. There is also degeneration of mesolimbic and reuptake by 5-HT nerve terminals and conversion to
LEVODOPA 41
dopamine (Markianos et al., 1982; Tohgi et al., before bedtime. The administration of clozapine, how-
1993). Compatible with this, the administration of ever, is limited for pragmatic reasons due to its poten-
the 5-HT precursor L-tryptophan may reduce the fre- tial to induce neutropenia. Patients treated with
quency and severity of hallucination in PD (Rabey clozapine require frequent monitoring of white blood
et al., 1977). On the other hand, serotonergic dener- counts, particularly during treatment initiation (Dewey
vation and chronic administration of levodopa have and OSuilleabhain, 2000; Brandstadter and Oertel,
been thought to lead to hypersensitivity of postsynap- 2002; Morgante et al., 2004). Monitoring of white
tic 5-HT receptors (Nausieda et al., 1983). Indeed, blood cell count and absolute neutrophil count should
blockage of serotonergic receptors using methyser- be based on stage of therapy with monitoring weekly
gide or the 5-HT3 antagonist ondansetron appears to for 6 months, every 2 weeks for 6 months and every
alleviate psychotic symptoms in PD patients (Nau- 4 weeks ad infinitum (Novartis Pharmaceuticals
sieda et al., 1983; Zoldan et al., 1995). Corporation, 2005).
Treatment of psychosis in PD can be difficult. Alternatively, quetiapine, another atypical neuro-
Although data on empirical validity are lacking, cur- leptic can be used. Quetiapine also blocks dopaminer-
rently established treatment algorithms usually recom- gic and serotonergic receptors and has affinity to
mend the identification of those agents in the histamine H1 with only small affinity to muscarinergic
individual treatment schema carrying the biggest and cholinergic receptors (Dewey and OSuilleabhain,
potential for the induction of hallucinations. In gen- 2000; Ondo et al., 2005). In a randomized, open-label,
eral, it is thought that agents that are major risk blinded-rater, parallel-group trial, quetiapine and
factors for PD psychosis are those with the weakest clozapine demonstrated equal efficacy and side-effect
antiparkinsonian efficacy. If possible, agents should profile in 45 patients with PD with drug-induced
be eliminated in the following order: anticholiner- psychosis (Morgante et al., 2004). However, a recent,
gics, amantadine, MAO-B inhibitors, dopamine ago- smaller placebo-controlled study failed to demonstrate
nists and finally levodopa (Olanow et al., 2001). significant improvement on psychosis rating scales
Nevertheless, overall reduction of PD medication and (Ondo et al., 2005). The starting dose for quetiapine
in particular of dopaminergic agents is often done at is 2550 mg at bedtime and the dose may be increased
the expense of deterioration of motor functions. In about every third day up to 125 mg/day.
order to prevent worsening of motor functions, the Another atypical antipsychotic medication with low
alternative to decreasing dopaminergic medication is affinity for D2-receptors is olanzapine. However,
the introduction of antipsychotic agents (Merims double-blind, placebo-controlled evaluations of effi-
et al., 2004). cacy and safety profile revealed no significant
In order to avoid extrapyramidal side-effects and improvement of hallucinations but worsening of motor
worsening of PD symptoms, for the treatment of function (Breier et al., 2002; Ondo et al., 2002).
drug-induced psychosis, only atypical neuroleptics Recently, a new class of neuroleptics with function-
should be used. A variety of atypical neuroleptics are ally selective action mechanisms was introduced. For
available. Atypical neuroleptics have lower D2 occu- example, aripriprazole has partial D2 dopamine ago-
pancy than typical neuroleptics and block 5-HT2A nistic and antagonistic characteristics as well as partial
receptors. The latter mechanism may be responsible agonism at 5-HT1A and partial antagonism at 5-HT2A
for their antipsychotic effects (Meltzer, 1999). The receptors (Naber and Lambert, 2004). Its pharmacolo-
current gold standard for the treatment of psychosis gical characteristics suggest a low-profile risk of extra-
in PD is clozapine. Clozapine has relatively low D2 pyramidal side-effects. However, since preliminary
but strong affinity to 5HT2A receptors, as well as affi- case studies reported significant motor side-effects,
nity to histamine H1, muscarinergic and cholinergic aripriprazole cannot currently be recommended as
receptors. Clozapine has a strong antipsychotic effect first-line treatment for drug-induced psychosis in PD
and does not induce motor side-effects (Friedman (Fernandez et al., 2004; Schonfeldt-Lecuona and
and Lannon, 1989a; Kahn et al., 1991; French Cloza- Connemann, 2004).
pine Parkinson Study Group, 1999; Parkinson Study
Group, 1999). Clozapine is slowly titrated, starting 32.6.3.2. Impulse control deficits
with 12.5 mg at bedtime and may be increased to Recently, it has been appreciated that impulse control
75100 mg/day, which usually allows psychotic symp- deficits may develop in PD associated with dopaminergic
toms to be controlled. If necessary, daily doses may be therapy (Vogel and Schiffter, 1983; Uitti et al., 1989;
administered spread out throughout the day. Due to its Fernandez and Friedman, 1999; Molina et al., 2000;
sedating effects, clozapine helps to control psychotic van Deelen et al., 2002; Driver-Dunckley et al., 2003;
symptoms, agitation and sleep disturbance when given Kurlan, 2004). Impulsivity is the failure to resist an
42 T. D. HALBIG AND W. C. KOLLER
impulse, drive or temptation that is harmful to oneself clinical studies have shown that the monoamines
or others (Hollander and Evers, 2001). The Diagnos- 5-HT and dopamine affect sexual behavior (Melis
tic and Statistical Manual of Mental Disorders and Argiolas, 1995; Kafka, 2003). Furthermore, the
(DSM-IV: APA, 2000) classifies conditions such mesolimbic-mesocortical dopaminergic system pre-
as intermittent explosive disorder (failure to resist sumably has an important role in several aspects of
aggressive impulses), kleptomania (failure to resist reward mechanisms and behavioral impulse control
urges to steal items) or pathological gambling (failure which have been shown to be dysfunctional in patholo-
to resist the impulse to gamble despite severe perso- gical gambling (Fiorillo et al., 2003). The occurrence
nal, family or vocational consequences) as impulse of impulse control deficits in PD during dopaminergic
control disorders. Although classified separately, treatment might therefore be interpreted as behavioral
hypersexuality and punding (see below) are other abnormalities induced by overstimulation of dopamine
examples of deficits of impulse control. receptors in denervated mesolimbic structures. Geneti-
Pathological gambling and hypersexuality have cally determined traits might explain why these pro-
recently been reported to be present in PD (Molina blems are induced in only some PD patients (Menza
et al., 2000; Seedat et al., 2000; Gschwandtner et al., et al., 1993; Noble, 2000).
2001; Driver-Dunckley et al., 2003; Montastruc However, for methodological restrictions, the
et al., 2003; Avanzi et al., 2004; Dodd et al., 2005). above-cited studies are inconclusive. Furthermore,
For example, Molina et al. presented a series of 12 data on the prevalence of impulse control deficits in
PD patients (out of a cohort of 250: 4.9%) with patho- the general population are not well established and
logical gambling. Symptoms in 9 patients developed vary depending on the methods of assessment (Shaffer
after initiation of levodopa therapy. Other reports note and Hall, 1996). In summary, due to methodological
the first occurrence of pathological gambling in asso- restrictions and limited data, the implications of these
ciation with a dose increase of dopaminergic drugs findings are still open. Yet, patients rarely volunteer
(Gschwandtner et al., 2001). One retrospective data- to report behavioral abnormalities and their assessment
base study identified 9 out of 1884 patients with patho- is not part of the clinical routine in most movement
logical gambling (Driver-Dunckley et al., 2003). The disorders centers. The prevalence of impulse control
study reports an overall incidence of 0.05% and a deficits induced by dopaminergic agents, including
slightly higher incidence of 1.5% in patients treated levodopa, therefore may be considerable.
with levodopa and a dopamine agonist.
Hypersexuality (paraphilia-related disorder) has 32.6.4. Melanoma
repeatedly been reported in smaller case series and case
studies of PD patients in association with the adminis- Levodopa treatment has been thought to be related to
tration of dopaminergic drugs (Quinn et al., 1983; the development of malignant melanoma. Based on
Vogel and Schiffter, 1983; Uitti et al., 1989; van Deelen several case reports on the co-occurrence of PD and
et al., 2002; Berger et al., 2003). Compulsive behavior malignant melanoma, the diagnosis of malignant mel-
has also been described in PD. Compulsive behavior anoma or the presence of suspicious, undiagnosed skin
is characterized by stereotypical motor behavior accom- lesions has been determined as a contraindication for
panied by an intense fascination with a particular the use of levodopa (Physicians Desk Reference,
object. For example, repetitive handling and examining 2003).
of mechanical objects (punding) have been observed in The putative causal connection between levodopa
single cases of PD and could be related to the overall treatment and malignant melanoma was based on the
dose of dopaminergic medication (Fernandez and fact that levodopa is an important substrate for the
Friedman, 1999; Evans et al., 2004). formation of melanin. Within the melanocyte, tyrosine
These observations have raised concern that dopa- is converted to dopa and then to dopaquinone via
minergic agents, including levodopa, may be asso- the bifunctional enzyme tyrosinase. Dopaquinone
ciated with the induction of impulse control deficits. is further oxidized to form the pigment melanin
Although the mentioned impulse control deficits differ (Kincannon and Boutzale, 1999). It was hypothesized
regarding their phenomenological presentation and that exogenous levodopa may increase the production
neurobiological basis, they have commonalities. For of melanin and promote malignant transformation of
example, compulsive and impulsive control deficits the melanocytes. However, there is no experimental
are characterized by the decreased ability of the indivi- evidence for this assumption. In contrast, in experi-
dual to inhibit motor responses to affective states mental tumor systems with human and murine mela-
which might critically depend on dopaminergic neuro- noma cells, levodopa and dopamine had antitumor
modulation. More specifically, animal research and activity (Wick, 1979, 1980, 1987, 1989).
LEVODOPA 43
Since the first report of malignant melanoma in PD turnover and the conjunction with an AADC shifts per-
in 1972 (Skibba et al., 1972), a total of 54 cases have ipheral levodopa metabolization to the COMT pathway,
been reported in the literature (Weiner et al., 1993; levodopa therapy may lead to increased plasma homo-
Fiala et al., 2003). Interpretation of these rare cases cysteine levels (Morris, 2003).
with respect to a causal link of levodopa administra- Yet, the clinical importance of elevated plasma
tion and tumor induction is difficult for a variety of homocysteine levels in levodopa-treated PD patients is
reasons. Firstly, both PD and malignant melanoma far from understood. First of all, data on atherosclerosis
are highly prevalent conditions, in particular in the and PD are controversial. Several studies found an
elderly population (Thompson et al., 2005). Based on increased risk in PD patients for death from stroke and
the small reported numbers and deficient documenta- coronary artery disease (Gorell et al., 1994; Ben-
tion of cases, it is therefore not possible to determine Shlomo and Marmot, 1995) and an increased risk for
whether co-occurrence of levodopa and malignant intima media thickness of the carotid artery in patients
melanoma is coincidental or not (Fiala et al., 2003). undergoing levodopa treatment (Nakaso et al., 2003).
Secondly, pathogenesis of malignant melanoma and These findings were not confirmed by other studies
the time gap between exposure to carcinogens and (Struck et al., 1990; Korten et al., 2001; Mastaglia
tumor manifestation are thought to take years to dec- et al., 2002; Jellinger, 2003). A reason for these conflic-
ades. Given the variety of risk factors for melanoma, tual findings may be that several other vascular risk fac-
including genetic predisposition with tyrosine kinase tors appear to be less prevalent in PD patients. For
gene mutation, family history, sun exposure, skin example, blood pressure is lower in PD patients
phenotype and history of skin lesion (Thompson (Brevetti et al., 1990; Hakamaki et al., 1998) and smok-
et al., 2005), it is difficult to determine statistical ing is less frequent in PD patients (Hernan et al., 2001).
probabilities for the role of exogenous levodopa. It is therefore conceivable that homocysteine-associated
In conclusion and despite continued warning, based increased risk for stroke and coronary heart disease is
on experimental and epidemiological data, there is at outweighed by other diminished vascular risk factors
present no evidence for a causal relation between (Postuma and Lang, 2004).
levodopa therapy and malignant melanoma. It is well established that PD is a risk factor for the
development of dementia (Emre, 2003). Elevated
32.6.5. Hyperhomocysteinemia plasma homocysteine levels in some, but not all, studies
have been found to increase the risk for Alzheimers
Recently, elevated plasma homocysteine levels have dementia (Seshadri et al., 2002; Luchsinger et al.,
been described in PD patients treated with levodopa 2004), possibly independently of vascular factors
(Allain et al., 1995; Kuhn et al., 1998; Muller et al., (Dufouil et al., 2003; den Heijer et al., 2003). One
1999, 2001; Lamberty and Klitgaard, 2000; Yasui may thus hypothesize that hypercysteinemia in PD
et al., 2000, 2003; Miller, 2002; Miller et al., 2003; may contribute to the development of dementia in PD.
Nakaso et al., 2003; Rogers et al., 2003; Genedani Indeed, a recent preliminary study reported that patients
et al., 2004; OSuilleabhain et al., 2004a). These findings with PD and hyperhomocysteinemia are more likely to
raised the question of whether levodopa treatment is a be depressed and to perform worse on psychometric
risk factor for hyperhomocysteinemia (Postuma and tasks compared with normohomocysteinemic patients
Lang, 2004). Hyperhomocysteinemia in turn is a risk (OSuilleabhain et al., 2004b). However, further
factor for vascular disease (Perry et al., 1995; Bostom research is needed to see whether hyperhomocys-
et al., 1999a, b), possibly for dementia (Seshadri et al., teinemia is a risk factor for neuropsychiatric burden in
2002; Luchsinger et al., 2004; Wright et al., 2004) and patients with PD.
is potentially neurotoxic (Duan et al., 2002). It is thus Experimental and animal research suggested that
possible that the risk for these conditions is increased hyperhomocysteinemia may exert neurotoxic effects on
in PD patients receiving levodopa. the substantia nigra. More specifically, hyperhomocystei-
The mechanisms for increased plasma homocysteine nemia is associated with excitotoxicity via N-methyl-D-
are most likely associated with peripheral metabolization aspartate (NMDA) activation (Lipton et al., 1997) and
of levodopa and more specifically the O-methylation free radical formation (Cook et al., 2002; Lawrence de
of levodopa by COMT (see section 32.3) (Postuma and Koning et al., 2003) and also may have proinflammatory
Lang, 2004). O-methylation of levodopa by COMT effects (Lawrence de Koning et al., 2003). Interestingly,
requires S-adenosylmethionine (SAM) as methyl donor. a study using a mouse model of PD demonstrated that
Demethylation of SAM forms S-adenosylhomocysteine when homocysteine is infused directly into either the
which is converted to homocysteine. Since levodopa substantia nigra or striatum, homocysteine exacerbates
administration leads to increased metabolic levodopa 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-
44 T. D. HALBIG AND W. C. KOLLER
induced dopamine depletion, neuronal degeneration and (LDR) is reflected as gradual improvement of motor
motor dysfunction (Duan et al., 2002). Furthermore, the functions after repeated administration and prolonged
same authors report that homocysteine exacerbates oxi- wearing-off after cessation of therapy (Muenter and
dative stress, mitochondrial dysfunction and apoptosis Tyce, 1971). Benefits from this LDR may even be
in human dopaminergic cells exposed to the pesticide maintained if doses are skipped (Fig. 32.5, early PD).
rotenone or the pro-oxidant ferrous iron (Fe2). How- Apart from the LDR, there is a short-duration response
ever, there are no clinical data on possible neurotoxic which reflects the plasma dopamine concentration
effects relevant for the development of PD or and may be present even early in the disease; however,
hastening of PD progression. it may be masked by the LDR (Nutt et al., 1995). Evi-
Taken together, there is considerable, converging dence suggests that the LDR progressively diminishes
evidence for the potential of levodopa long-term within the course of disease progression. Efficacy of
treatment to increase homocysteine plasma levels. levodopa in terms of duration of symptom control
The latter has been shown to be a risk factor for vas- decreases and eventually becomes a function of the
cular disease and possibly cognitive decline in levodopa plasma concentration with progressively
humans and to be neurotoxic in experimental models shortened (Fig. 32.5, moderate PD), and eventually
of PD. Yet, presently, there is no evidence for the even unpredictable, levodopa responses. Moreover,
clinical relevance of these findings, which certainly involuntary movements (dyskinesias) may occur
merits further investigation. Although it appears associated with levodopa administration (Fig. 32.5,
rational to screen PD patients presenting with or advanced PD). Interestingly, Cotzias et al. (1967), in
being at risk for vascular disease and dementia for their seminal early report on the symptomatic effects
hyperhomocysteinemia and to supplement vitamin B of levodopa in PD, already described involuntary
or COMT inhibitors which have been shown to (athetoid) movements of tongue and extremities
decrease homocysteine plasma levels (Nissinen in those patients who had an excellent response to
et al., 2005; Lamberti et al., 2005), further data are levodopa.
needed before a general guideline regarding screen- Motor complications can seriously interfere with
ing and prevention should be given. activities of daily living and impair quality of life
and their treatment is a challenge (Chapuis et al.,
2004). From a clinical point of view, hypokinetic
32.7. Long-term motor effects: motor
symptoms can be distinguished from hyperkinetic
fluctuations and dyskinesias
manifestations (Table 32.3).
32.7.1. Clinical presentation The first motor complication that developes in
most patients is the wearing-off phenomenon, i.e.
Early during treatment, the response to levodopa is patients experience a re-emergence of symptoms
sustained and extends by far the pharmacological before intake of the next dose. The wearing off may
half-life of 1.5 hours. This long-duration response present clinically in the morning as early-morning

Early PD Moderate PD Advanced PD

Dyskinesia Dyskinesia
Threshold Threshold
Clinical Effect

Clinical Effect

Clinical Effect

Dyskinesia
Response Threshold
Response Threshold Response
Threshold Threshold

Levodopa 2 4 6 Levodopa 2 4 6 Levodopa 2 4 6


Time (h) Time (h) Time (h)
Smooth, extended duration Diminished duration of target clinical
of target clinical response response Short duration of target
Low incidence of dyskinesias clinical response
Increased incidence of dyskinesias
"On" time is associated
with dyskinesias

Fig. 32.5. Motor complications in early, moderate and advanced Parkinsons disease (PD). Pharmacologic response to a levo-
dopa dose (arrow) depending on the stage of disease progression. In the early stage, the response is slow in onset, and has a
small magnitude and a very long duration. In the intermediate stage, the severity of motor dysfunction in the off state has
increased and the response is of greater magnitude and shorter duration. Dyskinesias may be elicited at this stage. In the late
stage, the motor response is abrupt and has a very large magnitude but the duration of response is short and the threshold for
dyskinesia is reduced. Reproduced from Obeso and Olanow (1997), published by Wells Medical, Kent.
LEVODOPA 45
Table 32.3 of symptoms before the beneficial effects kick in
(beginning-of-dose worsening) (Merello and Lees,
Motor complications
1992). Finally, motor functions at the end of the dose
Motor fluctuations interval may deteriorate below the dose interval base-
End-of-dose worsening or wearing off line level for a prolonged time, before the following
 Re-emergence of symptoms at the end of a dose interval dose becomes effective (rebound) (Gancher et al.,
at daytime 1988; Nutt et al., 1988). Related to the onoff motor
 Nocturnal symptoms manifestations, patients may suffer from neuropsy-
 Early-morning symptoms chiatric symptoms which are present in particular dur-
Beginning-of-dose worsening
ing off phases. These may include depression, anxiety
Dose failures
and panic attacks which may be associated with
Unpredictable worsening (on/off)
Freezing marked autonomic symptoms (Menza et al., 1990;
Rebound worsening Siemers et al., 1993). During on periods patients
Hyperkinetic responses may experience hypomania or, less frequently, mania
Peak-dose dyskinesias (Giovannoni et al., 2000).
Square-wave dyskenias Apart from these fluctuations of levodopa response
Diphasic dyskinesias in terms of temporary restoration of motor functions,
Off-period dystonia/early-morning dystonia long-term levodopa treatment may be associated with
involuntary movements which most often present as
dyskinesias (Nutt, 1990) (Table 32.3). Dyskinesias
are usually initially present during on phases. Later
akinesia, throughout the day (wearing-off) or at night on they may occur at virtually any time of the dose
(Fahn, 1974; Marsden et al., 1982; Nutt, 1987). In interval. Dyskinesias are typically choreiform; they
more advanced stages, drug responses may become may, however, also resemble dystonia or athetotic
more and more unpredictable and patients experience movements or akathisia or present as myoclonic jerks
symptomatic periods of varying duration at any time or ballism (Nutt, 1990; Comella and Goetz, 1994;
during the dose interval (onoff). Related to this, Marconi et al., 1994). Dyskinesias can be distin-
delayed onset of dose effects as well as complete guished regarding their temporal relation with the
doseresponse failures occur. Furthermore, patients intake of a levodopa dose (Fig. 32.6). Most frequently,
may experience a levodopa intake-related worsening they present as peak-dose dyskinesias. Peak-dose

Dyskinesia Dyskinesia

"off" "off"

Levodopa Levodopa Levodopa Levodopa Levodopa Levodopa

Diphasic dyskinesia Diphasic dyskinesia


"on" dyskinesia "on" dyskinesia
A B "off" dystonia
Fig. 32.6. Motor complications: dyskinesias. (A) Schematic representation of dyskinesias in patients with Parkinsons disease.
Each levodopa dose (arrow) induces a predictable motor improvement lasting for about 34 hours. Peak-of-dose dyskinesias
(open squares) occur at the time of maximal improvement but may also occupy the entire on period. Diphasic dyskinesias
(black squares) may occur at the beginning and/or the end of the cycle. (B) An example of a patient with an on response
in which the patient fails to respond to a given dose of levodopa, often associated with off-period dystonia (hatched rectan-
gles). Failure to respond to a dose of levodopa tends to occur in the afternoon or evening after repeated doses. Reproduced from
Obeso et al. (2000), with permission from the American Academy of Neurology.
46 T. D. HALBIG AND W. C. KOLLER
dyskinesias tend to occur at the time of peak plasma Schrag and Quinn, 2000; Ahlskog and Muenter,
concentration, usually in the middle between two levo- 2001; Garcia Ruiz et al., 2004).
dopa doses when patients are without typical parkinso-
nian features. Dyskinesias may persist during the 32.7.3. Determining factors for motor complications
entire on period (square-wave dyskinesias). In con-
trast, diphasic dyskinesias occur at the beginning or There is substantial evidence that the disease duration
the end of a levodopa dose interval, i.e. when levodopa is a major factor contributing to the development of
plasma concentrations begin to rise and fall, but not motor complications. According to a meta-analysis
during peak concentrations (Fig. 32.6). Diphasic dys- (Ahlskog and Muenter, 2001), clinical studies on
kinesias more often affect the lower limbs and in par- patients with PD onset well before levodopa avail-
ticular asymmetrically the side of the body that is ability (pre-levodopa era) report high frequencies of
more affected by PD. Movements are often stereotypi- dyskinesias early after treatment initiation, with
cal and may present with dystonic components. In con- approximately half of patients experiencing dyskine-
trast to peak-dose dyskinesias, the presence of diphasic sias after 56 months. This complies with the early
dyskinesias may overlap with parkinsonian features. observations of Cotzias et al. (1967). In contrast, in
Diphasic dyskinesias are thought to be associated with more recent series, frequencies were minimal until
rising or falling, yet insufficient levodopa concentra- 12 years and then amounted up to only 25% through
tions. Finally, severe off states with marked immobi- the first 2.53.5 years. At 46 years of follow-up,
lity may be followed by abrupt onset periods of modern-era series still had lower median dyskinesia
violent dyskinesias (yo-yoing). In contrast, off-period frequencies (3639%). Several other clinic-based and
dystonia develops when levodopa levels are low at the community studies confirmed that longer disease dura-
end of a dose interval or when levodopa doses fail to tions and hence more advanced neurodegenerative
kick in (Fig. 32.6). changes may play a critical role in the development
of levodopa-induced dyskinesias (Lesser et al., 1979;
32.7.2. Time of onset of motor complications Parkinson Study Group, 1996a; Schrag and Quinn,
2000; Kostic et al., 2002).
Various studies examined the time of onset of motor Furthermore, clinical and experimental evidence
complications. Data, however, vary depending on the suggests that time of onset and severity of dyskinesias
methods and patient characteristics. Ideally, in order are associated with the total exposure to levodopa
to examine the effects of long-term levodopa treatment (Agid et al., 1985; Smith et al., 2002). Another factor
for the development of motor complications, prospec- determining onset and frequency of motor complica-
tive long-term studies should compare drug-naive and tions is the age of PD onset. Several studies demon-
levodopa-treated de novo patients over long time inter- strated that motor complications and in particular
vals. Yet, the only study meeting this criterium is the dyskinesias tend to develop earlier in patients with
ELLDOPA study (see section 32.5). Results of the young onset of symptoms and are less likely to occur
ELLDOPA study indicate that motor complications in patients with onset of symptoms after the age 70
may develop after less than 1 year of levodopa therapy (Golbe, 1991; Kostic et al., 1991; Stern et al., 1991;
(Parkinson Study Group, 2004a). Within a treatment Schrag and Quinn, 2000). Another controversial factor,
period of 40 weeks, dyskinesias were significantly however, is gender. Few studies provided evidence for
more frequent in patients treated with a daily dose of higher frequency of dyskinesias in women (Parkinson
600 mg levodopa (15/91) as compared to patients on Study Group, 1996a; Lyons et al., 1998).
placebo (3/90) or patients receiving 150 mg (3/92) or
300 mg (2/88) per day. Likewise, there was a strong 32.7.4. Mechanisms
trend for those patients on 600 mg to present with
more frequent wearing-off phenomenon compared to The mechanisms that contribute to the development of
all other treatment groups. Other studies allowing con- motor complications are controversial. Since there is
clusions regarding onset of motor complications in no change in the peripheral pharmacokinetics of levo-
levodopa-treated patients compared the effects of levo- dopa and dopamine during the course of disease pro-
dopa with other antiparkinsonian agents or are based gression, the underlying mechanisms have to be
on chart reviews (Table 32.4). It is generally agreed assumed to be central. A variety of factors have been
that, after 5 years of treatment, about 50% of patients discussed. For the maintenance of motor functioning,
and after 10 years 80100% of patients have devel- postsynaptic striatal receptors have to be continuously
oped motor complications (Marsden and Parkes, activated (DeLong et al., 1983). Early in the disease,
1977; Burguera et al., 1999; Grandas et al., 1999; remaining endogenous dopamine supplemented by
LEVODOPA 47
Table 32.4
Prevalence of levodopa-induced motor complications

Prevalence of
Study complications Length of study Method of evaluation

Rajput et al. (1984) 10% fluctuations 5 years Physician evaluation


25% dyskinesias
Poewe et al. (1986) 52% wearing off 6 years Webster Scale
54% dyskinesias Modified Columbia Scale
Hely et al. (1994) 41% wearing off 5 years Modified Columbia Scale
55% dyskinesias Dyskinesia Scale
Physician evaluation
Montastruc et al. (1994) 40% wearing off 5 years Columbia Scale
56% dyskinesias UPDRS
Dupont et al. (1996) 59% fluctuations 5 years UPDRS, part 4
41% dyskinesias
Parkinson Study Group (1996b) 50% wearing off 2 years Physician evaluation
30% dyskinesias UPDRS, part 4
Koller et al. (1999) 20% wearing off 5 years Patient diary
20% dyskinesias Physician-recorded questionnaire/diary
Rascol et al. (2000) 45% dyskinesias 5 years UPDRS, dyskinesia scale
Parkinson Study Group (2000) 38% wearing off 2 years Physician determination
31% dyskinesias
Whone et al. (2003) 27% dyskinesias 2 years Physician evaluation
UPDRS, dyskinesia scale
Parkinson Study Group (2004b) 63% wearing off 4 years Physician evaluation
54% dyskinesias UPDRS, dyskinesia scale
Parkinson Study Group (2004a) 21% wearing off 10 months Physician evaluation
7% dyskinesias UPDRS, dyskinesia scale

UPDRS, Unified Parkinsons Disease Rating Scale.


Adapted from Olanow et al. (2001) and supplemented.

exogenous levodopa is sufficient for continuous dopami- monkeys treated with MPTP, where the cell loss
nergic stimulation (CDS). During progressive degenera- amounts to 9095% (Clarke et al., 1987). Compati-
tion and loss of nigrostriatal neurons, the postsynaptically ble with this, compared to patients without motor
available dopamine depends more and more on externally fluctuations, patients experiencing wearing off show
administered dopamine. more pronounced reduction in striatal levodopa
One reason for this is the progressive decrease in uptake (Leenders et al., 1986).
the production of endogenous dopamine. Further- However, motor fluctuations probably also reflect
more, presynaptical reuptake and storage of intrasy- changes of postsynaptic receptor responses to dopami-
naptical dopamine progressively diminish. Due to nergic stimulation. The duration of motor improvements
this loss of presynaptic buffering, fluctuations in due to administration of levodopa or the dopamine ago-
the levodopa plasma levels are reflected as fluctua- nist apomorphine decreases with disease progression,
tions in postsynaptic receptor activations which which cannot be explained by a reduced presynaptic
translate in turn into motor fluctuations. This dopamine storage (Fabbrini et al., 1988; Bravi et al.,
hypothesis is supported by the notion that motor 1994; Verhagen Metman et al., 1997).
fluctuations develop much faster with severe nigros- Apart from the degree of dopaminergic denervation,
triatal degeneration, as demonstrated by the shor- the manner in which missing dopamine is replaced by
tened delay between treatment initiation and dopaminergic agents also appears to contribute to the
emergence of motor fluctuations in PD patients with establishment of motor complications. Dopaminergic
severe disease (Ahlskog and Muenter, 2001) or in agents used for the symptomatic treatment of PD differ
48 T. D. HALBIG AND W. C. KOLLER
regarding their pharmacological half-life. Dopamine strate that the incidence of dyskinesias is higher in
metabolized from levodopa has a half-life of about patients treated with levodopa only than in patients trea-
1.5 hours, whereas dopamine agonists have half-lives ted with a dopamine agonist and hence a dopaminergic
of up to 65 hours (see Ch. 33). Due to its short half-life, agent with a longer half-life.
repeated oral administration of levodopa leads to Different postsynaptic changes related to the nigros-
rapidly changing plasma levels with pulsatile stimula- triatal degenerative process and associated with exter-
tion of postsynaptic receptors. In contrast, continuous nal administration of levodopa that may account for
levodopa infusions or administration of long-acting motor fluctuations and the establishment of dyskinesias
dopamine agonists is associated with stable CDS. have been identified in experimental models. These
Converging evidence from animal models (Bedard include gene changes with upregulation of preproenke-
et al., 1986; Pearce et al., 1998; Maratos et al., 2001) phalin mRNA and downregulation of dynorphin and
as well as from clinical trials (Rascol et al., 2000; substance P as well as changes of Fos genes and prody-
Parkinson Study Group, 2004b) suggests that the short norphin) (Gerfen et al., 1990; Jolkkonen et al., 1995;
half-life of dopaminergic agents importantly contributes Duty and Brotchie, 1997; Calon et al., 2000; Zeng
to the development of motor complications and in et al., 2000; Westin et al., 2001) and GABA-A receptor
particular dyskinesias. upregulation of the globus pallidus internus (Calon
For example, levodopa and short-acting dopamine et al., 1995). Although the exact mechanisms to date
agonists are much more likely to induce dyskinesias are unknown, it is thought, that these gene changes are
in MPTP-treated monkeys than dopamine agonists associated with abnormal firing patterns of the major
with a long half-life (Bedard et al., 1986; Pearce basal ganglia output nuclei and in particular the globus
et al., 1998). Furthermore, there is evidence that the pallidus internus, as assessed during dyskinetic states
potential of the same dopamine agonist to induce (Papa et al., 1999; Lozano et al., 2000).
dyskinesias depends on whether it has been adminis-
tered pulsatile or in a continuous fashion via infusion 32.8. Toxicity
(Blanchet et al., 1995).
Compatible with this are results from several clinical Though levodopa is considered the gold standard for
trials comparing the incidence of dyskinesias in patients the sympomatic treatment of PD symptoms, its long-
treated with levodopa and dopamine agonists term effects on disease progression are unknown.
(Table 32.5). Patients in the dopamine agonist treatment Different levels of research, including in vitro studies,
arm in these studies usually received supplementary in vivo animal experiments and clinical and neuroima-
levodopa if symptoms remained inadequately con- ging data obtained in humans, raised the possibilities
trolled. Results of these studies consistently demon- of both neurotoxic and neuroprotective effects of
levodopa.
Table 32.5 There is conclusive evidence that oxidative stress is
present in the substantia nigra pars compacta (SNpc)
Prevalence of dyskinesias in patients treated with either in PD (increased Fe2: Dexter et al., 1987, 1989; Sofic
levodopa or dopamine agonists levodopa
et al., 1991; decreased glutathione: Perry et al., 1982;
Riederer et al., 1989; decreased mitochondrial complex
Length of Prevalence of
study dyskinesias 1 activity: Parker et al., 1989; Schapira et al., 1990;
Study (years) Agent (%) Tretter et al., 2004). Furthermore, oxidative stress has
been shown to impair DNA (Schapira et al., 1990),
Rascol et al. 5 Ropinirole 20 proteins (Parker et al., 1989; Tretter et al., 2004) and
(2000) Levodopa 45 lipids (Emerit et al., 2004). On the other hand, in vitro
Parkinson 2 Pramipexole 10 evidence suggests that under certain conditions both
Study Group Levodopa 30
dopamine and levodopa may exert toxic effects on
(2000)
cultured dopaminergic neurons (Mena et al., 1992;
Whone et al. 2 Ropinirole 3.4
(2003) Levodopa 27 Mytilineou et al., 1993) via auto-oxidation, leading to
Parkinson 4 Pramipexole 24.5 the formation of highly reactive oxygen species
Study Group Levodopa 54 (Graham et al., 1978). Compatible with this, high doses
(2004b) of dopamine and levodopa have been shown to decrease
Rinne et al. 4 Cabergoline 6* TH-positive staining neurons in various dopaminergic
(1998b) Levodopa 14* cell cultures (Michel and Hefti, 1990; Steece-Collier
et al., 1990; Mytilineou et al., 1993), probably via apop-
*Peak-dose dyskinesias. totic mechanisms (Ziv et al., 1997; Corona-Morales
LEVODOPA 49
et al., 2000). Based on these findings, it is reasonable to (Parkinson Study Group, 2002a; Whone et al., 2003).
assume that the increased oxidative stress already pre- As a surrogate marker for disease progression, neuroi-
sent in PD is potentiated by chronic administration of maging measures indicative of the functional integrity
dopaminergic agents. of the nigrostriatal system were used. For example, in
However, for a variety of methodological reasons, the an imaging substudy of the CALM-PD trial (Compar-
in vitro findings supporting the neurotoxic potential of ison of the Agonist pramipexole vs. Levodopa on
dopaminergic agents do not simply translate into in vivo Motor complications in Parkinsons Disease), 82 de
effects. For example, the toxic concentration of levodopa novo PD patients were randomized to receive either
used in most studies was much higher than concentrations 300 mg levodopa or 1.5 mg pramipexole per day
usually achieved by therapeutic levodopa supplementation. (Parkinson Study Group, 2002a). Open-label levodopa
Furthermore, most in vitro studies lack neuroprotective fea- could be added, if clinical symptoms could not be suf-
tures such as antioxidant ascorbic acid or glial cells which ficiently controlled by the study drug. The main out-
are present in in vivo animal models as well as in humans. come variable was the change of iodine-123-labeled
Indeed, it has been demonstrated that culture systems that 2-beta-carboxymethoxy-3-beta-(4-iodophenyl)tropane
were accommodated for these factors were protected (b-CIT) uptake on single-photon emission computed
against levodopa toxicity (Kalir and Mytilineou, 1991; tomography (SPECT) from baseline to month 46 after
Mena et al., 1993, 1997a; Pardo et al., 1993). Interestingly, trial initiation. Results of the study showed that the 41
low-dose levodopa administration may exert neurotrophic patients treated initially with pramipexole had a signifi-
effects in in vivo models using other pro-oxidant agents. cantly slower mean decline in striatal b-CIT uptake
For example, in cultures using glia-conditioned media, (16.0%) than the 42 subjects treated initially with levo-
levodopa was shown to enhance neurite outgrowth and to dopa (25.5%). Interestingly, mean total and motor
increase cell survival (Mytilineou et al., 1993; Mena et al., UPDRS scores obtained in the defined off state in both
1997a, b). Furthermore, levodopa may protect mesence- the levodopa- and the pramipexole-treated groups did
phalic cultures against oxidative stress, probably via upre- not differ significantly between baseline and assess-
gulation of glutathione (Han et al., 1996). Taken together, ments at 34 or 46 months.
in vitro data are at present inconclusive. Another prospective, randomized trial, the REAL-
In vivo animal studies also tried to assess the putative PET study (Requip as Early Therapy versus L-dopa
cell toxic effects of levodopa supplementation Positron Emission Tomography) (Whone et al., 2003),
in experimental models of PD. One study evaluated the assessed the neuroprotective potential of ropinirole
effect of 6-month oral levodopa treatment on several using the rate of loss of dopamine-terminal function in
dopaminergic markers in rats with moderate or severe 162 de novo patients over 24 months, as assessed by
6-hydroxydopamine (6-OHDA)-induced lesions of fluoro-dopa-PET as primary outcome measure. Patients
mesencephalic dopamine neurons and sham-lesioned were randomized to receive either ropinirole or levo-
animals (Murer et al., 1998). Counts of TH-immunoreac- dopa. Over the first 4 weeks of the study, doses were
tive neurons in the substantia nigra and ventral tegmental escalated to daily regimens of 3 mg ropinirole, or
area showed no significant difference between levodopa- 300 mg levodopa per day. Titration was flexible, based
treated and vehicle-treated rats. In addition, for rats of the on clinical response and tolerability, to a maximum
sham-lesioned and severely lesioned groups, immunora- 24 mg of ropinirole or 1000 mg of levodopa per day. If
diolabeling for TH, DAT and vesicular monoamine trans- symptoms were inadequately controlled, titration to
porter (VMAT2) at the striatal level was not significantly the maximum tolerated dose of the assigned medication
different between rats treated with levodopa or vehicle. was encouraged. The primary outcome measure was
Unexpectedly, quantification of immunoautoradiograms reduction in putamen 18F-dopa uptake (Ki) between
showed a partial recovery of all three dopaminergic mar- baseline and 2-year PET. Analysis of the putamen as
kers (TH, DAT and VMAT2) in the denervated territories the central region of interest revealed that there was a
of the striatum of moderately lesioned rats receiving levo- significantly slower rate of deterioration in the ropinir-
dopa. Compatible with these neuroprotective effects, ole group than in the levodopa group (13% versus
another study, also using a PD rat model with 6-OHDA- 20%). Interestingly, UPDRS measures indicated that
induced dopaminergic lesions and chronic levodopa motor function during treatment at 2 years was superior
treatment for about 6 months (Datla et al., 2001), also with levodopa compared with ropinirole.
revealed an increased number of dopaminergic neurons. Both the CALM-PD and REAL-PET studies
Recently, two prospective randomized clinical trials demonstrated a more rapid decline on imaging biomar-
have compared the effects of levodopa versus the kers of nigrostriatal functioning in patients treated with
non-ergot derivative D2-binding dopamine agonists levodopa. Since there was no placebo group in both
pramipexole and ropinirole on disease progression studies, it is not clear whether the different rate of
50 T. D. HALBIG AND W. C. KOLLER
decline reflects toxic effects of levodopa or rather neu- DAT. Thus, the results of the ELLDOPA study with
roprotective effects of the dopamine agonists. Further- diverging clinical and controversial imaging findings
more, although the same results were seen in both are not conclusive.
studies despite the use of different agonists, different In summary, even though there is some controversial
tracers and different imaging techniques, it is possible evidence from laboratory and human imaging studies
that levodopa and dopamine agonists have different suggesting a toxic effect of levodopa, in the final ana-
pharmacological effects and regulate components of lysis it remains unclear whether levodopa is toxic.
the nigrostriatal system in opposite directions In contrast, there is no clinical evidence supporting the
(Ahlskog et al., 1999). In this case, the different notion of levodopa toxicity and some data even support
SPECT and PET findings would simply reflect these the possibility of neuroprotective effects of levodopa.
pharmacological differences rather than indicate dif-
ferent rates of disease progression. It is also not clear
32.9. Strategies to improve levodopa therapy
why the apparently more rapid decline on PET and
SPECT in the levodopa groups did not reflect clini- 32.9.1. Oral formulations aiming at faster onset of
cally. Indeed, UPDRS motor scores in patients treated symptomatic effects
with levodopa were equal or superior to scores
obtained in patients treated with agonists. Several different approaches have been developed to face
The ELLDOPA (see section 32.5) study was the first the shortcomings and failures of levodopa therapy. Even
study ever comparing the effects of levodopa with pla- in earlier stages of PD, the time to effect of a single dose
cebo. The aim of the ELLDOPA study was to deter- of levodopa may be delayed for various reasons. The
mine the effect of levodopa on the rate of progression most important factor is usually related to the speed of
of PD (Parkinson Study Group, 2004a). The ELLDOPA gastric emptying, which varies and may be prolonged
study evaluated clinical and imaging functions in 361 in PD (Edwards et al., 1992; Byrne et al., 1994; Kaneoke
patients randomized to receive a daily carbidopa-levo- et al., 1995; Quigley, 1996; Pfeiffer, 1998, 2003; Hardoff
dopa dose of 37.5 and 150 mg, 75 and 300 mg, or et al., 2001; Goetze et al., 2005). Furthermore, food may
150 and 600 mg, respectively, or a matching placebo delay gastrointestinal transit time and LNAA from
for a period of 40 weeks. Patients then underwent with- protein-rich food may lead to interference and delay of
drawal of treatment for 2 weeks. The primary outcome transmucosal and transendothelial levodopa transport
was a change in scores on the UPDRS between baseline (Djaldetti et al., 1996) (see section 32.3). In later stages
and 42 weeks. Neuroimaging studies of 142 subjects of the disease, swallowing may also be a problem, further
were performed at baseline and at week 40 to assess delaying the time until a levodopa dose kicks in. To
striatal dopamine transporter density by measuring b- shorten the time to therapeutic effect, conservative mea-
CIT uptake. The clinical component of the study sures such as avoiding food and in particular protein-rich
demonstrates that the severity of symptoms increased diets 45 minutes before and 90 minutes after intake of a
more in the placebo group than in all the groups receiv- levodopa dose may be helpful. Furthermore, special for-
ing levodopa. The mean difference between the total mulations of levodopa and new forms of administration
score on the UPDRS at baseline and at 42 weeks was of levodopa have been developed.
7.8 units in the placebo group, 1.9 units in patients In order to speed up levodopa transfer to the jeju-
receiving levodopa at a dose of 150 or 300 mg daily num, the major site of resorption, several studies com-
and 1.4 in those receiving 600 mg daily. This result pared the effect of liquefied levodopa with standard
is consistent with a protective, not a toxic, effect. How- oral administration (Kurth et al., 1993; Metman et al.,
ever, the possibility of a confounding, long-lasting, 1994; Pappert et al., 1996; Kurth, 1997). In a double-
symptomatic effect that persists for months after wash- blind, cross-over study (Pappert et al., 1996), either
out cannot be entirely excluded. In contrast, the imaging liquid levodopa or standard levodopa was administered
component of the study demonstrated that, in a sub- to 23 PD patients. Liquid levodopa was produced using
group of 116 patients, the mean decline in the b-CIT 10 tablets of levodopa/carbidopa (LD/CD: 100/25 mg)
uptake was significantly greater with levodopa than and 1 g of ascorbate dissolved in 1 liter of tap water.
placebo (6% among those receiving levodopa at Patients were evaluated hourly between 9 a.m. and
150 mg daily, 4% in those receiving it at 300 mg daily 4 p.m. for plasma levodopa levels. Furthermore, the
and 7.2% among those receiving it at 600 mg daily, as UPDRS, a dyskinesia rating scale and onoff ratings
compared with 1.4% among those receiving placebo). were employed. Patients receiving liquid LD/CD
Again, as in the CALM-PD and REAL-PET studies, ingested significantly higher doses and had significantly
the imaging findings suggest either a toxic effect of improved motor function and total on time, without an
levodopa or a pharmacologic modification of the increase in dyskinesia severity. Levodopa levels and
LEVODOPA 51
variability were equivalent with the two formulations. concentration time curve (AUC: 0.5 hours). Apparent
However, overall dosing frequencies in this study were elimination half-life was similar for both formulations.
much higher for the liquid preparation (17.22 versus Other recent strategies to shorten Tmax used chemi-
6.96) and the overall ingested levodopa dose was higher cally modified levodopa preparations (Stocchi et al.,
(1321 versus 890 mg). In an open trial comparing a 1994, 1996; Djaldetti et al., 2002, 2003). For example,
levodopa/carbidopa/ascorbic acid solution (LCAS) and levodopa ethylester (etilevodopa, LDEE) is a highly
standard levodopa, 4 patients with PD with severe soluble prodrug of levodopa passing unchanged
motor fluctuations presented with reduced bradykinesia, through the stomach to the duodenum, where it is
decreased dysfunctional dyskinesias and increased func- rapidly hydrolyzed by local esterases and absorbed as
tional on time when treated with LCAS (Kurth et al., levodopa. In a recent open-label, randomized, four-
1993). According to the authors, oral LCAS allowed way cross-over study, different oral preparations of
better titration of levodopa dosage and offered a more LDEE/carbidopa and LD/CD were compared in 29
predictable response than LD/CD tablets. Compatible PD patients with motor fluctuations (Djaldetti et al.,
with this, in a small double-blind, placebo-controlled, 2002, 2003). Results showed significantly shorter
cross-over study in 5 PD patients, orally administered plasma levodopa Tmax, higher AUC and Cmax for
liquid LD/CD led to slightly earlier peak plasma LDEE/carbidopa. Compatible with these results, the
levodopa levels (Metman et al., 1994). same authors were able to demonstrate the clinical
A dispersible formulation of levodopa (LD)/benser- superiority of LDEE in 62 patients with delayed-on
azide (Madopar LT) is an alternative to the prepara- and no-on subtypes of response fluctuation in a con-
tions of liquefied levodopa, as described above. trolled double-blind study (Djaldetti et al., 2002).
Madopar LT can be dissolved in water or taken like Results showed that mean latency to turning on was
a regular tablet and is available in Europe (Contin reduced by 21% (morning dose) and 17% (postlunch
et al., 1999). It is characterized by earlier release, rapid dose) and percentage of no-on episodes after the post-
absorption and shorter time to peak plasma levodopa lunch dose was decreased by 21% in the LDEE/CD
concentration (Tmax) with no difference in remaining group but increased by 36% in the LD/CD group.
pharmacokinetic and clinical characteristics (Haglund Levodopa methyl ester is another highly water-soluble
and Hoogkamer, 1995). Several open-label trials have levodopa derivative that can be administered orally
shown its usefulness in the treatment of early-morning and intravenously and has been shown to decrease
akinesia (Steiger et al., 1992) and in patients with latency to on in smaller studies (Juncos et al., 1987;
swallowing difficulties (Bayer et al., 1988). In sum- Stocchi et al., 1994, 1996).
mary, even though controlled clinical data are sparse, Recently, an orally disintegrating LD/CD tablet
based on the shorter Tmax, dispersible and liquid for- containing phenylalanine has been introduced in the
mulations may be worthwhile to shorten the delay to USA (Parcopa) (Nausieda et al., 2005). It rapidly dis-
effect and may be offered to patients as first dose integrates on the tongue without chewing and does
in the morning, as rescue medication or to shorten not require water to aid dissolution. It is available in
diphasic dyskinesias. the USA in dosages corresponding to the usual
A dual-release preparation has been developed and is dosages of standard levodopa preparations. According
currently marketed in Switzerland. In a double-blind to an open-label study, the clinical efficacy of orally
single-dose study 200 mg levodopa (plus 50 mg benser- disintegrating LD/CD tablets is equivalent to standard
azide) was administered either as dual-release prepara- preparations (Nausieda et al., 2005). This new formu-
tion consisting of a three-layer tablet combining lation may be an alternative to standard levodopa for
immediate and slow-release properties or a conven- patients who have difficulties swallowing conventional
tional slow-release formulation (see section 32.9.3) tablets or when water is unavailable or inappropriate.
(Descombes et al., 2001). The cross-over study enrolled Finally, nasal and transdermal formulations have
16 fluctuating patients with PD. Compared to the slow- been evaluated in rats in in vivo and in vitro studies;
release formulation, the time to on was shown to be however, clinical evidence in humans is lacking (Sudo
shorter with the dual-release formulation (43  31 and et al., 1998; Kao et al., 2000; Iwase et al., 2000).
81  39 minutes, respectively), whereas the mean
time to relapse to off was similar for both for- 32.9.2. Infusions and other non-oral ways of
mulations. UPDRS improvement and dyskinesias levodopa administration
scores were similar for either preparation. There were
no differences in tolerability. The dual-release formula- In order to control better motor fluctuations related to
tion had significantly shorter Tmax and greater maxi- the short levodopa half-life, several strategies are
mum concentration (Cmax) and area under the plasma available to provide more continous dopaminergic
52 T. D. HALBIG AND W. C. KOLLER
stimulation. For example, constant intravenous infu- keted as Sinemet CR and Madopar CR (or HBS, or
sion of levodopa replacing standard oral levodopa or Depot). Madopar CR, when in contact with gastric fluid
in addition to standard levodopa has been shown to and after dissolution of the gelatin shell of the capsule,
result in stable plasma dopa concentrations and dra- forms a mucous body that continuously releases its
matic improvement of motor fluctuations in patients contents via a hydrated layer by diffusion over a pro-
with predictable wearing off, unpredictable onoff longed period of time (Erni and Held, 1987). Sinemet
fluctuations and prolonged offs (Shoulson et al., CR is embedded in an erodible polymeric matrix and
1975; Quinn et al., 1982). dissolves during passage along the duodenal-jejunal
Furthermore, liquid solutions may be used in order mucosa (LeWitt et al., 1989; Yeh et al., 1989).
to bypass the oral route. Bypassing the oral route may Due to their gradual release, CR preparations are
be necessary in patients with swallowing difficulties characterized by delayed Tmax by about 4590 minutes
or in order to smoothen the variable and sometimes and by prolonged plasma half-life of up to 2 hours.
erratic time till levodopa kicks in which is often caused Furthermore, due to variable gastrointestinal transit, less
by delayed gastric emptying. Liquid solutions have efficient absorption in the distal bowel or greater first-
been used in patients with percutaneous endoscopic gas- pass systemic metabolism, bioavailability is decreased
trotomy, with a portable duodenal or jejunal pump or by about 30% compared to standard levodopa (Yeh
via nasoduodenal catheter (Cedarbaum et al., 1990a; et al., 1989). Finally, Cmax is reduced (Grahnen et al.,
Metman et al., 1994; Syed et al., 1998). For enteral 1992; for Madopar see also Crevoisier et al., 1987; Da
application, a stable gel suspension of LD/CD (20/5 Prada et al., 1987; Jensen et al., 1987; Malcolm et al.,
mg/ml) has been developed (Duodopa) (Nilsson et al., 1987; Nordera et al., 1987; Poewe et al., 1987; Quinn
1998, 2001; Nyholm et al., 2003). Continuous intraduo- et al., 1987; for Sinemet see also Cedarbaum et al.,
denal infusion has been shown to be associated with 1987; Yeh et al., 1989; Wilding et al., 1991).
stable plasma levodopa concentrations compared to
controlled-release (CR) preparations (see section 32.9.3.1. Effects on motor fluctuations
32.9.3). Compatible with this, on time increased and The effects of CR preparations on motor fluctuations
off time and dyskinesias decreased (Nyholm et al., are mixed. Overall, there are fewer data available on
2003, 2005). Long-term evaluations for more than the effects of CR LD/benserazide than for LD/CD pre-
9 years demonstrated that continuous intraduodenal parations. A double-blind, cross-over study in 14
levodopa infusion may be efficient in patients with patients with PD and motor fluctuations examined
advanced disease and severe onoff fluctuations the effects of Madopar HBS or placebo twice a day
(Nilsson et al., 2001). Patients are usually given a bolus in addition to the optimal standard Madopar treatment
as the first morning dose and then continuous infusion (De Michele et al., 1989). Clinical response as evalu-
during the day. In addition to decreased motor fluctua- ated by the Kings College Hospital Parkinsons Dis-
tions, infusion therapy permits the reduction of the daily ease Rating Scale and the Mobility in Bed Scale
overall dose of levodopa. improved significantly, whereas evaluation by self-
In contrast, rectally administered levodopa was scoring diaries did not show significant changes.
shown to be of no value in patients who are unable Another open-label study on 22 patients reported longer
to take oral medication. In a series of 12 PD patients on periods, less severe off periods and decrease in
postsurgery there was no rise in levodopa plasma early-morning akinesia (Chouza et al., 1990).
levels and no clinical benefit when levodopa was Compatible with the results obtained for LD/ben-
administered rectally (Eisler et al., 1981). serazide, several double-blind (Hutton et al., 1989;
Mark and Sage, 1989a; Lieberman et al., 1990; Wol-
32.9.3. Levodopa slow-release preparations ters et al., 1992; Wolters and Tesselaar, 1996) and
open studies (Goetz et al., 1988; Deleu et al., 1989;
In order to address the fluctuating motor response after Rondot et al., 1989; Rodnitzky et al., 1989; Pahwa
long-term treatment with levodopa and in particular et al., 1993) demonstrated significant off-time reduc-
the wearing-off phenomenon, controlled/slow-release tion and improvement of functional clinical abilities for
levodopa preparations have been developed. Their LD/CD CR preparations. In particular, the combined
specific aim is to extend the short half-life of levodopa administration of controlled- and immediate-release for-
to prolong its clinical effects. CR preparations com- mulations allowed significant improvement regarding
prise levodopa/AADC embedded in a slowly eroding disability from dyskinesias and number of on hours
matrix (Erni and Held, 1987; LeWitt et al., 1989; without dyskinesias (Sage and Mark, 1988; Friedman
Yeh et al., 1989). CR preparations are available in and Lannon, 1989b; Mark and Sage, 1989b; Wolters
combination with carbidopa or benserazide and mar- et al., 1992).
LEVODOPA 53
Other possible advantages of CR preparations are levodopa (plus carbidopa or benserazide) can be transi-
less frequent dose failures (Pahwa et al., 1993) and ently useful in moderately advanced stages of PD with
diminished early-morning dystonia and nocturnal awa- motor fluctuations characterized mainly by the wear-
kenings (Goetz et al., 1989; Pahwa et al., 1993). ing-off phenomenon and nocturnal and early-morning
However, other studies failed to provide evidence motor symptoms. In contrast, in later stages CR pre-
for the superior efficacy of slow-release Madopar in parations may contribute to dopaminergic side-effects
improving nocturnal and early-morning disabilities. such as dyskinesias.
For example, in a double-blind cross-over study, 103
patients with nocturnal and/or early-morning disabilities 32.9.3.2. Effects in de novo patients and prevention
received a bedtime dose of either Madopar CR or of motor complications
standard Madopar in addition to their usual daytime According to the concept of CDS, non-physiologic,
levodopa regimen. Both Madopar CR and standard pulsatile receptor stimulation contributes to the devel-
Madopar improved nocturnal and early-morning dis- opment of motor fluctuations and dyskinesias. Based
ability equally (UK Madopar CR Study Group, 1989). on this consideration, the administration of levodopa
Likewise, for LD/CD CR, studies in patients with motor CR preparations has been suggested as pre-emptive
fluctuations were not able to detect significant differ- strategy to prevent or delay the development of motor
ences between standard formulation and CR regarding complications. Two controlled prospective multicenter
number of off hours (Ahlskog et al., 1988) or effects trials assessed the potential of CR preparations to
on wearing off (Feldman et al., 1989). reduce the incidence of motor complications compared
The long-term efficacy of CR preparations is also with immediate-release LD/CD (Block et al., 1997;
controversial. Although some studies found efficacy Koller et al., 1999) or LD/benserazide (Dupont et al.,
for several months only (Pezzoli et al., 1988; Klee- 1996). The primary endpoints of these studies were
dorfer and Poewe, 1992), other investigators report the incidence of motor fluctuations and dyskinesias
satisfactory symptom control for more than 2 years after 5 years of treatment. Though both treatment
with Madopar CR (Siegfried, 1987), in particular for groups responded well to therapy and CR-treated
mild to moderate fluctuations or in addition to regular patients were found to score better in terms of measures
levodopa (Pacchetti et al., 1990). of activities of daily living (Dupont et al., 1996; Block
Another controversial issue is the effect of CR pre- et al., 1997), there was no significant difference regard-
parations on dyskinesias. Although some authors ing the primary endpoints between both treatment
report increase in peak-dose or diphasic dyskinesias groups: About 20% of patients presented with motor
for LD/benserazide (Pezzoli et al., 1988; De Michele complications after 5 years of continuous treatment.
et al., 1989; Kleedorfer and Poewe, 1992) and LD/ Thus, compared to standard levodopa, CR levodopa
CD (Jankovic et al., 1989; Cedarbaum et al., 1990b; has not proven to be efficacious in preventing or delaying
Hutton and Morris, 1991), others report decrease in the development of motor complications.
dyskinesias (Chouza et al., 1990).
In summary, evidence for the efficacy of CR pre-
32.10. Dopamine-enhancing agents
parations to control motor fluctuations is only derived
from studies with standard levodopa as comparator 32.10.1. Monoamine oxidase-B inhibitors
and not including comparisons with placebo. In general,
studies report a reduction in dose frequency (Hutton To increase central dopaminergic availability and
et al., 1989; Lieberman et al., 1990). However, most, to extend levodopa half-life, different agents that
but not all, studies (Dupont et al., 1996) report an block the breakdown of levodopa and dopamine
increase of overall doses of levodopa by 30%, which have been introduced into clinical practice. One thera-
is consistent with the pharmacokinetic profile of peutic principle to block the biotransformation of
CR preparations. Furthermore, efficacy in reducing dopamine is the selective inhibition of MAO-B (see
off time, dose failures, nocturnal and early-morning section 32.2.2 this chapter and Ch. 34). MAO-B inhi-
akinesia and rigidity may be transient. A problem bitors block the deamination of residual endogenous
related to prolonged on- and offset of clinical effects dopamine as well as dopamine synthesized from
may be the development or worsening of diphasic dys- externally administered levodopa. However, several
kinesias. Furthermore, associated with erratic levodopa other mechanisms, including reuptake inhibition and
resorption, levodopa plasma levels may build up unpre- action on presynaptic autoreceptors, are involved in
dictably, with variably high peak dose levels, which the enhancement of dopaminergic neurotransmission
may translate into peak-dose dyskinesias. In conclusion, by MAO-B inhibitors (Riederer et al., 1978). Selegi-
the administration of the CR levodopa formulation of line and rasagiline are both irreversible inhibitors of
54 T. D. HALBIG AND W. C. KOLLER
MAO-B with mild antiparkinsonian effects when used (Keranen et al., 1994; Heikkinen et al., 2001) and 1.7
as monotherapy or in conjunction with levodopa in and 1.8 hours for tolcapone (Dingemanse et al.,
early PD (Parkinson Study Group, 1989, 2002b; 1995b; Jorga et al., 1998b).
Pahwa et al., 1993; Lees, 1995; Block et al., 1997; Both agents are highly metabolized and only 0.5% of
Iwase et al., 2000) as well as in fluctuating patients an oral dose is excreted with the urine. Degradation is
(Rabey et al., 2000; Parkinson Study Group, 2005; primarily via hepatic glucuronidation. Tolcapone is
Rascol et al., 2005). Furthermore, there is evidence primarily eliminated via the biliary route and about
that the use of MAO-B inhibitors may delay the need 40% of orally administered doses is excreted in the feces
for dopaminergic therapy (Parkinson Study Group, (Wikberg et al., 1993b; Jorga et al., 1999; Heikkinen
1989). Whether this results from mild symptomatic et al., 2001; Valeant Pharmaceuticals International,
effects of MAO-B inhibition or rather from neuropro- 2006). Entacapone is almost entirely glucuronized in the
tective, PD progression-delaying effects, as suggested liver and eliminated by the kidneys and via the biliary
by recent studies (Olanow et al., 1995; Parkinson route, undergoing enterohepatic circulation (Wikberg
Study Group, 2004c), is debated. For a more indepth et al., 1993a; Jorga et al., 1998b; Heikkinen et al., 2001).
evaluation of MAO-B inhibitors in the treatment of Pharmacokinetics of both COMT inhibitors is not
PD, see Chapter 34. significantly affected by co-administration of levodopa
and AADC inhibitors (Dingemanse et al., 1995b; Smith
32.10.2. Catechol-O-methyltransferase inhibitors et al., 1997). There is no difference in pharmacokinetics
between healthy young and old subjects and PD patients
Another therapeutic principle that aims at the enhance- and no accumulation in plasma takes place during
ment of externally administered levodopa is the inhibi- chronic administration of both COMT inhibitors (Din-
tion of COMT. Levodopa is mainly metabolized by gemanse et al., 1995b; Jorga et al., 1997). Elimination
AADC and COMT and, when administered orally, half-life for entacapone is shorter than for tolcapone
only about 1% reaches the CNS to supplement missing (0.40.7 hours (Keranen et al., 1994; Heikkinen et al.,
dopamine in PD (see section 32.3). The previous intro- 2001) and 23 hours (Jorga, 1998a), respectively).
duction of AADC inhibitors blocked one of the main Entacapone is not lipophilic and does not cross the
metabolic pathways, allowing a dramatic levodopa bloodbrain barrier. It thus exerts its effects on COMT
dose reduction of up to 75%. By further blocking the only peripherally (Nutt, 1998; Troconiz et al., 1998). In
remaining major peripheral metabolic pathway contrast, tolcapone is able to penetrate the bloodbrain
(COMT), central levodopa bioavailability could be barrier, as shown in rodents and monkeys (Zurcher
significantly further increased. Two different COMT et al., 1990), as well as by 18F-dopa PET imaging in
inhibitors, tolcapone and entacapone, have been PD patients (Ceravolo et al., 2002). Nevertheless, the
approved in the 1990s as adjunctive therapy to levo- main clinical effects of both drugs are thought to be
dopa. Both agents have been shown to result in more mediated by peripheral actions (Nutt, 2000).
CDS with potentiated and prolonged clinical effects Entacapone and tolcapone inhibit COMT in a dose-
of each single dose of levodopa. dependent fashion (Jorga, 1998b). Compatible with their
pharmakokinetic differences, COMT inhibition differs
32.10.2.1. Catechol-O-methyltransferase inhibition: between entacapone and tolcapone. As reflected by inhi-
pharmacokinetics and dynamics bition of COMT activity in red blood cells, a measure of
Both COMT inhibitors are rapidly absorbed and their peripheral COMT inhibition, 200 mg entacapone inhibits
Cmax is reached within less than 2 hours (Keranen COMT by 60% at peak plasma concentration and by 10%
et al., 1994; Dingemanse et al., 1995b; Jorga, 1998a). 4 hours after administration. In contrast, 200 mg tolca-
Oral bioavailability is about 60% for tolcapone (Jorga pone inhibits COMT by 80% at peak plasma concentra-
et al., 1998b) and between 30% and 46% for entaca- tion and by 70% 4 hours after intake (Jorga, 1998b).
pone (Keranen et al., 1994; Heikkinen et al., 2001). The effects of both entacapone and tolcapone on COMT
Cmax and AUC are higher for tolcapone (Keranen are fully reversible, as studied in in vitro models (Lotta
et al., 1994; Dingemanse et al., 1995b). Entacapone et al., 1995; Borges et al., 1997). COMT is recovered
and tolcapone are highly protein-bound (98% and within 8 hours (entacapone) and 16 hours (tolcapone),
99.9%, respectively). At therapeutic oral doses (see respectively (Keranen et al., 1994; Dingemanse et al.,
Table 32.6) both COMT inhibitors have an elimination 1995b). COMT-inhibitory effects do not decrease after
half-life between 1.6 and 3.4 hours (Keranen et al., repeated administration (Dingemanse et al., 1996;
1994; Dingemanse et al., 1995b; Jorga et al., 1998b; Ruottinen and Rinne, 1996a; Jorga et al., 1997) and are
Heikkinen et al., 2001). Tmax upon oral administration maintained independently of PD progression (Ruottinen
of 100 or 200 mg is 0.6 or 0.7 hours for entacapone and Rinne, 1996a).
LEVODOPA 55
32.10.2.2. Effects of catechol-O-methyltransferase Wurtman, 1993; Tornwall et al., 1994; for tolcapone, see
inhibitors on levodopa pharmacokinetics Acquas et al., 1992; Napolitano et al., 1995).
Both COMT inhibitors increase the AUC without
increasing Cmax for levodopa, as shown in healthy 32.10.2.3. Clinical effects of catechol-O-
volunteers when administered as single doses of 100 methyltransferase inhibition
or 200 mg (Keranen et al., 1993; Dingemanse et al.,
32.10.2.3.1. Effects in patients with motor fluctuations
1995a; Sedek et al., 1997) and PD patients (Myllyla
et al., 1993; Roberts et al., 1993; Nutt et al., 1994; Several prospective, double-blind placebo-controlled
Tohgi et al., 1995; Ruottinen and Rinne, 1996b). Data clinical trials assessed the effects of the COMT inhibitors
obtained in healthy volunteers after administration of tolcapone and entacapone as add-on to levodopa on
200 mg entacapone or tolcapone revealed increases of motor fluctuations (entacapone: Parkinson Study Group,
AUC by 40% or 80%, suggesting better clinical efficacy 1997; Rinne et al., 1998a; Myllyla et al., 2001; Poewe
for tolcapone (Keranen et al., 1993; Jorga et al., 1997; et al., 2002a; Brooks et al., 2003; tolcapone: Baas
Sedek et al., 1997). Single doses of entacapone and et al., 1997; Kurth et al., 1997; Myllyla et al., 1997;
tolcapone prolong levodopa half-life in PD patients by Rajput et al., 1997; Adler et al., 1998). Primary efficacy
2080% (Myllyla et al., 1993; Roberts et al., 1993; Nutt measures were daily on and off time. Table 32.6
et al., 1994; Tohgi et al., 1995; Ruottinen and Rinne, reports the outcome for treatment with either 100 or
1996b; Sedek et al., 1997). 200 mg tolcapone or 200 mg entacapone. On time in
Moreover, repeated administration of COMT in- all studies was significantly increased by 515%. Com-
hibitors leads to smoothening of levodopa plasma patible with this, off time was reduced between 10
concentration (Rouru et al., 1999). After repeated admin- and 22%. Improvements were found to be more pro-
istration, the AUC of levodopa is increased (by 2550%) found in patients treated with tolcapone. All studies
and levodopa trough levels increase (Keranen et al., report that levodopa doses could be decreased by
1993; Nutt et al., 1994; Dingemanse et al., 1995b; Jorga 1229%, again with higher dose reduction in tolcapone-
et al., 1997). Increases of AUC appear to be mildly smal- treated patients. These differences are compatible with
ler and elimination half-life shorter after administration the pharmacological properties of both agents. However,
of 200 mg entacapone compared to 200 mg tolcapone based on the differences in observational periods, with
(Kaakkola, 2000). Long-term treatment of 8 weeks by longer study durations for entacapone, it cannot be
600 mg/day of tolcapone and 1200 mg/day of entacapone excluded that higher efficacy measures for tolcapone at
correspondingly increased the AUC by 33% and 43%, least partially result from a higher impact of placebo
respectively (Nutt et al., 1994; Yamamoto et al., 1997; effects early during the trial and loss of efficacy in the
Kaakkola, 2000). Even though Cmax is not increased after entacapone-treated patients who were observed longer
single-dose administration, repeated doses will increase (Nutt, 2000).
Cmax (Nutt et al., 1994), which may be explained by the
fact that levodopa plasma concentrations at the end of 32.10.2.3.2. Catechol-O-methyltransferase inhibition
each dose interval are higher with COMT inhibition to prevent dyskinesias
(Nutt, 2000). Apart from its role as levodopa adjunct in the treatment of
When entacapone is administered with standard levo- motor fluctuations, COMT inhibitors have been impli-
dopa or CR levodopa, the AUC increased for both formu- cated in the delay of the development of dyskinesias.
lations (Ahtila et al., 1995; Piccini et al., 2000). Again, Long-term treatment with levodopa has been shown
when co-administered with CR levodopa, the efficacy of to be associated with motor complications, including
a single dose of 200 mg tolcapone was higher, with an wearing off and dyskinesias. Although the pathophy-
increase of AUC by 70% compared to a 20% increase after siological basis of these complications is incompletely
200 mg entacapone (Ahtila et al., 1995; Jorga et al., understood, current pathophysiological and clinical
1998a). evidence suggests that the half-life of dopaminergic
Increased central dopamine availability by COMT inhi- agents substantially contributes to the development of
bition is reflected in studies using fluoro-dopa (FD)-PET. dyskineseas (see section 32.7.4). The administration of
Both entacapone and tolcapone plus fluorodopa in con- levodopa with its short pharmacological half-life of about
junction with an AADC inhibitor increased striatal fluoro- 1.5 hours and pulsatile receptor stimulation is therefore
dopa uptake significantly (Sawle et al., 1994; Routtinen thought to be a major risk factor for the early develop-
et al. 1995; Ceravolo et al., 2002). This is compatible with ment of dyskinesias (Smith et al., 2003). Based on these
an enhancement of striatal dopamine, as shown by considerations it has been postulated that more conti-
microdialysis in rats (for entacapone, see Kaakkola and nous dopaminergic stimulation by an extension of the
56 T. D. HALBIG AND W. C. KOLLER
Table 32.6
Effects of catechol-O-methyltransferase (COMT) inhibitors on on and off time in fluctuating patients

Number of
patients on Study
COMT duration Change in Change in Change in daily
Study inhibitor (months) on time* off time* levodopa dose

Entacapone
(200 mg with each LD dose)
Parkinson Study Group (1997) 103 6 1.0 h NR 100 mg
Rinne et al. (1998) 85 6 1.4 h 1.3 h 87 mg
Brooks et al. (2003) 115 6 1.3 h 1.1 h 33 mg
Poewe et al. (2002) 172 6 1.7 h 1.6 h 54 mg
Tolcapone
Adler et al. (1998) 69/74 1.5 2.1 h/2.3 h 2h/2.5h 186 mg/252 mg
(100 mg/200 mg)
Rajput et al. (1997) 69/67 3 NR 2.3h/3.2h 166 mg/207 mg
(100 mg/200 mg)
Baas et al. (1997) 60/59 3 2.6 h/2.6 h 3.0h/2.4h 108 mg/122 mg
(100 mg/200 mg)
Kurth et al. (1997) 41/40 1.5 0.9 h/0.3 h 1.7h/1.3h 139 mg/200 mg
(50 mg/200 mg)

*Change in time as a percent of waking day.


NR, not reported.

levodopa half-life by co-administration of COMT inhi- both entacapone and tolcapone are primarily associated
bition may delay or prevent the development of dyski- with dopaminergic overstimulation and may present as
nesias. Indeed, experimental evidence obtained in dyskinesia, nausea, vomiting and hallucinations. More-
MPTP-treated monkeys revealed that the co-adminstra- over, non-dopaminergic side-effects such as diarrhea,
tion of levodopa and entacapone resulted in significantly abdominal pain, constipation, urine discoloration and
less severe dyskinesias compared to levodopa-only treat- fatigue have been reported for both agents and liver
ment (Smith et al., 2005). Interestingly, and in support of toxicity for tolcapone only.
the hypothesis that pulsatile stimulation contributes to the
establishment of dyskinesias, this effect was only present 32.10.2.4.1. Dopaminergic side-effects
when the levodopa/entacapone were administered four Dopaminergic side-effects usually develop early during
times daily. When levodopa/entacapone were adminis- treatment. The most common dopaminergic side-
tered only twice daily, there was no difference in dyskine- effects are dyskinesias, which most often present in
sia rate compared to levodopa-only treatment. In another the form of peak-dose dyskinesias. Treatment with both
study by the same group, the combination of entacapone COMT inhibitors presented with significantly higher
with levodopa resulted in even more rapid appearance of incidence of dyskinesia compared to patients on pla-
dyskinesias (Smith et al., 2003) when administered twice cebo. Differences in percentage of patients reporting
daily. Currently, clinical trials in patients with early PD dyskinesias as adverse event varied between 7% and
test the notion that more frequent dosing and the co- 47% depending on the study. Predictors for the devel-
administration of entacapone may delay the development opment of dyskinesias are pre-existing dyskinesias
of dyskinesias. and higher doses of levodopa (Poewe et al., 2002a;
Reinikainen et al., 2002). Dyskinesias can usually be
32.10.2.4. Side-effects of catechol- managed successfully by reducing the amount of levo-
O-methyltransferase inhibitors dopa per dose, by increasing the dose intervals or both
The side-effect profile of entacapone and tolcapone is (Poewe et al., 2002a; Brooks et al., 2003).
in general favorable. Safety data reported here are Nausea has been reported to be the second most
derived from the mentioned controlled phase III enta- important dopaminergic side-effect. It also emerges
capone and tolcapone studies (Table 32.7). Compatible during the first days of treatment. Nausea developed
with their levodopa-enhancing effects, side-effects of in about 1323% of patients treated with entacapone
LEVODOPA 57
Table 32.7
Incidence of the more frequent side-effects in controlled clinical trials assessing the effects of tolcapone1 or entacapone2
against placebo

Tolcapone Tolcapone
Agent (100 mg t.i.d.) (200 mg t.i.d.) Placebo Entacapone Placebo

n 296 298 298 603 400


Side-effects
Dyskinesia 42 51 20 25 15
Nausea 30 35 18 14 8
Diarrhea 16 18 8 10 4
Dizziness 13 6 10 8 6
Hallucination 8 10 5 4 4
Vomiting 8 10 4 4 1
Constipation 6 8 5 6 4
Fatigue 7 3 6 6 4
Abdominal pain 5 6 3 8 4
Urine discoloration 2 7 1 10 0

Numbers indicate percentage of patients.


1
Valeant Pharmaceuticals International (2006).
2
Novartis Pharmaceuticals Corporation (2000).
Adapted from Kaakkola (2000) and modified and updated.

(Parkinson Study Group, 1997; Rinne et al., 1998a; diarrhea is a class effect of the COMT inhibitors, it is
Brooks, 2004) and in about 30% of patients treated with not clear why frequency and severity are clearly more
tolcapone (Baas et al., 1997; Kurth et al., 1997; Rajput pronounced for tolcapone. However, an association
et al., 1997; Waters et al., 1997; Brooks, 2004). Again, with levodopa/AADC inhibitor treatment seems unli-
adjustment of the overall levodopa dose usually allows kely, since diarrhea is also induced when tolcapone is
control of this side-effect (Brooks, 2004). administered without levodopa (Hauser et al., 1998).
Interestingly, the incidence of neuropsychiatric side- Another frequent side-effect emerging in 737% of
effects after introduction of COMT inhibitors is rather patients is discoloration of urine to dark yellow or red-
low. However, in an open-label extension study (Larsen dish brown, which is harmless but worrisome to
et al., 2003), 19 out of 132 patients (14.4%) of patients trea- patients who were not previously informed about it.
ted with entacapone developed hallucinations. There is A rare but more serious side-effect associated with
evidence that single patients, in particular elderly patients, tolcapone treatment is liver toxicity (Brooks, 2004). Tol-
may be more vulnerable to confusion and hallucinations capone-treated patients in phase III trials developed
when treated with entacapone (Henry and Wilson, 1998). clinically relevant increases of liver enzymes and 3%
of patients had to discontinue the study (Brooks, 2004).
32.10.2.4.2. Non-dopaminergic effects After tolcapone was approved by regulative authorities,
In contrast to dopaminergic side-effects, non-dopami- 4 out of 60 000 patients receiving tolcapone developed
nergic adverse events were reported to occur throughout severe liver dysfunction, which was fatal in 3 patients
the entire study period. The most important non-dopa- (Assal et al., 1998; Olanow, 2000). One of these 3
minergic side-effect is diarrhea. Diarrhea usually did patients died despite adequate liver function laboratory
not occur during the first days of treatment. Diarrhea monitoring. In consequence tolcapone was withdrawn
in clinical trials was more frequent in patients receiving from the European and Canadian markets. In the US,
tolcapone (1618%) than in patients treated with enta- the use of tolcapone was restricted to patients who
capone (820%). In tolcapone-treated patients, severe experience motor fluctuations refractory to other treat-
explosive diarrhea led to treatment discontinuation in ments and who show no clinical evidence of liver disease
810% of patients (Baas et al., 1997; Kurth et al., or two serum glutamic-pyruvic transaminase (SGPT/
1997; Rajput et al., 1997), whereas only 14% receiving ALT) or serum glutamic-oxaloacetic transaminase
entacapone discontinued treatment for diarrhea (Rinne (SGOT/AST) values greater than the upper limit of nor-
et al., 1998a; Poewe et al., 2002a; Brooks et al., 2003). mal. Furthermore, in patients treated with tolcapone,
The underlying pathophysiological mechanism leading serum SGPT/ALT and SGOT/AST levels should be
to diarrhea is unknown. Even though it appears as if determined at baseline and periodically (i.e. every 24
58 T. D. HALBIG AND W. C. KOLLER
weeks) for the first 6 months of therapy. After the first 6 diseases. However, since its approval by the FDA in
months, periodic monitoring is recommended at inter- 1970, several other pharmacological compounds
vals deemed clinically relevant. Tolcapone should be mimicking the effects of dopamine or enhancing levo-
discontinued if SGPT/ALT or SGOT/AST levels exceed dopa availability have been introduced. Compared to
two times the upper limit of normal or if clinical signs newer agents and, in particular, dopamine agonists,
and symptoms suggest the onset of hepatic dysfunction. levodopa is still the most efficacious drug, as revealed
Finally, patients who fail to show substantial clinical by consistently better symptom control measured by
benefit within 3 weeks of initiation of treatment should the UPDRS (Rascol et al., 2000; Parkinson Study
be withdrawn from tolcapone (Valeant Pharmaceuticals Group, 2004b). Compatible with this, even though
International, 2006). dopamine agonists allow symptoms to be satisfactorily
All five mentioned phase III entacapone trials reported controlled during the first years of disease, eventually
occasionally elevations of liver enzymes (Gordin et al., nearly all dopamine agonist-treated patients need levo-
2003). However, a clear association with entacapone treat- dopa supplementation. The particular role of levodopa
ment could not be established. Furthermore, these studies is further substantiated by its favorable side-effect
did not report instances of serious liver dysfunction. One profile (Table 32.2 and see Ch. 33).
report, however, describes 3 patients who received entaco- Apart from its symptomatic clinical effects and side-
pone and developed symptopmatic liver dysfunction effects, the impact of levodopa therapy on the
which improved after withdrawal (Fisher et al., 2002). disease process has been the subject of considerable
Interpretation of these cases is complicated by several fac- controversy and neuroprotective effects as well as
tors, including poor documentation and lack of rechal- toxic effects have been examined widely. However, to
lenge with tolcapone, multimorbidity of patients and date, there is no convincing evidence for toxic effects
polypharmacy. Thus, the diagnosis of entacapone-induced of levodopa in humans. Recent data even show some
liver dysfunction is probable, although not proven controversial evidence for mild neuroprotective effects
(Brooks, 2004). Apart from these cases, no further evi- of levodopa therapy (Parkinson Study Group, 2004a).
dence for liver toxicity has been presented and indeed However, after more than 30 years of experience
more than 300 000 patient-years of entacapone exposure with levodopa, the limitations and shortcomings of
were not reported to be associated with liver dysfunction. levodopa therapy also became evident. A wealth of
Presently, the liver toxicity seen in COMT inhibitor- data indicated that long-term levodopa treatment con-
treated patients may therefore not be interpreted as a tributes to the early development of motor complica-
COMT inhibitor class effect, but rather a problem asso- tions and in particular dyskinesias (see section 32.7.4
ciated with tolcapone. Compatible with this view, there and Tables 32.4 and 32.5). Today it is estimated that,
are important pharmacological differences between both after 5 years of levodopa treatment, between 20 and
agents. Tolcapone has a much higher lipid solubility, 50% of patients have developed motor complications,
which may enhance penetration in mitochondrial inner including dyskinesias and onoff fluctuations, whereas
membrane (Nissinen et al., 1997). Furthermore, in contrast the incidence of motor complications in dopamine ago-
to entacapone, tolcapone may uncouple oxidative phos- nist-treated patients is significantly lower (510%).
phorylation. On the other hand, entacopone is nearly These observations led to a reassessment of the role of
entirely glucuronized (Wikberg et al., 1993a; Lautala levodopa in symptomatic PD therapy and in particular
et al., 2000), whereas talcapone is glucuronized but also the debate of when and how to initiate dopaminergic
methylated and oxidized (Jorga et al., 1998c). Both factors therapy.
may be important for the putative toxicity of tolcapone. Based on the potential of levodopa to induce long-
In summary, due to its safety profile, entacapone is term motor complications and the lack of evidence for
clearly the first-line COMT inhibitor. However, due to neuroprotection, there is presently no rationale for
its superior efficacy in the control of motor complica- treating patients with early PD without functional
tions, tolcapone may be considered for selected patients deficits relevant to activities of daily living and quality
in whom entacapone or other therapeutic measures do of life with levodopa. Instead, based on preliminary
not allow sufficient symptom control, provided the clinical evidence for their mild neuroprotective poten-
required safety monitoring is ensured (Factor et al., tial, MAO-B inhibitors (Ch. 34) or coenzyme Q10
2001; Onofrj et al., 2001). (Ch. 31) may be tried. In patients with minor symptoms
and a need for treatment, less potent non-dopaminergic
32.11. Conclusion and practical implications agents such as MAO-B inhibitors or amantadine
for the use of levodopa (Ch. 36) can be tried.
In patients with more pronounced symptoms needing
Levodopa revolutionized the symptomatic treatment of symptomatic improvement, dopaminergic agents have
one of the most prevalent progressive degenerative to be employed. Based on the potential of levodopa to
LEVODOPA 59
induce motor complications early, current treatment levodopa and a COMT inhibitor may be able to pre-
algorithms often recommend that the use of levodopa vent or delay the development of motor complications.
be spared until necessary to control motor symptoms Thus, the development of levodopa therapy in PD as
satisfactorily (Olanow et al., 2001). Instead, dopamine one of the few true success stories in the treatment of
agonists are recommended as initial treatment. How- a neurodegenerative has by no means come to an end.
ever, the question of which is the best strategy to initiate
treatment is controversial. There is general agreement References
that motor complications compromise patients func- Aarsland D, Karlsen K (1999). Neuropsychiatric aspects of
tionality and well-being. Nevertheless, in the several Parkinsons disease. Curr Psychiatry Rep 1: 6168.
controlled studies which compare dopamine agonists Abate G, Polimeni RM, Cuccurullo F et al. (1979). Effects of
and levodopa and which demonstrate superiority of indomethacin on postural hypotension in Parkinsonism. Br
dopamine agonists in terms of delay of motor complica- Med J 2: 14661468.
tions, no difference was found on quality of life and Acquas E, Carboni E, de Ree RH et al. (1992). Extracellular
activity of daily living measures (Rinne et al., 1998b; concentrations of dopamine and metabolites in the rat
caudate after oral administration of a novel catechol-O-
Rascol et al., 2000; Parkinson Study Group, 2004b).
methyltransferase inhibitor Ro 407592. J Neurochem
Moreover, the use of dopamine agonists may be limited 59: 326330.
due to their side-effect profile, in particular in vulner- Adler CH, Singer C, OBrien C et al. (1998). Randomized,
able patients where levodopa, as the generally best tol- placebo-controlled study of tolcapone in patients with
erated dopaminergic drug, should be considered. fluctuating Parkinson disease treated with levodopa-
Furthermore, dopamine agonists also have the potential carbidopa. Tolcapone Fluctuator Study Group III. Arch
to induce motor complications, although precise data on Neurol 55: 10891095.
the temporal profile and exact incidence are as yet Agid Y, Bonnet AM, Ruberg M et al. (1985). Pathophysiol-
unknown (Jenner, 2003). Further, there is some evi- ogy of L-dopa-induced abnormal involuntary movements.
dence that establishment of motor complications is Psychopharmacology Suppl 2: 145159.
reversible (Jenner, 2002). No controlled study ever Agid Y, Javoy-Agid F, Ruberg M (1987). Biochemistry of
neurotransmitters in Parkinsons disease. In: CD Marsden,
assessed whether patients who were initially treated
S Fahn (Eds.), Movement Disorders 2. Butterworths,
with a dopamine agonist and who received levodopa London, pp. 166230.
later on are doing better in terms of motor functions, Ahlskog JE, Muenter MD (2001). Frequency of levodopa-
activities of daily living and quality of life than patients related dyskinesias and motor fluctuations as estimated
who were initially treated with levodopa and developed from the cumulative literature. Mov Disord 16: 448458.
motor complications early in the disease and then Ahlskog JE, Muenter MD, McManis PG et al. (1988). Con-
received a dopamine agonist as add-on in order to con- trolled-release Sinemet (CR-4): a double-blind crossover
trol the motor complications via more CDS. Based on study in patients with fluctuating Parkinsons disease.
these considerations, the more favorable side-effect Mayo Clin Proc 63: 876886.
profile and its higher efficacy, levodopa administration Ahlskog JE, Uitti RJ, OConnor MK et al. (1999). The effect
may be also considered a rational choice for individual of dopamine agonist therapy on dopamine transporter ima-
ging in Parkinsons disease. Mov Disord 14: 940946.
patients, even in earlier stages of disease progression.
Ahtila S, Kaakkola S, Gordin A et al. (1995). Effect of enta-
This holds in particular for older de novo patients with capone, a COMT inhibitor, on the pharmacokinetics
less risk for the development of motor complications and metabolism of levodopa after administration of
than patients with early onset of PD. controlled-release levodopa-carbidopa in volunteers. Clin
In conclusion, although it is now more than 30 years Neuropharmacol 18: 4657.
after the introduction of levodopa, pharmacological PD Allain P, Le Bouil A, Cordillet E et al. (1995). Sulfate and
therapy in the beginning of the 21st century involves in cysteine levels in the plasma of patients with Parkinsons
most patients who have access to the full armamentar- disease. Neurotoxicology 16: 527529.
ium of PD therapy a complex, growing array of agents APA (2000). Diagnostic and Statistical Manual of Mental
that is usually continuously modified in the course Disorders, DSM-IV-TR Fourth Edn. American Psychiatric
of disease progression. For most patients levodopa Association, Washington DC.
Arnulf I, Bonnet AM, Damier P et al. (2000). Hallucinations,
sooner or later is indispensable for satisfactory symp-
REM sleep, and Parkinsons disease: a medical hypothesis.
tom control. Neurology 55: 281288.
Finally, apart from new modes of administration, Assal F, Spahr L, Hadengue A et al. (1998). Tolcapone and
recent conceptual developments may again lead to fulminant hepatitis. Lancet 352: 958.
changes in current therapeutic thinking. For example, Avanzi M, Uber E, Bonfa F (2004). Pathological gambling in
based on the concept of CDS, clinical trials currently two patients on dopamine replacement therapy for Parkin-
evaluate whether the combined administration of sons disease. Neurol Sci 25: 98101.
60 T. D. HALBIG AND W. C. KOLLER

Baas H, Beiske AG, Ghika J et al. (1997). Catechol-O- Bouhaddi M, Vuillier F, Fortrat JO et al. (2004). Impaired
methyltransferase inhibition with tolcapone reduces the cardiovascular autonomic control in newly and long-
wearing off phenomenon and levodopa requirements in term-treated patients with Parkinsons disease: involve-
fluctuating parkinsonian patients. J Neurol Neurosurg ment of L-dopa therapy. Auton Neurosci 116: 3038.
Psychiatry 63: 421428. Bradbrook ID, Gillies HC, Morrison PJ et al. (1986). The
Barbeau A, Sourkes T, Murphy G (1962). Les Catechola- effects of domperidone on the absorption of levodopa in
mines dans la Maladie de Parkinson Georg, Geneva. normal subjects. Eur J Clin Pharmacol 29: 721723.
Barnes J, David AS (2001). Visual hallucinations in Parkin- Brandstadter D, Oertel WH (2002). Treatment of drug-
sons disease: a review and phenomenological survey. induced psychosis with quetiapine and clozapine in Par-
J Neurol Neurosurg Psychiatry 70: 727733. kinsons disease. Neurology 58: 160161.
Barone JA (1999). Domperidone: a peripherally acting dopa- Bravi D, Mouradian MM, Roberts JW et al. (1994). Wearing-
mine2-receptor antagonist. Ann Pharmacother 33: 429440. off fluctuations in Parkinsons disease: contribution of
Bartholini G, Burkard WP, Pletscher A (1967). Increase of postsynaptic mechanisms. Ann Neurol 36: 2731.
cerebral catecholamines caused by 3,4-dihydroxyphenyla- Breier A, Sutton VK, Feldman PD et al. (2002). Olanzapine in
lanine after inhibition of peripheral decarboxylase. Nature the treatment of dopamimetic-induced psychosis in patients
215: 852853. with Parkinsons disease. Biol Psychiatry 52: 438445.
Bayer AJ, Day JJ, Finucane P et al. (1988). Bioavailability Brevetti G, Bonaduce D, Breglio R et al. (1990). Parkinsons
and acceptability of a dispersible formulation of levodopa- disease and hypotension: 24-hour blood pressure recording
benserazide in parkinsonian patients with and without in ambulant patients. Clin Cardiol 13: 474478.
dysphagia. J Clin Pharm Ther 13: 191194. Brooks DJ (2004). Safety and tolerability of COMT inhibi-
Bedard PJ, Di Paolo T, Falardeau P et al. (1986). Chronic tors. Neurology 62: S39S46.
treatment with L-DOPA, but not bromocriptine induces Brooks DJ, Sagar H (2003). UK-Irish Entacapone Study
dyskinesia in MPTP-parkinsonian monkeys. Correlation Group. Entacapone is beneficial in both fluctuating and
with [3H]spiperone binding. Brain Res 379: 294299. non-fluctuating patients with Parkinsons disease: a rando-
Ben-Shlomo Y, Marmot MG (1995). Survival and cause of mised, placebo controlled, double blind, six month study.
death in a cohort of patients with parkinsonism: possible clues J Neurol Neurosurg Psychiatry 74: 10711079.
to aetiology? J Neurol Neurosurg Psychiatry 58: 293299. Burguera JA, Grandas F, Horga de la Parte JF et al. (1999).
Berger C, Mehrhoff FW, Beier KM et al. (2003). [Sexual [Entacapone: is it useful as complimentary treatment with
delinquency and Parkinsons disease.] Nervenarzt 74: levodopa?] Rev Neurol 28: 817834.
370375. Byrne KG, Pfeiffer R, Quigley EM (1994). Gastrointestinal
Birkmayer W (1969). The importance of monoamine meta- dysfunction in Parkinsons disease. A report of clinical
bolism for the pathology of the extrapyramidal system. experience at a single center. J Clin Gastroenterol 19:
J Neurovisc Relat 31 (Suppl 9): 297. 1116.
Birkmayer W, Hornykiewicz O (1961). [The L-3,4-dioxy- Calne DB (1970). L-Dopa in the treatment of parkinsonism.
phenylalanine (DOPA)-effect in Parkinson-akinesia.] Clin Pharmacol Ther 11: 789801.
Wien Klin Wochenschr 73: 787788. Calne DB, Brennan J, Spiers AS et al. (1970). Hypotension
Blanchet PJ, Calon F, Martel JC et al. (1995). Continuous caused by L-dopa. Br Med J 1: 474475.
administration decreases and pulsatile administration Calne DB, Reid JL, Vakil SD et al. (1971). Idiopathic Parkin-
increases behavioral sensitivity to a novel dopamine sonism treated with an extracerebral decarboxylase inhibitor
D2 agonist (U-91356A) in MPTP-exposed monkeys. in combination with levodopa. Br Med J 3: 729732.
J Pharmacol Exp Ther 272: 854859. Calon F, Goulet M, Blanchet PJ et al. (1995). Levodopa or
Block G, Liss C, Reines S et al. (1997). Comparison of D2 agonist induced dyskinesia in MPTP monkeys: corre-
immediate-release and controlled release carbidopa/levo- lation with changes in dopamine and GABAA receptors
dopa in Parkinsons disease. A multicenter 5-year study. in the striatopallidal complex. Brain Res 680: 4352.
The CR First Study Group. Eur Neurol 37: 2327. Calon F, Grondin R, Morissette M et al. (2000). Molecular
Borges N, Vieira-Coelho MA, Parada A et al. (1997). Studies basis of levodopa-induced dyskinesias. Ann Neurol 47:
on the tight-binding nature of tolcapone inhibition S70S78.
of soluble and membrane-bound rat brain catechol- Camerlingo M, Ferraro B, Gazzaniga GC et al. (1990). Car-
O-methyltransferase. J Pharmacol Exp Ther 282: 812817. diovascular reflexes in Parkinsons disease: long-term
Bostom AG, Rosenberg IH, Silbershatz H et al. (1999a). effects of levodopa treatment on de novo patients. Acta
Nonfasting plasma total homocysteine levels and stroke Neurol Scand 81: 346348.
incidence in elderly persons: the Framingham Study. Cedarbaum JM, Kutt H, Dhar AK et al. (1986). Effect of
Ann Intern Med 131: 352355. supplemental carbidopa on bioavailability of L-dopa. Clin
Bostom AG, Silbershatz H, Rosenberg IH et al. (1999b). Neuropharmacol 9: 153159.
Nonfasting plasma total homocysteine levels and Cedarbaum JM, Breck L, Kutt H et al. (1987). Controlled-
all-cause and cardiovascular disease mortality in elderly release levodopa/carbidopa. II. Sinemet CR4 treatment of
Framingham men and women. Arch Intern Med 159: response fluctuations in Parkinsons disease. Neurology
10771080. 37: 16071612.
LEVODOPA 61

Cedarbaum JM, Silvestri M, Kutt H (1990a). Sustained ent- de Maindreville AD, Fenelon G, Mahieux F (2005). Halluci-
eral administration of levodopa increases and interrupted nations in Parkinsons disease: a follow-up study. Mov
infusion decreases levodopa dose requirements. Neurology Disord 20: 212217.
40: 995997. De Michele G, Mengano A, Filla A et al. (1989). A double-
Cedarbaum JM, Silvestri M, Clark M et al. (1990b). Results blind, cross-over trial with madopar HBS in patients with
of long-term treatment with controlled-release levodopa/ Parkinsons disease. Acta Neurol (Napoli) 11: 408414.
carbidopa (Sinemet CR). J Neural Transm Park Dis de Smet Y, Ruberg M, Serdaru M et al. (1982). Confusion,
Dement Sect 2: 205213. dementia and anticholinergics in Parkinsons disease. J
Ceravolo R, Piccini P, Bailey DL et al. (2002). 18F-dopa PET Neurol Neurosurg Psychiatry 45: 11611164.
evidence that tolcapone acts as a central COMT inhibitor Deleu D, Jacques M, Michotte Y et al. (1989). Clinical and
in Parkinsons disease. Synapse 43: 201207. pharmacokinetic evaluation of controlled-release levodopa/
Chapuis S, Ouchchane L, Metz O et al. (2004). Impact of the carbidopa (CR-4) in parkinsonian patients with severe
motor complications of Parkinsons disease on the quality motor fluctuations: a six month follow-up study. Clin
of life. Mov Disord 20: 224230. Neurol Neurosurg 91: 303309.
Chouza C, Aljanati R, Caamano JL et al. (1990). Long-term DeLong MR, Crutcher MD, Georgopoulos AP (1983). Rela-
treatment with Madopar HBS in parkinsonians with tions between movement and single cell discharge in the
fluctuations. Adv Neurol 53: 519526. substantia nigra of the behaving monkey. J Neurosci 3:
Clarke CE, Sambrook MA, Mitchell IJ et al. (1987). Levodo- 15991606.
pa-induced dyskinesia and response fluctuations in den Heijer T, Vermeer SE, Clarke R et al. (2003). Homocys-
primates rendered parkinsonian with 1-methyl-4-phenyl- teine and brain atrophy on MRI of non-demented elderly.
1,2,3,6- tetrahydropyridine (MPTP). J Neurol Sci 78: Brain 126: 170175.
273280. Descombes S, Bonnet AM, Gasser UE et al. (2001). Dual-
Comella CL, Goetz CG (1994). Akathisia in Parkinsons release formulation, a novel principle in L-dopa treatment
disease. Mov Disord 9: 545549. of Parkinsons disease. Neurology 56: 12391242.
Contin M, Riva R, Martinelli P et al. (1999). Concentration- Dewey RB Jr, OSuilleabhain PE (2000). Treatment of drug-
effect relationship of levodopa-benserazide dispersible induced psychosis with quetiapine and clozapine in
formulation versus standard form in the treatment of Parkinsons disease. Neurology 55: 17531754.
complicated motor response fluctuations in Parkinsons Dexter DT, Wells FR, Agid F et al. (1987). Increased nigral
disease. Clin Neuropharmacol 22: 351355. iron content in postmortem parkinsonian brain. Lancet 2:
Cook JW, Taylor LM, Orloff SL et al. (2002). Homocysteine 12191220.
and arterial disease. Experimental mechanisms. Vascul Dexter DT, Wells FR, Lees AJ et al. (1989). Increased nigral
Pharmacol 38: 293300. iron content and alterations in other metal ions occurring
Corona-Morales AA, Castell A, Zhang L (2000). L-DOPA- in brain in Parkinsons disease. J Neurochem 52:
induced neurotoxic and apoptotic changes on cultured 18301836.
chromaffin cells. Neuroreport 11: 503506. Dingemanse J, Jorga K, Zurcher G et al. (1995a). Pharmacoki-
Cotzias GC, Van Woert MH, Schiffer LM (1967). Aromatic netic-pharmacodynamic interaction between the COMT
amino acids and modification of parkinsonism. N Engl inhibitor tolcapone and single-dose levodopa. Br J Clin
J Med 276: 374379. Pharmacol 40: 253262.
Cotzias GC, Papavasiliou PS, Gellene R (1969). Modification Dingemanse J, Jorga KM, Schmitt M et al. (1995b). Integrated
of Parkinsonismchronic treatment with L-dopa. N Engl pharmacokinetics and pharmacodynamics of the novel
J Med 280: 337345. catechol-O-methyltransferase inhibitor tolcapone during
Crevoisier C, Hoevels B, Zurcher G et al. (1987). Bioavail- first administration to humans. Clin Pharmacol Ther 57:
ability of L-dopa after Madopar HBS administration in 508517.
healthy volunteers. Eur Neurol 27 (Suppl 1): 3646. Dingemanse J, Jorga K, Zurcher G et al. (1996). Multiple-dose
Da Prada M, Kettler R, Zurcher G et al. (1987). Inhibition of clinical pharmacology of the catechol-O-methyl-transferase
decarboxylase and levels of dopa and 3-O-methyldopa: a inhibitor tolcapone in elderly subjects. Eur J Clin Pharmacol
comparative study of benserazide versus carbidopa in 50: 4755.
rodents and of Madopar standard versus Madopar HBS Djaldetti R, Baron J, Ziv I et al. (1996). Gastric emptying in
in volunteers. Eur Neurol 27 (Suppl 1): 920. Parkinsons disease: patients with and without response
Datla KP, Blunt SB, Dexter DT (2001). Chronic L-DOPA fluctuations. Neurology 46: 10511054.
administration is not toxic to the remaining dopaminergic Djaldetti R, Inzelberg R, Giladi N et al. (2002). Oral solution
nigrostriatal neurons, but instead may promote their func- of levodopa ethylester for treatment of response fluctua-
tional recovery, in rats with partial 6-OHDA or FeCl(3) tions in patients with advanced Parkinsons disease. Mov
nigrostriatal lesions. Mov Disord 16: 424434. Disord 17: 297302.
de la Fuente-Fernandez R, Nunez MA, Lopez E (1999). The Djaldetti R, Giladi N, Hassin-Baer S et al. (2003). Pharmacoki-
apolipoprotein E epsilon 4 allele increases the risk of netics of etilevodopa compared to levodopa in patients with
drug-induced hallucinations in Parkinsons disease. Clin Parkinsons disease: an open-label, randomized, crossover
Neuropharmacol 22: 226230. study. Clin Neuropharmacol 26: 322326.
62 T. D. HALBIG AND W. C. KOLLER

Dodd ML, Klos KJ, Bower JH et al. (2005). Pathological Disease, Vol. 2. Macmillan Health Care Information,
gambling caused by drugs used to treat Parkinson disease. Florham Park, NJ, pp. 153163, 293304.
Arch Neurol 62: 13771381. Fehling C (1966). Treatment of Parkinsons syndrome with L-
Driver-Dunckley E, Samanta J, Stacy M (2003). Pathological dopa. A double blind study. Acta Neurol Scand 42: 367372.
gambling associated with dopamine agonist therapy in Feldman RG, Mosbach PA, Kelly MR et al. (1989). Double-
Parkinsons disease. Neurology 61: 422423. blind comparison of standard Sinemet and Sinemet CR in
Duan W, Ladenheim B, Cutler RG et al. (2002). Dietary patients with mild-to-moderate Parkinsons disease.
folate deficiency and elevated homocysteine levels endan- Neurology 39: 96101; discussion 105.
ger dopaminergic neurons in models of Parkinsons Fenelon G, Mahieux F, Huon R et al. (2000). Hallucinations
disease. J Neurochem 80: 101110. in Parkinsons disease: prevalence, phenomenology and
Dufouil C, Alperovitch A, Ducros V et al. (2003). Homocys- risk factors. Brain 123 (Pt 4): 733745.
teine, white matter hyperintensities, and cognition in Fernandez HH, Friedman JH (1999). Punding on L-dopa.
healthy elderly people. Ann Neurol 53: 214221. Mov Disord 14: 836838.
Dupont E, Andersen A, Boas J et al. (1996). Sustained- Fernandez HH, Friedman JH, Jacques C et al. (1999). Quetia-
release Madopar HBS compared with standard Madopar pine for the treatment of drug-induced psychosis in Par-
in the long-term treatment of de novo parkinsonian kinsons disease. Mov Disord 14: 484487.
patients. Acta Neurol Scand 93: 1420. Fernandez HH, Trieschmann ME, Friedman JH (2004). Aripi-
Duty S, Brotchie JM (1997). Enhancement of the behavioral prazole for drug-induced psychosis in Parkinson disease:
response to apomorphine administration following preliminary experience. Clin Neuropharmacol 27: 45.
repeated treatment in the 6-hydroxydopamine-lesioned Fiala KH, Whetteckey J, Manyam BV (2003). Malignant
rat is temporally correlated with a rise in striatal preproen- melanoma and levodopa in Parkinsons disease: causality
kephalin-B, but not preproenkephalin-A, gene expression. or coincidence? Parkinsonism Relat Disord 9: 321327.
Exp Neurol 144: 423432. Fiorillo CD, Tobler PN, Schultz W (2003). Discrete coding
Duvoisin RC (1972). Diphenidol for levodopa induced of reward probability and uncertainty by dopamine
nausea and vomiting. JAMA 221: 1408. neurons. Science 299: 18981902.
Edwards LL, Quigley EM, Pfeiffer RF (1992). Gastrointest- Fisher A, Croft-Baker J, Davis M et al. (2002). Entacapone-
inal dysfunction in Parkinsons disease: frequency and induced hepatotoxicity and hepatic dysfunction. Mov
pathophysiology. Neurology 42: 726732. Disord 17: 13621365; discussion 13971400.
Ehringer H, Hornykiewicz O (1960). [Distribution of nora- French Clozapine Parkinson Study Group (1999). Clozapine
drenaline and dopamine (3-hydroxytyramine) in the in drug-induced psychosis in Parkinsons disease. The
human brain and their behavior in diseases of the extrapyr- French Clozapine Parkinson Study Group. Lancet 353:
amidal system]. Klin Wochenschr 38: 12361239. 20412042.
Eisler T, Eng N, Plotkin C et al. (1981). Absorption of levo- Friedman JH (1991). The management of the levodopa psy-
dopa after rectal administration. Neurology 31: 215217. choses. Clin Neuropharmacol 14: 283295.
Emerit J, Edeas M, Bricaire F (2004). Neurodegenerative dis- Friedman JH, Lannon MC (1989a). Clozapine in the treat-
eases and oxidative stress. Biomed Pharmacother 58: 3946. ment of psychosis in Parkinsons disease. Neurology 39:
Emre M (2003). Dementia associated with Parkinsons 12191221.
disease. Lancet Neurol 2: 229237. Friedman JH, Lannon MC (1989b). An open trial of con-
Erni W, Held K (1987). The hydrodynamically balanced trolled release carbidopa/L-dopa (sinemet CR) for the
system: a novel principle of controlled drug release. Eur treatment of mild-to-moderate Parkinsons disease. Clin
Neurol 27 (Suppl 1): 2127. Neuropharmacol 12: 220223.
Evans AH, Katzenschlager R, Paviour D et al. (2004). Pund- Gancher ST, Nutt JG, Woodward W (1988). Response to
ing in Parkinsons disease: its relation to the dopamine brief levodopa infusions in parkinsonian patients with
dysregulation syndrome. Mov Disord 19: 397405. and without motor fluctuations. Neurology 38: 712716.
Fabbrini G, Mouradian MM, Juncos JL et al. (1988). Motor Garcia Ruiz PJ, Meseguer E, Del Val J et al. (2004). Motor
fluctuations in Parkinsons disease: central pathophysiolo- complications in Parkinson disease: a prospective follow-
gical mechanisms, Part I. Ann Neurol 24: 366371. up study. Clin Neuropharmacol 27: 4952.
Factor SA, Molho ES, Feustel PJ et al. (2001). Long-term Genedani S, Rasio G, Cortelli P et al. (2004). Studies on
comparative experience with tolcapone and entacapone homocysteine and dehydroepiandrosterone sulphate
in advanced Parkinsons disease. Clin Neuropharmacol plasma levels in Alzheimers disease patients and in Par-
24: 295299. kinsons disease patients. Neurotox Res 6: 327332.
Fahn S (1974). On-off phenomenon with levodopa therapy in Gerfen CR, Engber TM, Mahan LC et al. (1990). D1 and D2
Parkinsonism. Clinical and pharmacologic correlations and dopamine receptor-regulated gene expression of striatoni-
the effect of intramuscular pyridoxine. Neurology 24: gral and striatopallidal neurons. Science 250: 14291432.
431441. Giovannoni G, OSullivan JD, Turner K et al. (2000).
Fahn S, Elton R, Members of the UPDRS Development Hedonistic homeostatic dysregulation in patients with
Committee (1987). In: S Fahn, CD Marsden, DB Calne, Parkinsons disease on dopamine replacement therapies.
M Goldstein (Eds.), Recent Developments in Parkinsons J Neurol Neurosurg Psychiatry 68: 423428.
LEVODOPA 63

Giros B, Caron MG (1993). Molecular characterization of the Haglund L, Hoogkamer JFW (1995). MadoparW dispensible:
dopamine transporter. Trends Pharmacol Sci 14: 4349. pharmacokinetics and bioavailability in healthy volun-
Goetz CG, Stebbins GT (1993). Risk factors for nursing teers. J Neurol 242 (Suppl 2): S140.
home placement in advanced Parkinsons disease. Neurol- Hakamaki T, Rajala T, Lehtonen A (1998). Ambulatory
ogy 43: 22272229. 24-hour blood pressure recordings in patients with Parkin-
Goetz CG, Tanner CM, Shannon KM et al. (1988). Controlled- sons disease with or without fludrocortisone. Int J Clin
release carbidopa/levodopa (CR4-Sinemet) in Parkinsons Pharmacol Ther 36: 367369.
disease patients with and without motor fluctuations. Neurol- Han SK, Mytilineou C, Cohen G (1996). L-DOPA up-
ogy 38: 11431146. regulates glutathione and protects mesencephalic cultures
Goetz CG, Tanner CM, Gilley DW et al. (1989). Develop- against oxidative stress. J Neurochem 66: 501510.
ment and progression of motor fluctuations and side Hardie RJ, Malcolm SL, Lees AJ et al. (1986). The pharma-
effects in Parkinsons disease: comparison of Sinemet cokinetics of intravenous and oral levodopa in patients
CR versus carbidopa/levodopa. Neurology 39: 6366; dis- with Parkinsons disease who exhibit on-off fluctuations.
cussion 7263. Br J Clin Pharmacol 22: 429436.
Goetz CG, Pappert EJ, Blasucci LM et al. (1998). Intravenous Hardoff R, Sula M, Tamir A et al. (2001). Gastric emptying
levodopa in hallucinating Parkinsons disease patients: time and gastric motility in patients with Parkinsons dis-
high-dose challenge does not precipitate hallucinations. ease. Mov Disord 16: 10411047.
Neurology 50: 515517. Hauser RA, Molho E, Shale H et al. (1998). A pilot evalua-
Goetze O, Wieczorek J, Mueller T et al. (2005). Impaired tion of the tolerability, safety, and efficacy of tolcapone
gastric emptying of a solid test meal in patients with Par- alone and in combination with oral selegiline in untreated
kinsons disease using (13)C-sodium octanoate breath test. Parkinsons disease patients. Tolcapone De Novo Study
Neurosci Lett 375: 170173. Group. Mov Disord 13: 643647.
Golbe LI (1991). Young-onset Parkinsons disease: a clinical Heikkinen H, Saraheimo M, Antila S et al. (2001). Phar-
review. Neurology 41: 168173. macokinetics of entacapone, a peripherally acting cate-
Goldberg LI, Rajfer SI (1985). Dopamine receptors: applica- chol-O-methyltransferase inhibitor, in man. A study
tions in clinical cardiology. Circulation 72: 245248. using a stable isotope technique. Eur J Clin Pharmacol
Goldstein DS, Holmes CS, Dendi R et al. (2002). Orthostatic 56: 821826.
hypotension from sympathetic denervation in Parkinsons Hely MA, Morris JG, Reid WG et al. (1994). The Sydney
disease. Neurology 58: 12471255. multicentre study of Parkinsons disease: a randomised,
Gordin A, Kaakkola S, Teravainen H (2003). Position of prospective five year study comparing low dose bromo-
COMT inhibition in the treatment of Parkinsons disease. criptine with low dose levodopa-carbidopa. J Neurol
Adv Neurol 91: 237250. Neurosurg Psychiatry 57: 903910.
Gorell JM, Johnson CC, Rybicki BA (1994). Parkinsons dis- Henry C, Wilson JA (1998). Catechol-O-methyltransferase
ease and its comorbid disorders: an analysis of Michigan inhibitors in Parkinsons disease. Lancet 351: 19651966.
mortality data, 1970 to 1990. Neurology 44: 18651868. Hernan MA, Zhang SM, Rueda-deCastro AM et al. (2001).
Grace AA (1991). Phasic versus tonic dopamine release and the Cigarette smoking and the incidence of Parkinsons dis-
modulation of dopamine system responsivity: a hypothesis ease in two prospective studies. Ann Neurol 50: 780786.
for the etiology of schizophrenia. Neuroscience 41: 124. Hoehn M, Yahr M (1967). Parkinsonism: onset, progression
Graham DG, Tiffany SM, Bell WR Jr et al. (1978). Autoxida- and mortality. Neurology 17: 427442.
tion versus covalent binding of quinones as the mechanism Hoehn MM (1975). Levodopa-induced postural hypotension.
of toxicity of dopamine, 6-hydroxydopamine, and related Treatment with fludrocortisone. Arch Neurol 32: 5051.
compounds toward C1300 neuroblastoma cells in vitro. Hollander E, Evers M (2001). New developments in impul-
Mol Pharmacol 14: 644653. sivity. Lancet 358: 949950.
Grahnen A, Eckernas SA, Collin C et al. (1992). Comparative Holroyd S, Currie L, Wooten GF (2001). Prospective study
multiple-dose pharmacokinetics of controlled-release of hallucinations and delusions in Parkinsons disease.
levodopa products. Eur Neurol 32: 343348. J Neurol Neurosurg Psychiatry 70: 734738.
Grandas F, Galiano ML, Tabernero C (1999). Risk factors Horne MK, Cheng CH, Wooten GF (1984). The cerebral
for levodopa-induced dyskinesias in Parkinsons disease. metabolism of L-dihydroxyphenylalanine. An autoradio-
J Neurol 246: 11271133. graphic and biochemical study. Pharmacology 28: 1226.
Greenacre JK, Coxon A, Petrie A et al. (1976). Comparison Hornykiewicz O (1974). The mechanisms of action of
of levodopa with carbidopa or benserazide in parkinson- L-dopa in Parkinsons disease. Life Sci 15: 12491259.
ism. Lancet 2: 381384. Hornykiewicz O (2002). L-DOPA: from a biologically inac-
Gschwandtner U, Aston J, Renaud S et al. (2001). Pathologic tive amino acid to a successful therapeutic agent. Amino
gambling in patients with Parkinsons disease. Clin Neuro- Acids 23: 6570.
pharmacol 24: 170172. Howse PM, Matthews WB (1973). Brocresine in Parkinsons
Guldberg HC, Marsden CA (1975). Catechol-O-methyl disease. Action of a peripheral and central decarboxylase
transferase: pharmacological aspects and physiological inhibitor in potentiating levodopa. J Neurol Neurosurg
role. Pharmacol Rev 27: 135206. Psychiatry 36: 2729.
64 T. D. HALBIG AND W. C. KOLLER

Huang YC, Colaizzi JL, Bierman RH et al. (1986). Phar- Jorga KM, Kroodsma JM, Fotteler B et al. (1998c). Effect
macokinetics and dose proportionality of domperidone in of liver impairment on the pharmacokinetics of tolca-
healthy volunteers. J Clin Pharmacol 26: 628632. pone and its metabolites. Clin Pharmacol Ther 63:
Hutton JT, Morris JL (1991). Long-term evaluation of Sine- 646654.
met CR in parkinsonian patients with motor fluctuations. Jorga KM, Larsen JP, Beiske A et al. (1999). The effect of
Can J Neurol Sci 18: 467471. tolcapone on the pharmacokinetics of benserazide. Eur J
Hutton JT, Morris JL, Bush DF et al. (1989). Multicenter Neurol 6: 211219.
controlled study of Sinemet CR vs Sinemet (25/100) in Juncos JL, Mouradian MM, Fabbrini G et al. (1987). Levodopa
advanced Parkinsons disease. Neurology 39: 6772; methyl ester treatment of Parkinsons disease. Neurology
discussion 7263. 37: 12421245.
Inzelberg R, Kipervasser S, Korczyn AD (1998). Auditory Kaakkola S (2000). Clinical pharmacology, therapeutic use
hallucinations in Parkinsons disease. J Neurol Neurosurg and potential of COMT inhibitors in Parkinsons disease.
Psychiatry 64: 533535. Drugs 59: 12331250.
Iwase H, Sudo J, Terui J et al. (2000). Transdermal absorp- Kaakkola S, Wurtman RJ (1993). Effects of catechol-O-
tion of L-dopa from a new system composed of two sepa- methyltransferase inhibitors and L-3,4-dihydroxyphenyla-
rate layers of L-dopa and hydrogel in rats. Drug Dev Ind lanine with or without carbidopa on extracellular
Pharm 26: 755759. dopamine in rat striatum. J Neurochem 60: 137144.
Jankovic J, Schwartz K, Vander Linden C (1989). Compari- Kafka MP (2003). The monoamine hypothesis for the patho-
son of Sinemet CR4 and standard Sinemet: double blind physiology of paraphilic disorders: an update. Ann N Y
and long-term open trial in parkinsonian patients with Acad Sci 989: 8694; discussion 144153.
fluctuations. Mov Disord 4: 303309. Kahn N, Freeman A, Juncos JL et al. (1991). Clozapine is
Jankovic J, Gilden JL, Hiner BC et al. (1993). Neurogenic beneficial for psychosis in Parkinsons disease. Neurology
orthostatic hypotension: a double-blind, placebo-controlled 41: 16991700.
study with midodrine. Am J Med 95: 3848. Kalir HH, Mytilineou C (1991). Ascorbic acid in mesence-
Jellinger KA (2003). Prevalence of stroke in Parkinsons phalic cultures: effects on dopaminergic neuron develop-
disease. Mov Disord 18: 723724. ment. J Neurochem 57: 458464.
Jenner P (2002). Pharmacology of dopamine agonists in the Kaneoke Y, Koike Y, Sakurai N et al. (1995). Gastrointest-
treatment of Parkinsons disease. Neurology 58: S1S8. inal dysfunction in Parkinsons disease detected by
Jenner P (2003). The MPTP-treated primate as a model of electrogastroenterography. J Auton Nerv Syst 50:
motor complications in PD: primate model of motor 275281.
complications. Neurology 61: S4S11. Kao HD, Traboulsi A, Itoh S et al. (2000). Enhancement of
Jensen NO, Dupont E, Hansen E et al. (1987). Madopar the systemic and CNS specific delivery of L-dopa by the
HBS: slow-release levodopa and benserazide in parkinso- nasal administration of its water soluble prodrugs. Pharm
nian patients presenting marked fluctuations in symptoms Res 17: 978984.
on standard L-dopa treatment. Eur Neurol 27 (Suppl 1): Kaufmann H, Nahm K, Purohit D et al. (2004). Autonomic
6872. failure as the initial presentation of Parkinson disease and
Jolkkonen J, Jenner P, Marsden CD (1995). L-DOPA dementia with Lewy bodies. Neurology 63: 10931095.
reverses altered gene expression of substance P but not Keranen T, Gordin A, Harjola VP et al. (1993). The effect of
enkephalin in the caudate-putamen of common marmosets catechol-O-methyl transferase inhibition by entacapone on
treated with MPTP. Brain Res Mol Brain Res 32: the pharmacokinetics and metabolism of levodopa in
297307. healthy volunteers. Clin Neuropharmacol 16: 145156.
Jorga K, Fotteler B, Schmitt M et al. (1997). The effect of Keranen T, Gordin A, Karlsson M et al. (1994). Inhibition of
COMT inhibition by tolcapone on tolerability and pharma- soluble catechol-O-methyltransferase and single-dose
cokinetics of different levodopa/benserazide formulations. pharmacokinetics after oral and intravenous administration
Eur Neurol 38: 5967. of entacapone. Eur J Clin Pharmacol 46: 151157.
Jorga K, Fotteler B, Sedek G et al. (1998a). The effect of tol- Kincannon J, Boutzale C (1999). The physiology of pigmented
capone on levodopa pharmacokinetics is independent of nevi. Pediatrics 104: 10421045.
levodopa/carbidopa formulation. J Neurol 245: 223230. Kleedorfer B, Poewe W (1992). Comparative efficacy of two
Jorga KM (1998a). Pharmacokinetics, pharmacodynamics, oral sustained-release preparations of L-dopa in fluctuating
and tolerability of tolcapone: a review of early studies in Parkinsons disease. Preliminary findings in 20 patients. J
volunteers. Neurology 50: S31S38. Neural Transm Park Dis Dement Sect 4: 173178.
Jorga KM (1998b). COMT inhibitors: pharmacokinetic and Klein C, Kompf D, Pulkowski U et al. (1997). A study of
pharmacodynamic comparisons. Clin Neuropharmacol visual hallucinations in patients with Parkinsons disease.
21 (Suppl 1): 916. J Neurol 244: 371377.
Jorga KM, Fotteler B, Heizmann P et al. (1998b). Pharmaco- Koller WC, Hutton JT, Tolosa E et al. (1999). Immediate-
kinetics and pharmacodynamics after oral and intravenous release and controlled-release carbidopa/levodopa in PD:
administration of tolcapone, a novel adjunct to Parkinsons a 5-year randomized multicenter study. Carbidopa/Levo-
disease therapy. Eur J Clin Pharmacol 54: 443447. dopa Study Group. Neurology 53: 10121019.
LEVODOPA 65

Korten A, Lodder J, Vreeling F et al. (2001). Stroke and idio- Lees AJ (1995). Comparison of therapeutic effects and mortal-
pathic Parkinsons disease: does a shortage of dopamine ity data of levodopa and levodopa combined with selegiline
offer protection against stroke? Mov Disord 16: 119123. in patients with early, mild Parkinsons disease. Parkinsons
Kostic V, Przedborski S, Flaster E et al. (1991). Early devel- Disease Research Group of the United Kingdom. BMJ 311:
opment of levodopa-induced dyskinesias and response 16021607.
fluctuations in young-onset Parkinsons disease. Neurol- Lesser RP, Fahn S, Snider SR et al. (1979). Analysis of the
ogy 41: 202205. clinical problems in parkinsonism and the complications
Kostic VS, Marinkovic J, Svetel M et al. (2002). The effect of long-term levodopa therapy. Neurology 29: 12531260.
of stage of Parkinsons disease at the onset of levodopa LeWitt PA, Nelson MV, Berchou RC et al. (1989).
therapy on development of motor complications. Eur Controlled-release carbidopa/levodopa (Sinemet 50/200
J Neurol 9: 914. CR4): clinical and pharmacokinetic studies. Neurology 39:
Kuhn W, Roebroek R, Blom H et al. (1998). Elevated plasma 4553; discussion 59.
levels of homocysteine in Parkinsons disease. Eur Neurol Li ST, Dendi R, Holmes C et al. (2002). Progressive loss of
40: 225227. cardiac sympathetic innervation in Parkinsons disease.
Kurlan R (2004). Disabling repetitive behaviors in Parkinsons Ann Neurol 52: 220223.
disease. Mov Disord 19: 433437. Lieberman A, Gopinathan G, Miller E et al. (1990). Rando-
Kurth MC (1997). Using liquid levodopa in the treatment of mized double-blind cross-over study of Sinemet-controlled
Parkinsons disease. A practical guide. Drugs Aging 10: release (CR4 50/200) versus Sinemet 25/100 in Parkinsons
332340. disease. Eur Neurol 30: 7578.
Kurth MC, Tetrud JW, Irwin I et al. (1993). Oral levodopa/ Lieberman AN, Goldstein M, Gopinathan G et al.
carbidopa solution versus tablets in Parkinsons patients (1984). Combined use of benserazide and carbidopa in
with severe fluctuations: a pilot study. Neurology 43: Parkinsons disease. Neurology 34: 227229.
10361039. Lipton SA, Kim WK, Choi YB et al. (1997). Neurotoxicity
Kurth MC, Adler CH, Hilaire MS et al. (1997). Tolcapone associated with dual actions of homocysteine at the N-
improves motor function and reduces levodopa require- methyl-D-aspartate receptor. Proc Natl Acad Sci USA
ment in patients with Parkinsons disease experiencing 94: 59235928.
motor fluctuations: a multicenter, double-blind, rando- Lloyd KG, Davidson L, Hornykiewicz O (1975). The neuro-
mized, placebo-controlled trial. Tolcapone Fluctuator chemistry of Parkinsons disease: effect of L-dopa therapy.
Study Group I. Neurology 48: 8187. J Pharmacol Exp Ther 195: 453464.
Lamberti P, Zoccolella S, Iliceto G et al. (2005). Effects of Lotta T, Vidgren J, Tilgmann C et al. (1995). Kinetics of
levodopa and COMT inhibitors on plasma homocysteine human soluble and membrane-bound catechol O-methyl-
in Parkinsons disease patients. Mov Disord 20: 6972. transferase: a revised mechanism and description of the
Lamberty Y, Klitgaard H (2000). Consequences of pentyle- thermolabile variant of the enzyme. Biochemistry 34:
netetrazole kindling on spatial memory and emotional 42024210.
responding in the rat. Epilepsy Behav 1: 256261. Low PA, Gilden JL, Freeman R et al. (1997). Efficacy of
Lang AE (2001). Acute orthostatic hypotension when start- midodrine vs placebo in neurogenic orthostatic hypoten-
ing dopamine agonist therapy in parkinson disease: the sion. A randomized, double-blind multicenter study.
role of domperidone therapy. Arch Neurol 58: 835. Midodrine Study Group. JAMA 277: 10461051.
Langdon N, Malcolm PN, Parkes JD (1986). Comparison of Lozano AM, Lang AE, Levy R et al. (2000). Neuronal
levodopa with carbidopa, and levodopa with domperidone recordings in Parkinsons disease patients with dyskinesias
in Parkinsons disease. Clin Neuropharmacol 9: 440447. induced by apomorphine. Ann Neurol 47: S141S146.
Langer SZ (1980). Presynaptic regulation of the release of Luchsinger JA, Tang MX, Shea S et al. (2004). Plasma
catecholamines. Pharmacol Rev 32: 337362. homocysteine levels and risk of Alzheimer disease.
Larsen JP, Worm-Petersen J, Siden A et al. (2003). The toler- Neurology 62: 19721976.
ability and efficacy of entacapone over 3 years in patients Lyons KE, Hubble JP, Troster AI et al. (1998). Gender dif-
with Parkinsons disease. Eur J Neurol 10: 137146. ferences in Parkinsons disease. Clin Neuropharmacol
Lautala P, Ethell BT, Taskinen J et al. (2000). The specifi- 21: 118121.
city of glucuronidation of entacapone and tolcapone by Malcolm SL, Allen JG, Bird H et al. (1987). Single-dose
recombinant human UDP-glucuronosyltransferases. Drug pharmacokinetics of Madopar HBS in patients and effect
Metab Dispos 28: 13851389. of food and antacid on the absorption of Madopar HBS
Lawrence de Koning AB, Werstuck GH, Zhou J et al. (2003). in volunteers. Eur Neurol 27 (Suppl 1): 2835.
Hyperhomocysteinemia and its role in the development of Mannisto PT, Kaakkola S (1999). Catechol-O-methyltransferase
atherosclerosis. Clin Biochem 36: 431441. (COMT): biochemistry, molecular biology, pharmacology,
Leenders KL, Palmer AJ, Quinn N et al. (1986). Brain dopa- and clinical efficacy of the new selective COMT inhibitors.
mine metabolism in patients with Parkinsons disease Pharmacol Rev 51: 593628.
measured with positron emission tomography. J Neurol Mannisto PT, Tuomainen P, Toivonen M et al. (1990). Effect
Neurosurg Psychiatry 49: 853860. of acute levodopa on brain catecholamines after selective
66 T. D. HALBIG AND W. C. KOLLER

MAO and COMT inhibition in male rats. J Neural Transm L-DOPA toxicity through multiple mechanisms. J Neural
Park Dis Dement Sect 2: 3143. Transm 104: 317328.
Mannisto PT, Ulmanen I, Lundstrom K et al. (1992). Char- Menza MA, Sage J, Marshall E et al. (1990). Mood changes
acteristics of catechol O-methyl-transferase (COMT) and and on-off phenomena in Parkinsons disease. Mov
properties of selective COMT inhibitors. Prog Drug Res Disord 5: 148151.
39: 291350. Menza MA, Golbe LI, Cody RA et al. (1993). Dopamine-
Maratos EC, Jackson MJ, Pearce RK et al. (2001). Anti- related personality traits in Parkinsons disease. Neurology
parkinsonian activity and dyskinesia risk of ropinirole 43: 505508.
and L-DOPA combination therapy in drug naive MPTP- Merello M, Lees AJ (1992). Beginning-of-dose motor dete-
lesioned common marmosets (Callithrix jacchus). Mov rioration following the acute administration of levodopa
Disord 16: 631641. and apomorphine in Parkinsons disease. J Neurol Neurosurg
Marconi R, Lefebvre-Caparros D, Bonnet AM et al. Psychiatry 55: 10241026.
(1994). Levodopa-induced dyskinesias in Parkinsons Merello M, Pirtosek Z, Bishop S et al. (1992). Cardiovascular
disease phenomenology and pathophysiology. Mov Disord reflexes in Parkinsons disease: effect of domperidone and
9: 212. apomorphine. Clin Auton Res 2: 215219.
Mark MH, Sage JI (1989a). Long-term efficacy of controlled- Merims D, Shabtai H, Korczyn AD et al. (2004). Antiparkin-
release carbidopa/levodopa in patients with advanced sonian medication is not a risk factor for the development
Parkinsons disease. Ann Clin Lab Sci 19: 415421. of hallucinations in Parkinsons disease. J Neural Transm
Mark MH, Sage JI (1989b). Controlled-release carbidopa- 111: 14471453.
levodopa (Sinemet) in combination with standard Sinemet in Metman LV, Hoff J, Mouradian MM et al. (1994). Fluctua-
advanced Parkinsons disease. Ann Clin Lab Sci 19: 101106. tions in plasma levodopa and motor responses with liquid
Markham C, Diamond SG, Treciokas LJ (1974). Carbidopa and tablet levodopa/carbidopa. Mov Disord 9: 463465.
in Parkinson disease and in nausea and vomiting of Michel PP, Hefti F (1990). Toxicity of 6-hydroxydopamine
levodopa. Arch Neurol 31: 128133. and dopamine for dopaminergic neurons in culture. J Neu-
Markianos M, Hadjikonstantinou M, Bistolaki E (1982). rosci Res 26: 428435.
Urinary noradrenaline and serotonin metabolites in drug- Miller JW (2002). Homocysteine, folate deficiency, and
free Parkinson patients and the effect of L-dopa treatment. Parkinsons disease. Nutr Rev 60: 410413.
Acta Neurol Scand 66: 267275. Miller JW, Selhub J, Nadeau MR et al. (2003). Effect of
Marsden CD, Parkes JD (1977). Success and problems of L-dopa on plasma homocysteine in PD patients: relation-
long-term levodopa therapy in Parkinsons disease. Lancet ship to B-vitamin status. Neurology 60: 11251129.
1: 345349. Molina JA, Sainz-Artiga MJ, Fraile A et al. (2000). Patholo-
Marsden CD, Parkes JP, Qinn N (1982). Fluctuations of gic gambling in Parkinsons disease: a behavioral manifes-
disability in Parkinsons disease. In: CD Marsden, S Fahn tation of pharmacologic treatment? Mov Disord 15:
(Eds.), Movement Disorders. Butterworth, London. 869872.
Mastaglia FL, Johnsen RD, Kakulas BA (2002). Prevalence Montastruc JL, Rascol O, Senard JM et al. (1994). A rando-
of stroke in Parkinsons disease: a postmortem study. mised controlled study comparing bromocriptine to which
Mov Disord 17: 772774. levodopa was later added, with levodopa alone in previously
Meco G, Pratesi L, Bonifati V (1991). Cardiovascular untreated patients with Parkinsons disease: a five year
reflexes and autonomic dysfunction in Parkinsons follow up. J Neurol Neurosurg Psychiatry 57: 10341038.
disease. J Neurol 238: 195199. Montastruc JL, Schmitt L, Bagheri H (2003). [Pathological
Melamed E, Hefti F, Wurtman RJ (1980). Nonaminergic gambling behavior in a patient with Parkinsons disease
striatal neurons convert exogenous L-dopa to dopamine treated with levodopa and bromocriptine.] Rev Neurol
in parkinsonism. Ann Neurol 8: 558563. (Paris) 159: 441443.
Melis MR, Argiolas A (1995). Dopamine and sexual behavior. Moore H, Todd CL, Grace AA (1998). Striatal extracellular
Neurosci Biobehav Rev 19: 1938. dopamine levels in rats with haloperidol-induced depolar-
Meltzer HY (1999). The role of serotonin in antipsychotic ization block of substantia nigra dopamine neurons. J Neu-
drug action. Neuropsychopharmacology 21: 106S115S. rosci 18: 50685077.
Mena MA, Pardo B, Casarejos MJ et al. (1992). Neurotoxi- Morgante L, Epifanio A, Spina E et al. (2004). Quetiapine
city of levodopa on catecholamine-rich neurons. Mov and clozapine in parkinsonian patients with dopaminergic
Disord 7: 2331. psychosis. Clin Neuropharmacol 27: 153156.
Mena MA, Pardo B, Paino CL et al. (1993). Levodopa toxi- Morris MS (2003). Homocysteine and Alzheimers disease.
city in foetal rat midbrain neurones in culture: modulation Lancet Neurol 2: 425428.
by ascorbic acid. Neuroreport 4: 438440. Moskovitz C, Moses H 3rd, Klawans HL (1978). Levodopa-
Mena MA, Davila V, Sulzer D (1997a). Neurotrophic effects induced psychosis: a kindling phenomenon. Am J Psychiatry
of L-DOPA in postnatal midbrain dopamine neuron/corti- 135: 669675.
cal astrocyte cocultures. J Neurochem 69: 13981408. Muenter MD, Tyce GM (1971). L-dopa therapy of Parkin-
Mena MA, Casarejos MJ, Carazo A et al. (1997b). Glia sons disease: plasma L-dopa concentration, therapeutic
protect fetal midbrain dopamine neurons in culture from response, and side effects. Mayo Clin Proc 46: 231239.
LEVODOPA 67

Muller T, Werne B, Fowler B et al. (1999). Nigral endothe- Parkinsons disease, does not impair mitochondrial energy
lial dysfunction, homocysteine, and Parkinsons disease. production. Eur J Pharmacol 340: 287294.
Lancet 354: 126127. Nissinen E, Nissinen H, Larjonmaa H et al. (2005). The
Muller T, Woitalla D, Hauptmann B et al. (2001). Decrease COMT inhibitor, entacapone, reduces levodopa-induced
of methionine and S-adenosylmethionine and increase of elevations in plasma homocysteine in healthy adult rats.
homocysteine in treated patients with Parkinsons disease. J Neural Transm 112 (9) 12131221.
Neurosci Lett 308: 5456. Noble EP (2000). The DRD2 gene in psychiatric and neuro-
Murer MG, Dziewczapolski G, Menalled LB et al. (1998). logical disorders and its phenotypes. Pharmacogenomics
Chronic levodopa is not toxic for remaining dopamine 1: 309333.
neurons, but instead promotes their recovery, in rats with Nordera GP, Lorizio A, Lion P et al. (1987). Treatment of par-
moderate nigrostriatal lesions. Ann Neurol 43: 561575. kinsonian conditions with a controlled-release form of levo-
Myllyla VV, Sotaniemi KA, Illi A et al. (1993). Effect of dopapreliminary study. Eur Neurol 27 (Suppl 1): 7680.
entacapone, a COMT inhibitor, on the pharmacokinetics Novartis Pharmaceuticals Corporation (2000). COMTAN
of levodopa and on cardiovascular responses in patients Prescribing Information, East Hanover, New Jersey, USA.
with Parkinsons disease. Eur J Clin Pharmacol 45: Novartis Pharmaceuticals Corporation (2005). CLOZARIL
419423. Prescribing Information, East Hanover, New Jersey.
Myllyla VV, Jackson M, Larsen JP (1997). Efficacy and Nutt JG (1987). On-off phenomenon: relation to levodopa
safety of tolcapone in levodopa-treated Parkinsons dis- pharmacokinetics and pharmacodynamics. Ann Neurol
ease patients with wearing-off phenomenon: a multicen- 22: 535540.
tre, double blind, randomized placebo-controlled trial. Eur Nutt JG (1990). Levodopa-induced dyskinesia: review,
J Neurol 4: 333341. observations, and speculations. Neurology 40: 340345.
Myllyla VV, Kultalahti ER, Haapaniemi H et al. (2001). Nutt JG (1992). Pharmacokinetics and pharmacodynamics of
FILOMEN Study Group. Twelve-month safety of entaca- levodopa. In: WC Koller, (Ed.), Handbook of Parkinsons
pone in patients with Parkinsons disease. Eur J Neurol Disease. Marcel Dekker, New York, pp. 411732.
8: 5360. Nutt JG (1998). Catechol-O-methyltransferase inhibitors for
Mytilineou C, Han SK, Cohen G (1993). Toxic and protec- treatment of Parkinsons disease. Lancet 351: 12211222.
tive effects of L-dopa on mesencephalic cell cultures. Nutt JG (2000). Effect of COMT inhibition on the phar-
J Neurochem 61: 14701478. macokinetics and pharmacodynamics of levodopa in par-
Naber D, Lambert M (2004). Aripiprazole: a new atypical kinsonian patients. Neurology 55: S33S37; discussion
antipsychotic with a different pharmacological mechan- S38S41.
ism. Prog Neuropsychopharmacol Biol Psychiatry 28: Nutt JG, Fellman JH (1984). Pharmacokinetics of levodopa.
12131219. Clin Neuropharmacol 7: 3549.
Nakaso K, Yasui K, Kowa H et al. (2003). Hypertrophy of Nutt JG, Woodward WR, Anderson JL (1985). The effect of
IMC of carotid artery in Parkinsons disease is associated carbidopa on the pharmacokinetics of intravenously admi-
with L-DOPA, homocysteine, and MTHFR genotype. nistered levodopa: the mechanism of action in the treat-
J Neurol Sci 207: 1923. ment of parkinsonism. Ann Neurol 18: 537543.
Napolitano A, Zurcher G, Da Prada M (1995). Effects of tol- Nutt JG, Gancher ST, Woodward WR (1988). Does an inhi-
capone, a novel catechol-O-methyltransferase inhibitor, on bitory action of levodopa contribute to motor fluctuations?
striatal metabolism of L-dopa and dopamine in rats. Eur Neurology 38: 15531557.
J Pharmacol 273: 215221. Nutt JG, Woodward WR, Beckner RM et al. (1994). Effect
Nausieda PA, Tanner CM, Klawans HL (1983). Serotonergi- of peripheral catechol-O-methyltransferase inhibition on
cally active agents in levodopa-induced psychiatric toxicity the pharmacokinetics and pharmacodynamics of levodopa
reactions. Adv Neurol 37: 2332. in parkinsonian patients. Neurology 44: 913919.
Nausieda PA, Pfeiffer RF, Tagliati M et al. (2005). A multi- Nutt JG, Carter JH, Woodward WR (1995). Long-duration
center, open-label, sequential study comparing preferences response to levodopa. Neurology 45: 16131616.
for carbidopa-levodopa orally disintegrating tablets and Nyholm D, Askmark H, Gomes-Trolin C et al. (2003). Opti-
conventional tablets in subjects with Parkinsons disease. mizing levodopa pharmacokinetics: intestinal infusion ver-
Clin Ther 27: 5863. sus oral sustained-release tablets. Clin Neuropharmacol
Nilsson D, Hansson LE, Johansson K et al. (1998). Long- 26: 156163.
term intraduodenal infusion of a water based levodopa- Nyholm D, Nilsson Remahl AI, Dizdar N et al. (2005). Duo-
carbidopa dispersion in very advanced Parkinsons denal levodopa infusion monotherapy vs oral polyphar-
disease. Acta Neurol Scand 97: 175183. macy in advanced Parkinson disease. Neurology 64:
Nilsson D, Nyholm D, Aquilonius SM (2001). Duodenal 216223.
levodopa infusion in Parkinsons diseaselong-term Obeso JA, Olanow CW (1997). Dopamine agonists in early
experience. Acta Neurol Scand 104: 343348. Parkinsons disease. In: CW Olanow, JA Obeso (Eds.),
Nissinen E, Kaheinen P, Penttila KE et al. (1997). Entaca- Beyond the Decade of the Brain, Vol. 2. Wells Medical
pone, a novel catechol-O-methyltransferase inhibitor for Ltd, Tunbridge Wells, Kent, UK, pp. 1135.
68 T. D. HALBIG AND W. C. KOLLER

Obeso JA, Rodriguez-Oroz MC, Chana P et al. (2000). The Parker WD Jr, Boyson SJ, Parks JK (1989). Abnormalities of
evolution and origin of motor complications in Parkinsons the electron transport chain in idiopathic Parkinsons dis-
disease. Neurology 55: S13S20; discussion S21S13. ease. Ann Neurol 26: 719723.
Olanow CW (2000). Tolcapone and hepatotoxic effects. Parkinson Study Group (1989). Effect of deprenyl on the
Tasmar Advisory Panel. Arch Neurol 57: 263267. progression of disability in early Parkinsons disease.
Olanow CW, Hauser RA, Gauger L et al. (1995). The effect The Parkinson Study Group. N Engl J Med 321:
of deprenyl and levodopa on the progression of Parkin- 13641371.
sons disease. Ann Neurol 38: 771777. Parkinson Study Group (1996a). Impact of deprenyl and
Olanow CW, Watts RL, Koller WC (2001). An algorithm tocopherol treatment on Parkinsons disease in DATATOP
(decision tree) for the management of Parkinsons disease patients requiring levodopa. Ann Neurol 39: 3745.
(2001): treatment guidelines. Neurology 56: S1S88. Parkinson Study Group (1996b). Impact of deprenyl and
Ondo WG, Levy JK, Vuong KD et al. (2002). Olanzapine tocopherol treatment on Parkinsons disease in DATATOP
treatment for dopaminergic-induced hallucinations. Mov subjects not requiring levodopa. Ann Neurol 39: 2936.
Disord 17: 10311035. Parkinson Study Group (1997). Entacapone improves motor
Ondo WG, Tintner R, Dat Voung K et al. (2005). Double- fluctuations in levodopa-treated Parkinsons disease
blind, placebo-controlled, unforced titration parallel patients. Ann Neurol 42: 747755.
trial of quetiapine for dopaminergic-induced hallucina- Parkinson Study Group (1999). Low-dose clozapine for the
tions in Parkinsons disease. Mov Disord 20 (8): treatment of drug-induced psychosis in Parkinsons dis-
958963. ease. The Parkinson Study Group. N Engl J Med 340:
Onofrj M, Thomas A, Iacono D et al. (2001). Switch-over 757763.
from tolcapone to entacapone in severe Parkinsons Parkinson Study Group (2000). Pramipexole vs levodopa as
disease patients. Eur Neurol 46: 1116. initial treatment for Parkinson disease: a randomized
OSuilleabhain PE, Bottiglieri T, Dewey RB Jr et al. controlled trial. JAMA 284: 19311938.
(2004a). Modest increase in plasma homocysteine follows Parkinson Study Group (2002a). Dopamine transporter
levodopa initiation in Parkinsons disease. Mov Disord 19: brain imaging to assess the effects of pramipexole vs levo-
14031408. dopa on Parkinson disease progression. JAMA 287:
OSuilleabhain PE, Sung V, Hernandez C et al. (2004b). 16531661.
Elevated plasma homocysteine level in patients with Parkinson Study Group (2002b). A controlled trial of rasagi-
Parkinson disease: motor, affective, and cognitive associa- line in early Parkinson disease: the TEMPO Study. Arch
tions. Arch Neurol 61: 865868. Neurol 59: 19371943.
Pacchetti C, Martignoni E, Sibilla L et al. (1990). Effective- Parkinson Study Group (2004a). Levodopa and the progres-
ness of Madopar HBS plus Madopar standard in patients sion of Parkinsons disease. N Engl J Med 351:
with fluctuating Parkinsons disease: two years of follow- 24982508.
up. Eur Neurol 30: 319323. Parkinson Study Group (2004b). Pramipexole vs levodopa as
Pahwa R, Busenbark K, Huber SJ et al. (1993). Clinical initial treatment for Parkinson disease: a 4-year rando-
experience with controlled-release carbidopa/levodopa in mized controlled trial. Arch Neurol 61: 10441053.
Parkinsons disease. Neurology 43: 677681. Parkinson Study Group (2004c). A controlled, randomized,
Papa SM, Desimone R, Fiorani M et al. (1999). Internal delayed-start study of rasagiline in early Parkinson
globus pallidus discharge is nearly suppressed during disease. Arch Neurol 61: 561566.
levodopa-induced dyskinesias. Ann Neurol 46: 732738. Parkinson Study Group (2005). A randomized placebo-con-
Papavasiliou PS, Cotzias GC, Duby SE et al. (1972). Levodo- trolled trial of rasagiline in levodopa-treated patients with
pa in Parkinsonism: potentiation of central effects with a Parkinson disease and motor fluctuations: the PRESTO
peripheral inhibitor. N Engl J Med 286: 814. study. Arch Neurol 62: 241248.
Papavasiliou PS, Cotzias GC, Lawrence WH (1973). Levodo- Pearce RK, Banerji T, Jenner P et al. (1998). De novo
pa and dopamine in cerebrospinal fluid. Neurology 23: administration of ropinirole and bromocriptine induces
756759. less dyskinesia than L-dopa in the MPTP-treated marmoset.
Pappert EJ, Goetz CG, Niederman F et al. (1996). Liquid Mov Disord 13: 234241.
levodopa/carbidopa produces significant improvement in Perry EK, Kerwin J, Perry RH et al. (1990). Visual halluci-
motor function without dyskinesia exacerbation. Neurol- nations and the cholinergic system in dementia. J Neurol
ogy 47: 14931495. Neurosurg Psychiatry 53: 88.
Pappert EJ, Goetz CG, Niederman FG et al. (1999). Halluci- Perry IJ, Refsum H, Morris RW et al. (1995). Prospective
nations, sleep fragmentation, and altered dream phenom- study of serum total homocysteine concentration and risk
ena in Parkinsons disease. Mov Disord 14: 117121. of stroke in middle-aged British men. Lancet 346:
Pardo B, Mena MA, Fahn S et al. (1993). Ascorbic acid 13951398.
protects against levodopa-induced neurotoxicity on a cate- Perry TL, Godin DV, Hansen S (1982). Parkinsons disease:
cholamine-rich human neuroblastoma cell line. Mov a disorder due to nigral glutathione deficiency? Neurosci
Disord 8: 278284. Lett 33: 305310.
LEVODOPA 69

Pezzoli G, Tesei S, Ferrante C et al. (1988). Madopar HBS in Rajput AH, Stern W, Laverty WH (1984). Chronic low-dose
fluctuating parkinsonian patients: two-year treatment. levodopa therapy in Parkinsons disease: an argument for
Mov Disord 3: 3745. delaying levodopa therapy. Neurology 34: 991996.
Pfeiffer RF (1998). Gastrointestinal dysfunction in Parkinsons Rajput AH, Martin W, Saint-Hilaire MH et al. (1997). Tolca-
disease. Clin Neurosci 5: 136146. pone improves motor function in parkinsonian patients
Pfeiffer RF (2003). Gastrointestinal dysfunction in Parkinsons with the wearing-off phenomenon: a double-blind,
disease. Lancet Neurol 2: 107116. placebo-controlled, multicenter trial. Neurology 49:
Physicians Desk Reference (2003). 57th edn. Thomson PDR, 10661071.
Montvale, NJ. Rascol O, Brooks DJ, Korczyn AD et al. (2000). A five-year
Piccini P, Brooks DJ, Korpela K et al. (2000). The catechol-O- study of the incidence of dyskinesia in patients with early
methyltransferase (COMT) inhibitor entacapone enhances Parkinsons disease who were treated with ropinirole or
the pharmacokinetic and clinical response to Sinemet CR levodopa. 056 Study Group. N Engl J Med 342:
in Parkinsons disease. J Neurol Neurosurg Psychiatry 68: 14841491.
589594. Rascol O, Brooks DJ, Melamed E et al. (2005). Rasagiline as
Poewe WH, Lees AJ, Stern GM (1986). Low-dose L-dopa an adjunct to levodopa in patients with Parkinsons disease
therapy in Parkinsons disease: a 6-year follow-up study. and motor fluctuations (LARGO, Lasting effect in Adjunct
Neurology 36: 15281530. therapy with Rasagiline Given Once daily, study): a ran-
Poewe WH, Lees AJ, Stern GM (1987). Clinical and pharma- domised, double-blind, parallel-group trial. Lancet 365:
cokinetic observations with Madopar HBS in hospitalized 947954.
patients with Parkinsons disease and motor fluctuations. Reinikainen K, Leinonen M, Isojarvi J (2002). Predicting the
Eur Neurol 27 (Suppl 1): 9397. need for levodopa dose reduction after entacapone initia-
Poewe WH, Deuschl G, Gordin A et al. (2002a). Efficacy tion in PD patients with end-of-dose wearing-off. Mov
and safety of entacapone in Parkinsons disease patients Disord 17 (Suppl 5): P111.
with suboptimal levodopa response: a 6-month rando- Riederer P, Youdim MB, Rausch WD et al. (1978). On the
mized placebo-controlled double-blind study in Germany mode of action of L-deprenyl in the human central
and Austria (Celomen study). Acta Neurol Scand 105: nervous system. J Neural Transm 43: 217226.
245255. Riederer P, Sofic E, Rausch WD et al. (1989). Transition
Poewe WH, Deuschl G, Gordin A et al. (2002b). CELOMEN metals, ferritin, glutathione, and ascorbic acid in parkinso-
Study Group. Efficacy and safety of entacapone in Parkinsons nian brains. J Neurochem 52: 515520.
disease patients with suboptimal levodopa response: a Rinne UK, Molsa P (1979). Levodopa with benserazide
6-month randomized placebo-controlled double-blind study or carbidopa in Parkinson disease. Neurology 29:
in Germany and Austria (Celomen study). Acta Neurol Scand 15841589.
105: 245255. Rinne UK, Larsen JP, Siden A et al. (1998a). Entacapone
Postuma RB, Lang AE (2004). Homocysteine and levodopa: enhances the response to levodopa in parkinsonian
should Parkinson disease patients receive preventative patients with motor fluctuations. Nomecomt Study Group.
therapy? Neurology 63: 886891. Neurology 51: 13091314.
Quigley EM (1996). Gastrointestinal dysfunction in Parkinsons Rinne UK, Bracco F, Chouza C et al. (1998b). Early treat-
disease. Semin Neurol 16: 245250. ment of Parkinsons disease with cabergoline delays the
Quinn N, Marsden CD, Parkes JD (1982). Complicated onset of motor complications. Results of a double-blind
response fluctuations in Parkinsons disease: response to levodopa controlled trial. The PKDS009 Study Group.
intravenous infusion of levodopa. Lancet 2: 412415. Drugs 55 (Suppl 1): 2330.
Quinn NP, Toone B, Lang AE et al. (1983). Dopa dose- Roberts JW, Cora-Locatelli G, Bravi D et al. (1993). Cate-
dependent sexual deviation. Br J Psychiatry 142: 296298. chol-O-methyltransferase inhibitor tolcapone prolongs
Quinn NP, Marion MH, Marsden CD (1987). Open study of levodopa/carbidopa action in parkinsonian patients.
Madopar HBS, a new formulation of levodopa with Neurology 43: 26852688.
benserazide, in 13 patients with Parkinsons disease and Rodnitzky RL, Dickins QS, Dobson J (1989). Long-term
on-off fluctuations. Eur Neurol 27 (Suppl 1): 105113. clinical efficacy of Sinemet CR in patients with Parkin-
Rabey JM, Vardi J, Askenazi JJ et al. (1977). L-tryptophan sons disease. Neurology 39: 9295; discussion 95.
administration in L-dopa-induced hallucinations in elderly Rogers JD, Sanchez-Saffon A, Frol AB et al. (2003).
Parkinsonian patients. Gerontology 23: 438444. Elevated plasma homocysteine levels in patients treated
Rabey JM, Treves TA, Neufeld MY et al. (1995). Low-dose with levodopa: association with vascular disease. Arch
clozapine in the treatment of levodopa-induced mental Neurol 60: 5964.
disturbances in Parkinsons disease. Neurology 45: Rondot P, Ziegler M, Aymard N et al. (1989). Effect of con-
432434. trolled-release carbidopa/levodopa on motor performance
Rabey JM, Sagi I, Huberman M et al. (2000). Rasagiline in advanced Parkinsons disease. Neurology 39: 7477;
mesylate, a new MAO-B inhibitor for the treatment of discussion 95.
Parkinsons disease: a double-blind study as adjunctive Rouru J, Gordin A, Huupponen R et al. (1999). Pharmacoki-
therapy to levodopa. Clin Neuropharmacol 23: 324330. netics of oral entacapone after frequent multiple dosing
70 T. D. HALBIG AND W. C. KOLLER

and effects on levodopa disposition. Eur J Clin Pharmacol tration with levodopa/carbidopa to healthy volunteers.
55: 461467. Clin Neuropharmacol 20: 531541.
Ruottinen HM, Rinne JO, Ruotsalainen UH et al. (1995). Seedat S, Kesler S, Niehaus DJ et al. (2000). Pathological
Striatal [18F]fluorodopa utilization after COMT inhibition gambling behaviour: emergence secondary to treatment
with entacapone studied with PET in advanced Parkin- of Parkinsons disease with dopaminergic agents. Depress
sons disease. J Neural Transm Park Dis Dement Sect Anxiety 11: 185186.
10: 91106. Senard JM, Verwaerde P, Rascol O et al. (1995). Effects of
Ruottinen HM, Rinne UK (1996a). Effect of one months acute levodopa administration on blood pressure and heart
treatment with peripherally acting catechol-O-methyltrans- variability in never treated parkinsonians. Hypertens Res
ferase inhibitor, entacapone, on pharmacokinetics and 18 (Suppl 1): S175S177.
motor response to levodopa in advanced parkinsonian Senard JM, Rai S, Lapeyre-Mestre M et al. (1997). Preva-
patients. Clin Neuropharmacol 19: 222233. lence of orthostatic hypotension in Parkinsons disease.
Ruottinen HM, Rinne UK (1996b). A double-blind pharma- J Neurol Neurosurg Psychiatry 63: 584589.
cokinetic and clinical dose-response study of entacapone Seshadri S, Beiser A, Selhub J et al. (2002). Plasma homo-
as an adjuvant to levodopa therapy in advanced Parkin- cysteine as a risk factor for dementia and Alzheimers dis-
sons disease. Clin Neuropharmacol 19: 283296. ease. N Engl J Med 346: 476483.
Sachs C, Berglund B, Kaijser L (1985). Autonomic cardio- Shaffer H, Hall M (1996). Estimating the prevalence of ado-
vascular responses in parkinsonism: effect of levodopa lescent gambling disorders: a quantitative synthesis and
with dopa-decarboxylase inhibition. Acta Neurol Scand guide toward standard gambling nomenclature. J Gambl
71: 3742. Stud 12: 193214.
Sage JI, Mark MH (1988). Comparison of controlled-release Sharf B, Moskovitz C, Lupton MD et al. (1978). Dream phe-
Sinemet (CR4) and standard Sinemet (25 mg/100 mg) in nomena induced by chronic levodopa therapy. J Neural
advanced Parkinsons disease: a double-blind, crossover Transm 43: 143151.
study. Clin Neuropharmacol 11: 174179. Shoulson I, Glaubiger GA, Chase TN (1975). On-off
Sanchez-Ramos JR, Ortoll R, Paulson GW (1996). Visual response. Clinical and biochemical correlations during
hallucinations associated with Parkinson disease. Arch oral and intravenous levodopa administration in parkinso-
Neurol 53: 12651268. nian patients. Neurology 25: 11441148.
Sasahara K, Nitanai T, Habara T et al. (1980). Dosage form Siegfried J (1987). Therapeutic value of Madopar HBS: judg-
design for improvement of bioavailability of levodopa II: ment after 2 years experience. Eur Neurol 27 (Suppl 1):
bioavailability of marketed levodopa preparations in dogs 98104.
and parkinsonian patients. J Pharm Sci 69: 261265. Siemers ER, Shekhar A, Quaid K et al. (1993). Anxiety and
Sawle GV, Burn DJ, Morrish PK et al. (1994). The effect of motor performance in Parkinsons disease. Mov Disord 8:
entacapone (OR-611) on brain [18F]-6-L-fluorodopa meta- 501506.
bolism: implications for levodopa therapy of Parkinsons Sigurdardottir GR, Nilsson C, Odin P et al. (2001). Car-
disease. Neurology 44: 12921297. diovascular effects of domperidone in patients with
Schapira AH, Cooper JM, Dexter D et al. (1990). Mitochon- Parkinsons disease treated with apomorphine. Acta
drial complex I deficiency in Parkinsons disease. Neurol Scand 104: 9296.
J Neurochem 54: 823827. Skibba JL, Pinckley J, Gilbert EF et al. (1972). Multiple pri-
Schonfeldt-Lecuona C, Connemann BJ (2004). Aripiprazole mary melanoma following administration of levodopa.
and Parkinsons disease psychosis. Am J Psychiatry 161: Arch Pathol 93: 556561.
373374. Smith LA, Gordin A, Jenner P et al. (1997). Entacapone
Schrag A, Quinn N (2000). Dyskinesias and motor fluctua- enhances levodopa-induced reversal of motor disability
tions in Parkinsons disease. A community-based study. in MPTP-treated common marmosets. Mov Disord 12:
Brain 123 (Pt 11): 22972305. 935945.
Schultz E, Nissinen E (1989). Inhibition of rat liver and duo- Smith LA, Tel BC, Jackson MJ et al. (2002). Repeated
denum soluble catechol-O-methyltransferase by a tight- administration of piribedil induces less dyskinesia than
binding inhibitor OR-462. Biochem Pharmacol 38: L-dopa in MPTP-treated common marmosets: a beha-
39533956. vioural and biochemical investigation. Mov Disord 17:
Schultz E, Nissinen E, Kaakkola S (1989). Determination of 887901.
catechol-O-methyltransferase activity in erythrocytes by Smith LA, Jackson MJ, Hansard MJ et al. (2003). Effect of
high performance liquid chromatography with electroche- pulsatile administration of levodopa on dyskinesia induc-
mical detection. Biomed Chromatogr 3: 6467. tion in drug-naive MPTP-treated common marmosets:
Schultz W (1986). Responses of midbrain dopamine neurons effect of dose, frequency of administration, and brain
to behavioral trigger stimuli in the monkey. J Neurophy- exposure. Mov Disord 18: 487495.
siol 56: 14391461. Smith LA, Jackson MJ, Al-Barghouthy G et al. (2005). Multi-
Sedek G, Jorga K, Schmitt M et al. (1997). Effect of tolca- ple small doses of levodopa plus entacapone produce con-
pone on plasma levodopa concentrations after coadminis- tinuous dopaminergic stimulation and reduce dyskinesia
LEVODOPA 71

induction in MPTP-treated drug-naive primates. Mov Tornwall M, Kaakkola S, Tuomainen P et al. (1994). Com-
Disord 20: 306314. parison of two new inhibitors of catechol O-methylation
Sofic E, Paulus W, Jellinger K et al. (1991). Selective on striatal dopamine metabolism: a microdialysis study
increase of iron in substantia nigra zona compacta of in rats. Br J Pharmacol 112: 1318.
parkinsonian brains. J Neurochem 56: 978982. Tretter L, Sipos I, Adam-Vizi V (2004). Initiation of neuro-
Squires RF (1972). Multiple forms of monoamine oxidase in nal damage by complex I deficiency and oxidative stress
intact mitochondria as characterized by selective inhibitors in Parkinsons disease. Neurochem Res 29: 569577.
and thermal stability: a comparison of eight mammalian Troconiz IF, Naukkarinen TH, Ruottinen HM et al. (1998).
species. Adv Biochem Psychopharmacol 5: 355370. Population pharmacodynamic modeling of levodopa in
Steckler T, Sahgal A (1995). The role of serotonergic-choli- patients with Parkinsons disease receiving entacapone.
nergic interactions in the mediation of cognitive beha- Clin Pharmacol Ther 64: 106116.
viour. Behav Brain Res 67: 165199. Trugman JM, James CL, Wooten GF (1991). D1/D2 dopa-
Steece-Collier K, Collier TJ, Sladek CD et al. (1990). mine receptor stimulation by L-dopa. A [14C]-2-deoxy-
Chronic levodopa impairs morphological development of glucose autoradiographic study. Brain 114 (Pt 3):
grafted embryonic dopamine neurons. Exp Neurol 110: 14291440.
201208. U.K. Madopar CR Study Group (1989). A comparison of
Steiger MJ, Stocchi F, Bramante L et al. (1992). The clinical Madopar CR and standard Madopar in the treatment of
efficacy of single morning doses of levodopa methyl ester: nocturnal and early-morning disability in Parkinsons
dispersible Madopar and Sinemet plus in Parkinson dis- disease. Clin Neuropharmacol 12: 498505.
ease. Clin Neuropharmacol 15: 501504. Uitti RJ, Tanner CM, Rajput AH et al. (1989). Hypersexuality
Stern M, Dulaney E, Gruber SB et al. (1991). The epidemiol- with antiparkinsonian therapy. Clin Neuropharmacol 12:
ogy of Parkinsons disease. A case-control study of young- 375383.
onset and old-onset patients. Arch Neurol 48: 903907. Valeant Pharmaceuticals International (2006). Prescribing
Stocchi F, Barbato L, Bramante L et al. (1994). The clinical information Tasmar (tolcapone) USA. Valeant Pharmaceu-
efficacy of a single afternoon dose of levodopa methyl ticals International, Aliso Viejo, CA.
ester: a double-blind cross-over study versus placebo. van Deelen RA, Rommers MK, Eerenberg JG et al.
Funct Neurol 9: 259264. (2002). [Hypersexuality during use of levodopa]. Ned
Stocchi F, Barbato L, Bramante L et al. (1996). Fluctuating Tijdschr Geneeskd 146: 20952098.
parkinsonism: a pilot study of single afternoon dose of Verhagen Metman L, Locatelli ER, Bravi D et al. (1997).
levodopa methyl ester. J Neurol 243: 377380. Apomorphine responses in Parkinsons disease and the
Struck LK, Rodnitzky RL, Dobson JK (1990). Stroke and pathogenesis of motor complications. Neurology 48:
its modification in Parkinsons disease. Stroke 21: 369372.
13951399. Vogel HP, Schiffter R (1983). Hypersexualitya complica-
Sudo J, Iwase H, Terui J et al. (1998). Transdermal absorp- tion of dopaminergic therapy in Parkinsons disease. Phar-
tion of L-dopa from hydrogel in rats. Eur J Pharm Sci 7: macopsychiatria 16: 107110.
6771. von Bohlen und Halbach O, Dermietzel R (2002). Dopamine.
Sweet RD, Lee JE, McDowell FH (1972). Methyldopa as an In: Neurotransmitters and Neuromodulators: Wiley-VCH
adjunct to levodopa treatment of Parkinsons disease. Clin Verlag GmbH & Co. KGaA, Weinheim.
Pharmacol Ther 13: 2327. Wakabayashi K, Takahashi H (1997). Neuropathology of
Syed N, Murphy J, Zimmerman T Jr et al. (1998). Ten years autonomic nervous system in Parkinsons disease. Eur
experience with enteral levodopa infusions for motor fluc- Neurol 38 (Suppl 2): 27.
tuations in Parkinsons disease. Mov Disord 13: 336338. Wakabayashi K, Takahashi H, Ohama E et al. (1993). Lewy
Thompson JF, Scolyer RA, Kefford RF (2005). Cutaneous bodies in the visceral autonomic nervous system in Parkin-
melanoma. Lancet 365: 687701. sons disease. Adv Neurol 60: 609612.
Tohgi H, Abe T, Takahashi S et al. (1993). Alterations in the Waters CH, Kurth M, Bailey P et al. (1997). Tolcapone in
concentration of serotonergic and dopaminergic substances stable Parkinsons disease: efficacy and safety of long-
in the cerebrospinal fluid of patients with Parkinsons dis- term treatment. The Tolcapone Stable Study Group. Neu-
ease, and their changes after L-dopa administration. Neurosci rology 49: 665671.
Lett 159: 135138. Webster RA (2001). Dopamine (DA). In: RA Webster,
Tohgi H, Abe T, Yamazaki K et al. (1995). Effects of the (Ed.), Neurotransmitters, Drugs and Brain Function. John
catechol-O-methyltransferase inhibitor tolcapone in Par- Wiley & Sons Ltd, New York.
kinsons disease: correlations between concentrations of Weiner WJ, Singer C, Sanchez-Ramos JR et al. (1993). Levo-
dopaminergic substances in the plasma and cerebrospinal dopa, melanoma, and Parkinsons disease. Neurology 43:
fluid and clinical improvement. Neurosci Lett 192: 674677.
165168. Westin JE, Andersson M, Lundblad M et al. (2001). Persis-
Tolosa E, Marti MJ, Valldeoriola F et al. (1998). History of tent changes in striatal gene expression induced by long-
levodopa and dopamine agonists in Parkinsons disease term L-DOPA treatment in a rat model of Parkinsons
treatment. Neurology 50: S2S10; discussion S44S18. disease. Eur J Neurosci 14: 11711176.
72 T. D. HALBIG AND W. C. KOLLER

Whone AL, Watts RL, Stoessl AJ et al. (2003). Slower pro- Wright CB, Lee HS, Paik MC et al. (2004). Total homocys-
gression of Parkinsons disease with ropinirole versus teine and cognition in a tri-ethnic cohort: the Northern
levodopa: the REAL-PET study. Ann Neurol 54: 93101. Manhattan Study. Neurology 63: 254260.
Wick MM (1979). Levodopa and dopamine analogs: melanin Yahr MD, Duvoisin RC, Mendoza MR et al. (1971). Modifi-
precursors as antitumor agents in experimental human and cation of L-dopa therapy of Parkinsonism by alpha-
murine leukemia. Cancer Treat Rep 63: 991997. methyldopa hydrazine (MK-486). Trans Am Neurol Assoc
Wick MM (1980). Levodopa and dopamine analogs as DNA 96: 5558.
polymerase inhibitors and antitumor agents in human mel- Yamamoto M, Yokochi M, Kuno S et al. (1997). Effects of
anoma. Cancer Res 40: 14141418. tolcapone, a catechol-O-methyltransferase inhibitor, on
Wick MM (1987). Inhibition of transformation by levodopa- motor symptoms and pharmacokinetics of levodopa in
carbidopa in lymphocytes derived from patients with mel- patients with Parkinsons disease. J Neural Transm 104:
anoma. J Invest Dermatol 88: 535537. 229236.
Wick MM (1989). Levodopa/dopamine analogs as inhibitors Yasui K, Kowa H, Nakaso K et al. (2000). Plasma homocys-
of DNA synthesis in human melanoma cells. J Invest Der- teine and MTHFR C677T genotype in levodopa-treated
matol 92: 329S331S. patients with PD. Neurology 55: 437440.
Wikberg T, Ottoila P, Taskinen J (1993a). Identification of Yasui K, Nakaso K, Kowa H et al. (2003). Levodopa-
major urinary metabolites of the catechol-O-methyltrans- induced hyperhomocysteinaemia in Parkinsons disease.
ferase inhibitor entacapone in the dog. Eur J Drug Metab Acta Neurol Scand 108: 6667.
Pharmacokinet 18: 359367. Yeh KC, August TF, Bush DF et al. (1989). Pharmacoki-
Wikberg T, Vuorela A, Ottoila P et al. (1993b). Identification netics and bioavailability of Sinemet CR: a summary of
of major metabolites of the catechol-O-methyltransferase human studies. Neurology 39: 2538.
inhibitor entacapone in rats and humans. Drug Metab Zeng BY, Pearce RK, MacKenzie GM et al. (2000). Altera-
Dispos 21: 8192. tions in preproenkephalin and adenosine-2a receptor
Wilde MI, Markham A (1996). Ondansetron. A review of its mRNA, but not preprotachykinin mRNA correlate with
pharmacology and preliminary clinical findings in novel occurrence of dyskinesia in normal monkeys chroni-
applications. Drugs 52: 773794. cally treated with L-DOPA. Eur J Neurosci 12:
Wilding IR, Hardy JG, Davis SS et al. (1991). Characterisation of 10961104.
the in vivo behaviour of a controlled-release formulation of Ziv I, Zilkha-Falb R, Offen D et al. (1997). Levodopa
levodopa (Sinemet CR). Clin Neuropharmacol 14: 305321. induces apoptosis in cultured neuronal cellsa possible
Wolters EC, Tesselaar HJ (1996). International (NL-UK) accelerator of nigrostriatal degeneration in Parkinsons
double-blind study of Sinemet CR and standard Sinemet disease? Mov Disord 12: 1723.
(25/100) in 170 patients with fluctuating Parkinsons Zoldan J, Friedberg G, Livneh M et al. (1995). Psychosis
disease. J Neurol 243: 235240. in advanced Parkinsons disease: treatment with ondanse-
Wolters EC, Horstink MW, Roos RA et al. (1992). Clinical tron, a 5-HT3 receptor antagonist. Neurology 45:
efficacy of Sinemet CR 50/200 versus Sinemet 25/100 in 13051308.
patients with fluctuating Parkinsons disease. An open, Zurcher G, Colzi A, Da Prada M (1990). Ro 407592: inhi-
and a double-blind, double-dummy, multicenter treatment bition of COMT in rat brain and extracerebral tissues.
evaluation. The Dutch Sinemet CR Study Group. Clin J Neural Transm Suppl 32: 375380.
Neurol Neurosurg 94: 205211.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 33

Dopamine agonists

OLIVIER RASCOL1,2*, TARIK SLAOUI2, WAFA REGRAGUI2, FABIENE ORY-MAGNE2,


CHRISTINE BREFEL-COURBON1,2 AND JEAN-LOUIS MONTASTRUC1

1
Department of Clinical Pharmacology, Clinical Investigation Center, University Hospital, Toulouse, France
2
Department of Neurosciences, University Hospital, Toulouse, France

The discovery of a dopaminergic deficit in the nigros- which is used as subcutaneous injections or infusions.
triatal pathway of patients with Parkinsons disease All share the property of binding to the D2-like family
led to the introduction of levodopa therapy (Birkmayer of dopamine receptors (D2, D3, D4). All have specific
and Hornykiewicz, 1970). After 40 years, levodopa pharmacodynamic and pharmacokinetic profiles (Mon-
remains the gold-standard antiparkinsonian treament. tastruc et al., 1993; Uitti and Ahlskog, 1996; Deleu
Nevertheless, the initial good therapeutic efficacy of et al., 2002). There are many randomized controlled
levodopa is often confounded within a few years of trials (RCTs) and a large body of clinical experience to
the start of treatment by the development of motor support the usefulness of dopamine agonists in the treat-
complications (fluctuations, abnormal movements) and ment of Parkinsons disease. However several issues
other problems (Marsden et al., 1987; Nutt, 1990; Ras- remain a matter of controversy. Among these, important
col et al., 2003). Because of these limitations, the treat- ones are the place of the agonists (as a class) in the
ment of patients with Parkinsons disease has expanded global strategy of Parkinsons disease therapy (before
to incorporate additional pharmacologic approaches. or after levodopa) and the potential advantages or disad-
Among these other therapeutic options, dopamine- vantages of a given agonist versus another one.
receptor agonists are widely used. This chapter will
focus on the use of dopamine agonists as antiparkinso-
nian medications. It will not cover their role in the treat- 33.1. Pharmacological properties of dopamine
ment of other movement disorders, such as the restless- agonists
legs syndrome (Happe and Trenkwalder, 2004; Hening 33.1.1. Pharmacokinetic properties (for review, see
et al., 2004). Deleu et al., 2002; Tintner and Jankovic, 2003)
Bromocriptine was the first agonist to be indicated as
an adjunct to levodopa therapy for patients with Parkin- 33.1.1.1. Absorption and bioavailability
sons disease experiencing motor fluctuations (Calne Most dopamine agonists, except apomorphine, are
et al., 1974). There are now nine different dopamine ago- absorbed orally. The systemic bioavailability of the
nists presently approved and marketed for the treatment ergot agonists is low, due to an extensive first-pass
of Parkinsons disease: apomorphine, bromocriptine, hepatic metabolism. This is also true for piribedil.
cabergoline, dihydroergocryptine, lisuride, pergolide, pir- Because of poor oral absorption and low bioavailability,
ibedil, pramipexole and ropinirole (alphabetical order). apomorphine is only used via the subcutaneous route.
Among these nine dopamine agonists, five are ergot deri- The second generation of non-ergot agonists, including
vatives (bromocriptine, cabergoline, dihydroergocryp- pramipexole and ropinirole, has a better bioavailability
tine, lisuride and pergolide) whereas the four others are (Table 33.1). The usually recommended range of daily
not (apomorphine, piribedil, pramipexole and ropinirole). doses for the different dopamine agonists is listed in
All are used as oral medications, except apomorphine, Table 33.1. Rotigotine, a dopamine agonist designed

*
Correspondence to: Professor Olivier Rascol, Clinical Investigation Center, Department of Clinical Pharmacology and
Department of Neurosciences, Faculty of Medicine, 37 Allees Jules Guesde, Toulouse 31073, France. E-mail: rascol@cict.fr,
Tel: 33-(0)-5-61-14-5962, Fax: 33-(0)-5-61-25-5116.
74 O. RASCOL ET AL.
Table 33.1
Main pharmacokinetic and pharmacodynamic (binding) properties of dopamine agonists

Binding affinity
Recommended Times Oral bioavail- Elimination
Agonists daily dose (mg) daily ability (%) half-life (h) D1-like D2-like 5-HT a1 a2

Apomorphine * * * 0.5 0/ 0/
Bromocriptine 1060 3 6 38
Cabergoline 26 1 50 65110 0/
Dihydroergo-
cryptine 30120 3 5 15 /
Lisuride 15 3 1020 23 0/
Pergolide 1.54 3 2050 20
Piribedil 150300 3 10 20 0/ 0 0/
Pramipexole 1.54.5 3 >90 812 0/ 0/ 0/
Ropinirole 624 3 50 68 0 0 0 0

*Subcutaneous route.
5-HT, serotonin.

to be administered transdermally rather than orally, is dopamine agonists are thus supposed to offer a more
currently under development for the treatment of Par- continuous stimulation of dopamine receptors than
kinsons disease (Mucke, 2003; Jenner, 2005; Poewe levodopa. At present, there is however no convincing
and Lussi, 2005; Rascol, 2005). This drug is not yet evidence that this difference in elimination half-lives
approved for this indication. correlates with specific efficacy or safety clinical
parameters. For example, empirical practice does not
33.1.1.2. Elimination half-life suggest that cabergoline, with its long elimination
Interest has focused recently in Parkinsons disease on half-life, is more efficacious in reducing the risk of
drug plasma elimination half-lives because continuous long-term motor complications than lisuride, an agonist
dopamine stimulation (CDS) is an appealing hypothesis with a much shorter elimination half life. The sole prac-
to explain the development of motor complications tical clinical difference that might be related to elimina-
(fluctuations and abnormal involuntary movements) that tion half-life is that cabergoline is used once daily to
are frequently associated with long-term dopatherapy. treat the patients, whereas the other agonists are gener-
According to this theory, levodopa, when administrated ally used on a t.i.d. regimen. In theory, it is also concei-
orally, is supposed to stimulate in a pulsatile non- vable that cabergoline-induced adverse drug reactions,
physiological way the striatal dopamine receptors. This such as psychosis for example, could persist for longer
induces in turn a cascade of molecular and cellular periods of time than with other agonists when the drug
downstream events that change the basal ganglia outflow is withdrawn. Conversely, wearing-off effects have
and lead to abnormal motor programs (Chase, 1998; Ola- been observed in some patients on monotherapy with
now et al., 2000). shorter elimination half-life agonists like ropinirole
All orally active dopamine agonists have longer (F. Stocchi, personal communication).
elimination half-lives than levodopa. Apomorphine has
the shortest one (30 minutes), but this is overcome in
clinical practice when the drug is delivered using con- 33.1.1.3. Metabolism and drug interactions (for
tinuous subcutaneous pumps (Neef and van Laar, review, see Pfeiffer, 1996)
1999; LeWitt, 2004). The shortest half-life for an orally Ergot derivatives as well as ropinirole and piribedil
active agonist is that of lisuride (26 hours). Cabergo- have an extensive hepatic metabolism. Enzymatic
line has the longest one (65110 hours) (Fariello, inhibitors, like macrolide antibacterials, have been
1998), and the other ones have intermediate half-lives, reported to increase some adverse drug reactions
ranging from 6 to 20 hours (Rascol, 1997; Matheson induced by ergot agonists like bromocriptine (Montas-
and Spencer, 2000; Di Marco et al., 2002; Biglan and truc and Rascol, 1984; Periti et al., 1992). Ropinirole
Holloway, 2002; Blin, 2003; Albanese and Colosimo, is metabolized extensively by the hepatic microsomal
2003; Curran and Perry, 2004) (Table 33.1). Globally, enzyme system (CYP1A2, CYP3A4 and CYP2D6)
DOPAMINE AGONISTS 75
(Kaye and Nicholls, 2000). Ciprofloxacin, which is an nist). Apomorphine and pergolide are known as mixed
inhibitor of CYP1A2, can increase the area under the D2- and D1-agonists. Other agonists, such as ropinirole
curve of ropinirole plasma levels. In this case, a dose and pramipexole, are reported to be selective D2-like
reduction of the agonist might be considered. Estro- (and preferentially D3-) agonists (Piercey, 1998; Tul-
gens will decrease ropinirole clearance by about loch, 1997) (Table 33.1). Such differences could theo-
36%. Conversely, pramipexole, which undergoes mini- retically have clinical correlates, but the literature is
mal hepatic biotransformation and is excreted virtually quite controversial on that topic. It has been proposed,
unchanged in the urine by the renal tubular secretion, for example, that a synergistic activation of both D1-
is not exposed to such interaction. It is devoid of and D2-receptors may be necessary to induce a full
any CYP interaction. On the other hand, this drug can antiparkinsonian effect (Luquin et al., 1992; Robert-
theoretically lead to potential interactions with drugs son, 1992). According to this theory, mixed D1/D2
excreted by renal tubular secretion (H2-antagonists, agents could then be more potent antiparkinsonian
diuretics, verapamil, quinidine) and this may require medications than the D2-selective ones. An action at
dose adjustment. The same is true in patients with D1-receptors has also been implicated in the genesis
impaired renal function. Apomorphine is partly metabo- of adverse drug reactions, like dyskinesias for exam-
lized by the catechol-O-methyltransferase (COMT) ple. Some authors have proposed that clozapine, an
enzyme (LeWitt, 2004). However, no relevant interac- atypical neuroleptic, could exert antidyskinetic proper-
tion has been reported with a COMT inhibitor such as ties because of its antagonistic effects at D1-receptors
entacapone (Durif et al., 2004). Overall, there is no (Bennett et al., 1994). This claim is however incompa-
evidence that the co-prescription of levodopa with tible with the observation that selective D2-agonists
any orally active dopamine agonist will modify the can induce dyskinesia by themselves, even in non-
pharmacokinetic properties of any of these compounds primed levodopa-naive monkeys (Gomez-Mancilla
and vice versa. and Bedard, 1992; Luquin et al., 1992) and that the
selective D1-agonist, A-86929, has a lower propensity
33.1.2. Pharmacodynamics and receptor selectivity than D2-agonists to induce abnormal movements in the
primate model of levodopa-induced dyskinesias
33.1.2.1. Binding to non-dopamine receptors
(Grondin et al., 1997). However the prodrug of this
The dopamine agonists that belong to the ergot deriva- last compound, ABT-431, induced the same amount
tive family (bromocriptine, lisuride, pergolide, cabergo- of dyskinesias as levodopa when tested in dyskinetic
line) have high to moderate affinity for a variety of patients with Parkinsons disease (Rascol et al., 2001).
non-dopaminergic receptors such as a-adrenergic (a1 Potentially beneficial effects of preferential selec-
and a2) and serotonergic (5-HT1 and 5-HT2) receptors tivity for D3-receptors have also been speculated.
(Montastruc et al., 1993; Piercey et al., 1996; Fariello, The highest concentrations of D3-receptor mRNA are
1998; Piercey, 1998). These compounds, therefore, are found in the mesolimbic pathways, which are thought
less selective than non-ergot agents like ropinirole, pra- to be involved in motivation (Wise and Rompre,
mipexole and piribedil (Tulloch, 1997; Piercey, 1998) 1989). Some psychopharmacologic experiments in
(Table 33.1). It is not clear whether such differences animals suggest that D3-agonists might have antide-
influence clinical outcomes. A drug with actions at nor- pressant properties (Maj et al., 1997). This led to the
adrenergic or serotonergic receptors might theoretically proposal that drugs like pramipexole and ropinirole
induce better antiparkinsonian efficacy than selective could have antidepressant effects, but clinical evidence
dopaminergic agents, since the transformation of levo- is still weak to support this assumption.
dopa into norepinephrine and not solely into dopamine
could explain why this drug has such potent sympto-
matic antiparkinsonian effects. However, acting on 33.1.2.3. In vivo antiparkinsonian efficacy of
non-dopamine receptors could also lead to adverse dopamine agonists in experimental models of
reactions such as psychosis or dysautonomic symptoms. Parkinsons disease
Clinical studies have not yet provided undisputable Most D2-agonists have demonstrated that they can
evidence to support these assumptions. reverse the motor deficit that mimics the motor parkinso-
nian symptoms in different in vivo models of Parkinsons
33.1.2.2. Relative affinity for dopamine-receptor disease. This is true for the akinetic syndrome induced in
subtypes the rodent by reserpine, a dopamine-depleting agent, for
Dopamine agonists differ in their relative affinities for the abnormal rotational behavior induced by the unilat-
D1- and D2-like receptors (Table 33.1). Bromocriptine eral striatal injection of 6-hydroxydopamine in the rat
is a D2-agonist and a weak D1-antagonist (partial ago- and for the parkinsonian-like motor syndrome induced
76 O. RASCOL ET AL.
by the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine strate antiapoptotic effects in a variety of cell models
(MPTP) intoxication of various species of primates (Schapira and Olanow, 2003). In the laboratory,
(Eden et al., 1991; Arai et al., 1995; Domino et al., dopamine agonists protect dopaminergic and non-
1998; Pearce et al., 1998). An intriguing finding in these dopaminergic neurons from a variety of toxins in both
animal models is that most agonists, when used as mono- in vitro and in vivo models of parkinsonism (Olanow
therapy, are as efficacious as levodopa in reversing the et al., 1998; Le and Jankovic, 2001; Schapira, 2002).
symptoms, whereas in clinical practice, it is common However, to date, none of these promising preclinical
observation that orally active agonists have a less potent data have yet translated into undisputable clinical
symptomatic antiparkinsonian activity than levodopa. benefit (see below).
The reason for this discrepancy and for the non-predict-
ability of the model on this aspect remains unknown. 33.2. Clinical trials on dopamine agonists:
It may be due to tolerability-related dose limitations clinical efficacy in the treatment of Parkinsons
in humans. disease
Dopamine agonists induce significantly fewer dys-
kinesias than levodopa in levodopa-naive MPTP- Under the auspices of the Movement Disorders Society
intoxicated monkey (Bedard et al., 1986; Grondin et al., (MDS), a group of experts that included author O. Rascol
1996; Pearce et al., 1998). This property, which is also of the present chapter has performed an evidence-based
observed in patients with Parkinsons disease, as long systematic review to assess all antiparkinsonian thera-
as they have not been previously exposed and primed peutic interventions. The conclusions of this review
by levodopa for dyskinesias, may result from the long are based on the robustness of clinical evidence (Goetz
elimination half-life of the agonists, but other mechanisms et al., 2002; Rascol et al., 2002) and have recently
could also be implicated (see section 33.1.1.2). been updated (Goetz et al., 2005). This MDS review
includes, among other interventions, the nine dopamine
agonists discussed in this chapter. It extensively sum-
33.1.2.4. Neuroprotective properties of dopamine marizes the level of evidence supporting the use of each
agonists in experimental models of neuronal death individual agonist according to a number of different
Current theories on the pathogenesis of Parkinsons clinical objectives: (1) prevention of disease progres-
disease have centered for many years on the formation sion; (2) symptomatic control of parkinsonism as mono-
of reactive oxygen species and the onset of oxidative therapy; (3) symptomatic control of parkinsonism as
stress, leading to oxidative damage in the substantia adjunct to levodopa; (4) prevention of motor complica-
nigra pars compacta (Jenner and Olanow, 1996). tions; (5) control of motor complications; and (6) man-
Dopamine may produce toxic metabolites, via auto- agement of non-motor complications, each drug being
oxidation to toxic semiquinones, that lead to the pro- rated according to standardized definitions of efficacy,
duction of reactive oxygen species (Olanow, 1990). safety and implications for clinical practice. The pre-
Levodopa decarboxylation to dopamine may enhance sent chapter will synthesize the conclusions of this
this phenomenon. For this reason, dopamine agonists, review and the reader is invited to refer to the original
which allow delay and reduction of levodopa needs, documents for a more comprehensive and detailed
have been speculated to have a positive impact on description of the methods as well as for an extensive
the disease progression. However, levodopa toxicity description and citation of the level of evidence, agonist
is highly questioned in vivo and there are no clinical by agonist and trial by trial. These data will be pre-
data to support the concept that this drug could sented here according to the use of dopamine agonists
accelerate the progression of the disease in patients in two clinical situations: (1) the early de novo patient
(Olanow et al., 2004). not previously exposed to levodopa; and (2) the patient
Independent of levodopa toxicity, there are other suffering from more advanced disease and already
reasons why a dopamine agonist might be neuropro- treated with levodopa.
tective. Activity at presynaptic dopamine autorecep-
tors might reduce the amount of dopamine released 33.2.1. Evidence for the use of dopamine agonists in
into the synapse, thus reducing the rate of firing and levodopa-naive patients with early Parkinsons
dopamine turnover in the neuron (Sethy et al., 1997). disease
Several dopamine agonists also have free radical-
scavenging activity (Yoshikawa et al., 1994; Hall There are generally three main reasons to consider
et al., 1996). In addition, they protect cells in vitro using a dopamine agonist in this situation: (1) to slow
and ex vivo from oxidative damage (Iida et al., 1999; down disease progression; (2) to control the parkinso-
Ogawa et al., 1999) and excitotoxicity and demon- nian symptoms of the patients; and (3) to reduce the
DOPAMINE AGONISTS 77
risk of motor complications associated with the long- tion of the biomarker by levodopa than by the agonists
term use of levodopa. (Schapira and Olanow, 2004).
In summary, according to these findings, neuropro-
tection remains an unmet need in Parkinsons disease.
33.2.1.1. Dopamine agonists and the prevention of In spite of a seducing rationale, there is not strong
disease progression enough evidence definitely to recommend the use of
Our basic understanding of Parkinsons disease is that a dopamine agonist for this purpose in clinical prac-
the disorder is caused by the progressive death of tice. This remains an investigational topic. Interest-
dopamine nigrostriatal neurons. It is therefore in the ingly, in line with this negative conclusion, the few
earliest stages of the disease that one expects that the available controlled data assessing mortality after
largest number of dopamine neurons remain spared 10 years of follow-up showed that the early use of an
by the pathological process and could be available agonist like bromocriptine rather than levodopa in
for neuroprotection. Consequently, neuroprotective Parkinsons disease patients did not improve patients
strategies are often discussed at this stage. life expectancy (Hely and Morris, 1999; Lees et al.,
The clinical demonstration that a drug could posi- 2001; Montastruc et al., 2001a). No data are available
tively influence the progression of Parkinsons disease with other agonists.
is a major therapeutic challenge. This objective
remains an unsolved question. Up to now, even the
33.2.1.2. Dopamine agonists as monotherapy and
design of the trials assessing the putative disease-mod-
the symptomatic control of parkinsonian symptoms
ifying effects of a given drug remains a matter of
controversies. There is no way to measure directly Parkinsonism (tremor, bradykinesia and rigidity) is the
neuronal loss in vivo and it is unclear how clinical core of the clinical syndrome of Parkinsons disease.
symptoms correlate with neuronal death. Clinical end- It progressively causes motor disability in patients
points, such as measuring the hazard function of early with early Parkinsons disease who are not already
untreated patients to reach the need for symptomatic on levodopa. A dopamine agonist used as initial mono-
treatment, are biased by the intrinsic symptomatic anti- therapy can be proposed to control such symptoms and
parkinsonian properties of many drugs, including there are a number of RCTs to support this property.
dopamine agonists. The use of imaging biomarkers Such RCTs are usually parallel short-term (6-month)
which quantify for example the amount of dopamine comparisons with placebo or an active comparator
transporters or the production of dopamine in the stria- (levodopa or another agonist). The most widely stan-
tum with single-photon or positron emission tomogra- dardized scale used to assess parkinsonism in these
phy does not allow more definite conclusions because trials is the Unified Parkinsons Disease Rating Scale
of other potential biases. (UPDRS) (Fahn et al., 1987).
Bromocriptine, pramipexole and ropinirole have
been tested in RCTs specifically designed to assess 33.2.1.2.1. Randomized controlled trials comparing
their potential impact on disease progression (Olanow agonist monotherapy versus placebo (Table 33.2)
et al., 1995; Parkinson Study Group, 2002a; Whone There is at least one good-quality RCT and sometimes
et al., 2003). Due to methodological shortcomings, several to support the efficacy of dihydroergo-
none of these studies produced sufficiently convincing cryptine (Bergamasco et al., 2000), pergolide (Barone
evidence to establish that the drugs had a positive et al., 1999), pramipexole (Shannon et al., 1997) and
effect on disease progression. It has been reported in ropinirole (Adler et al., 1997) on parkinsonism in early
two long-term (24-year) RCTs comparing pramipex- Parkinsons disease (for a complete list of references,
ole or ropinirole to levodopa that two different neuroi- see the MDS evidence-based review: Goetz et al.,
maging biomarkers of striatal dopaminergic function 2002, 2005; Rascol et al., 2002). Pergolide is no longer
declined more slowly over time in patients treated with used as a first-line agonist for safety reasons (see sec-
an agonist than in those who received levodopa. This tion 33.3). There is less convincing but still supportive
can be interpreted as an indirect index of a neuropro- evidence for bromocriptine. This conclusion is mainly
tective effect. However, in these trials, the same based on levodopa-controlled RCTs showing, as a sec-
patients had a better clinical outcome on levodopa than ondary endpoint, that the drug was as efficacious as
on agonists. Moreover, it was impossible to disentan- levodopa (Riopelle, 1987; Montastruc et al., 1994)
gle in these studies if: (1) the imaging differences were (although these studies were not powered to demon-
due to a faster decline of the biomarker on levodopa or strate non-inferiority) or only slightly less efficacious
to a slower one on the agonist; and if (2) the difference (in a proportion suggesting that the difference in
was related or not to a more pronounced downregula- functional improvement was not important enough to
78 O. RASCOL ET AL.
Table 33.2
Level of evidence supporting the efficacy of the different dopamine agonists in levodopa-naive patients with early
Parkinsons disease based on the conclusions of the Movement Disorders Society evidence-based review (see Goetz et al.,
2002, 2005; Rascol et al., 2002)

Symptomatic control Prevention of motor


Agonists Disease progression of parkinsonism complications

Apomorphine* Insufficient Not used Not used


Bromocriptine Insufficient Likely Likely
Cabergoline Insufficient Insufficient Efficacious
Dihydroer-
gocryptine Insufficient Efficacious Insufficent
Lisuride Insufficient Likely Insufficient
Pergolide{ Insufficient Efficacious Insufficient
Piribedil Insufficient Insufficient Insufficient
Pramipexole Insufficient Efficacious Efficacious
Ropinirole Insufficient Efficacious Efficacious

*Apomorphine is not used in early Parkinsons disease because it is only active via the subcutaneous route.
{
Pergolide cannot be recommended as a first-line treatment for early Parkinsons disease because of the risk of valvular heart disorder.
Efficacious, positive effect of the agonists on studied outcomes in at least one high-quality randomized controlled trial (RCT) and no
conflicting data from other RCT(s); likely, efficacy likely, i.e. evidence suggests, but is not sufficient to show, that the agonist has a positive
effect on studied outcomes, based on any RCT and no conflicting data from other RCTs; insufficient, insufficient evidence, i.e. there are no
data or available data do not provide enough evidence either for or against the use of the agonist.

suggest that the choice of the treatment was critical 2000) and ropinirole (Rascol et al., 2000). For this rea-
within the first 3 years of follow-up; Parkinsons Dis- son, after some years of treatment, most patients who
ease Research Group of the United Kingdom, 1993). start on an agonist will receive levodopa as a replacing
The same is true for cabergoline (Rinne et al., 1997, or an adjunct treatment to keep control on the motor
1998). There are only open-label studies to support parkinsonian syndrome. The optimal timing when
the effect of lisuride (Rinne, 1989) and piribedil combining both drugs has never been assessed. In the
(Rondot and Ziegler, 1992) in early Parkinsons dis- last decade, the most frequently tested strategy has
ease. Apomorphine, being only used subcutaneously, been to start with an agonist and to postpone the
has never been tested as monotherapy for the treatment adjunction of levodopa as late as possible as a second
of Parkinsons disease at this early stage. step of the therapeutic strategy. However, in the pre-
vious decade, it was common practice to combine
33.2.1.2.2. Randomized controlled trials comparing early an agonist like bromocriptine or lisuride with
agonist monotherapy versus levodopa levodopa within the first months of treatment (early
Levodopa is more efficacious than any orally active combination strategy) (Przuntek et al., 1996; Allain
dopamine agonist monotherapy. This common clinical et al., 2000). There are no data to assess if one strategy
practice observation is documented in RCTs assessing is better than the other one.
agonists like cabergoline (Rinne et al., 1997), pramipex-
ole (Parkinson Study Group, 2000), ropinirole (Rascol 33.2.1.2.3. Randomized controlled trials comparing an
et al., 1998) and bromocriptine (Parkinsons Disease agonist monotherapy versus another agonist
Research Group of the United Kingdom, 1993; Olanow There are only few trials comparing the efficacy of
et al., 1995). There are no published levodopa-controlled a dopamine agonist versus that of another one as
head-to-head RCTs with the other agonists. monotherapy. When such data are available (bromo-
The proportion of patients with early Parkinsons criptine versus ropinirole (Korczyn et al., 1998, 1999)
disease capable of remaining on agonist monotherapy and versus pergolide (Mizuno et al., 1995), the clinical
falls progressively over years to less than 20% after relevancy of the reported difference, if any, remains
5 years of treatment with bromocriptine (Parkinsons questionable, especially since the exact dose equiva-
Disease Research Group of the United Kingdom, lence between the different agonists remains unknown.
1993; Montastruc et al., 1994), cabergoline (Rinne Such drugs should then be considered from a clinical
et al., 1998), pramipexole (Parkinson Study Group, perspective as having basically a comparable efficacy.
DOPAMINE AGONISTS 79
33.2.1.2.4. Randomized controlled trials comparing an (dihydroergocryptine, pergolide, piribedil), apomor-
agonist monotherapy versus another antiparkinsonian phine not having been used and tested in this situation.
medication The ability of several dopamine agonists to reduce
There are no published direct head-to-head compari- or to delay time to motor complications versus levo-
sons between any agonist monotherapy and other anti- dopa is therefore based on a good level of evidence.
parkinsonian medications in early Parkinsons disease However, the clinical importance and usefulness of
(monoamine oxidase-B (MAO-B) inhibitors, amanta- such results in the global management strategy of
dine, anticholinergics). The changes in UPDRS scores patients with early Parkinsons disease remain a matter
reported in placebo-controlled RCTs are usually of controversy (see section 33.4).
greater on agonists than on MAO-B inhibitors (Par-
kinson Study Group, 1989, 2002b). This could be 33.2.1.3.2. Randomized controlled trials comparing
interpreted as an indirect indication of a greater symp- agonist monotherapy versus another agonist
tomatic efficacy of the agonists, but one cannot firmly There is no indication that one agonist might be more
establish if this putative difference is of clinical efficacious than another one in preventing or delaying
importance. time to motor complications. The only published head-
to-head RCT comparing two agonists in this situation
33.2.1.3. Prevention of motor complications (ropinirole versus bromocriptine) (Korczyn et al.,
The incidence of motor complications is 10% per year 1999) did not show any difference at 3 years in the
on levodopa therapy. Over time, they usually become incidence of dyskinesias.
more disabling and difficult to manage. Symptomatic
control of symptoms is therefore not the only factor 33.2.1.3.3. Randomized controlled trials with
to be considered when initiating an antiparkinsonian agonist monotherapy versus other antiparkinsonian
treatment and doctors and patients are also often inter- medications
ested in considering the impact of an initial therapeutic No data are available to assess whether the early use of
strategy on the incidence of long-term motor compli- an agonist could be more efficacious in delaying time
cations. The strategy of using early a dopamine agonist to motor complications than other drugs like MAO-B
and to keep levodopa as a second-line rescue medica- inhibitors, anticholinergics, amantadine or the early
tion to delay the problem of motor complication has combination of a COMT inhibitor with levodopa. Such
been a growing matter of interest in the last decade. trials should be performed in the future.
Several RCTs compared the probability of developing
motor complications for up to 5 years in previously 33.2.2. Evidence to use dopamine agonists as
untreated patients who received an agonist versus adjunct to levodopa in patients with moderate to
levodopa-treated patients. There was no standardized advanced Parkinsons disease
method used to define when a patient began motor
complications and each trial used its own definition, Adding a dopamine agonist in patients already on
with consequent variable numbers from one study to levodopa therapy is a strategy that is generally consid-
another. ered and has been assessed for three main reasons:
(1) to improve the symptomatic control of parkinso-
33.2.1.3.1. Randomized controlled trials comparing an nian motor signs (parkinsonism); (2) to improve motor
agonist monotherapy versus levodopa (Table 33.2) complications; and (3) to try to improve non-motor
Prospective RCTs lasting 25 years and demonstrating signs.
the ability of the early use of an agonist to reduce the
incidence of motor complications versus levodopa are 33.2.2.1. Symptomatic control of parkinsonism as
available for cabergoline (Rinne et al., 1998), prami- adjunct to levodopa
pexole (Parkinson Study Group, 2000) and ropinirole The symptomatic efficacy of several agonists has been
(Rascol et al., 2000; Whone et al., 2003). Similar con- assessed in RCTs in patients with moderate to severe
clusions were reported according to less methodologi- forms of Parkinsons disease already treated with levo-
cally convincing trials with bromocriptine (Parkinson dopa. As for early monotherapy, such RCTs are
Diseases Research Group of the United Kingdom, usually limited to 36 months of follow-up and use
1993; Hely et al., 1994; Montastruc et al., 1994). Con- the UPDRS as an endpoint to assess the symptomatic
flicting results have been reported with lisuride effect of the drug on the cardinal signs of parkinson-
(Rinne, 1989; Allain et al., 2000) and published con- ism, in the off and/or in the on condition if the
trolled data are lacking for other orally active agonists patients are fluctuating.
80 O. RASCOL ET AL.
33.2.2.1.1. Randomized controlled trials comparing an strong impact on clinical practice because most studies
agonist versus placebo (Table 33.3) raised methodological problems (insufficient power,
Most agonists have shown to be effective in improving surrogate endpoints, arbitrary definition of dose
the cardinal motor signs of parkinsonism in patients equivalences) and because they reported only mod-
already treated with levodopa. This is true for apomor- est intergroup differences, posing the question of
phine (Dewey et al., 2001), bromocriptine (Guttman their relevance in clinical practice. This applies to
et al., 1997; Mizuno et al., 2003), cabergoline (Hutton trials with cabergoline (Inzelberg et al., 1996), lisur-
et al., 1996), pergolide (Olanow et al., 1994), piribedil ide (LeWitt et al., 1982; Laihinen et al., 1992),
(Ziegler et al., 2003) and pramipexole (Pinter et al., pergolide (LeWitt et al., 1983; Pezzoli et al., 1994;
1999; Pogarell et al., 2002). Due to lower-quality Mizuno et al., 1995; Boas et al., 1996), pramipexole
trials, the available evidence is less convincing but still (Mizuno et al., 2003) and ropinirole (Brunt et al., 2002).
supportive for dihydroergocryptine (Martignoni et al.,
1991), lisuride (Allain et al., 2000) and ropinirole 33.2.2.1.3. Randomized controlled trials comparing an
(Lieberman et al., 1998). Some trials suggested that agonist versus other antiparkinsonian medications
agonists such as pramipexole and ropinirole might Bromocriptine (Tolcapone Study Group, 1999) and per-
have a special impact on tremor (Pogarell et al., golide (Koller et al., 2001) have been the sole agonists
2002; Schrag et al., 2002), but there is no clear ratio- compared to the COMT inhibitor tolcapone in open-
nale for such a specific effect and this can probably label RCTs. No significant difference was reported in
apply to any efficacious symptomatic dopaminergic terms of efficacy on the parkinsonian cardinal signs,
medication (Navan et al., 2005). although the safety profile was somehow different
between the drugs. No data are available for other
33.2.2.1.2. Randomized controlled trials comparing an agonists and other antiparkinsonian drugs.
agonist versus another agonist
There are a number of RCTs of variable methodologi- 33.2.2.2. Symptomatic treatment of motor
cal quality that compare in cross-over or parallel complications
designs the symptomatic effect of two different dopa- Motor complications are frequent and can be disabling
mine agonists on parkinsonism when adjunct to levo- after several years of treatment with levodopa. They
dopa. In all these trials, bromocriptine has been used involve fluctuations, erratic or unstable responses to
as the reference comparator. Such data cannot have a medications, also known as the wearing-off or onoff

Table 33.3
Level of evidence supporting the efficacy of the different dopamine agonists as an adjunct to levodopa in moderate to
advanced Parkinsons disease according to the conclusions of the evidence-based review on antiparkinsonian
interventions (Goetz et al., 2002; Rascol et al., 2002)

Symptomatic control Control of motor Non-motor


Agonists of parkinsonism fluctuations symptoms

Apomorphine* Efficacious Efficacious Insufficient


Bromocriptine Efficacious Likely Insufficient
Cabergoline Efficacious Likely Insufficient
Dihydroergocryptine Insufficient Insufficient Insufficient
Lisuride Likely Insufficient Insufficient
Pergolide{ Efficacious Efficacious Insufficient
Piribedil Efficacious Insufficient Insufficient
Pramipexole Efficacious Efficacious Insufficient
Ropinirole Insufficient Efficacious Insufficient

*Apomorphine is used as subcutaneous route.


{
Pergolide cannot be recommended as a first-line treatment for Parkinsons disease because of the risk of valvular heart disorder.
Efficacious, positive effect of the agonists on studied outcomes in at least one high-quality randomized controlled trial (RCT) and no conflict-
ing data from other RCT(s); likely, efficacy likely, i.e. evidence suggests, but is not sufficient to show, that the agonist has a positive effect on
studied outcomes, based on any RCT and no conflicting data from other RCTs; insufficient, insufficient evidence, i.e. there are no data or avail-
able data and do not provide enough evidence either for or against the use of the agonist.
DOPAMINE AGONISTS 81
phenomena, and dyskinesias or involuntary move- of the duration and severity of levodopa-induced dys-
ments. Completing dopamine stimulation with drugs kinesias, since agonists could deliver a more stable
like dopamine agonists is expected to be potentially stimulation than levodopa and therefore reverse the
useful for such symptoms. RCTs have thus been underlying mechanisms causing dyskinesias. There
implemented to assess the effects of various agonists are only few open-label and generally uncontrolled
on motor complications. Such trials were usually reports in small cohorts of patients to support this
short-term (36-month) studies using home diaries as practice. The most convincing data have been
an endpoint (Hausser et al., 2000). Because agonists published with continuous subcutaneous infusions of
have differing effects on fluctuations and dyskinesias, apomorphine (Colzi et al., 1998; Manson et al., 2002;
these behaviors are considered separately. Katzenschlager et al., 2005). Some positive anecdotal
studies have also been presented with oral administra-
33.2.2.2.1. Motor fluctuations tion of pergolide (Facca and Sanchez-Ralos, 1996) and
ropinirole (Cristina et al., 2003).
There are several good-quality RCTs to support the
use of a number of dopamine agonists to reduce the
duration of off episodes (Table 33.3). This is true for 33.2.2.3. Symptomatic control of non-motor
pergolide (Olanow et al., 1994; Clarke and Speller, problems
2000a), pramipexole (Guttman et al., 1997; Clarke
Although Parkinsons disease is often considered a pro-
et al., 2000a; Mizuno et al., 2003), ropinirole (Rascol
totypic movement disorder, most patients have addi-
et al., 1996; Lieberman et al., 1998; Clarke and Deane,
tional non-motor symptoms. These include autonomic
2001a) and apomorphine (Ostergaard et al., 1995; Dewey
dysfunction, depression, cognitive decline, sleep pro-
et al., 2001). For safety reasons (see section 33.3), it is
blems, sensory complaints and pain. Available trials
not recommended to use pergolide as a first-line agonist.
on non-motor features of Parkinsons disease, when
Less convincing RCTs or less consistent results have
available, usually have major methodological limita-
been reported with bromocriptine (Hoehn and Elton,
tions. As a result, the evidence is weak in most
1985; Toyokura et al., 1985; Guttman et al., 1997) and
instances and does not allow the use of an agonist to
cabergoline (Hutton et al., 1996). There are only
be supported or recommended in such indications.
open-label or anecdotal data to suggest that other ago-
In RCTs conducted in non-parkinsonian subjects
nists like dihydroergocriptine, lisuride or piribedil can
with major or bipolar depression, pramipexole was
also improve motor fluctuations.
superior to placebo (Corrigan et al., 2000; Zarate
Comparative trials, when available, did not show
et al., 2004), but convincing data are missing in Parkin-
major differences between bromocriptine and other
sons disease patients. The only available evidence in
agonists such as cabergoline (Inzelberg et al., 1996),
Parkinsons disease is weak, limited to uncontrolled or
lisuride (Laihinen et al., 1992), pergolide (Mizuno et al.,
low-quality RCT trials with agonists like pergolide, pra-
1995; Clarke and Speller, 2000b) ropinirole (Clarke and
mipexole and ropinirole (Izumi et al., 2000; Perugi
Deane, 2001b; Brunt et al., 2002) and pramipexole
et al., 2001; Lattanzi et al., 2002; Rektorova et al.,
(Clarke et al., 2000b; Mizuno et al., 2003). The same
2003). This does not allow any firm conclusions to be
was true when comparing bromocriptine (Tolcapone
drawn.
Study Group, 1999) and pergolide (Koller et al., 2001)
The same is true for cognition, although it is com-
with the COMT inhibitor tolcapone. No other compari-
mon empirical knowledge that any agonist can aggra-
sons have been published.
vate cognitive and behavioral dysfunction in patients
with Parkinsons disease dementia or Lewy body dis-
33.2.2.2.2. Dyskinesias ease. Agonists are therefore not recommended or should
When levodopa-treated patients with advanced Parkin- even be withdrawn in such conditions. There is no
sons disease receive an agonist to reduce off episodes, strong evidence that symptoms like frozen gait, balance
this is usually at the cost of some worsening of dyskine- problems, anxiety, sleep disturbances or pain are
sia. This has been quite constantly observed in any RCT responsive to the effects of dopamine agonists. It is con-
assessing any agonist in this situation (see previous ceivable that such symptoms, if partly dopa-responsive
paragraph for references). In clinical practice, the daily and occurring or worsening during off episodes, might
dose of levodopa is then partly reduced when the be improved by such drugs as by any other dopaminer-
agonist is adjunct in order to minimize this problem. gic medication. No convincing undisputable clinical
On longer follow-up, in line with the CDS hypoth- data are available however. Conversely, dysautonomic
esis, there are theoretical reasons to expect that high parkinsonian symptoms, like orthostatic hypotension,
doses of dopamine agonists might permit the reduction are often aggravated by dopamine agonist (as by any
82 O. RASCOL ET AL.
dopaminergic medication), probably through dopami- although it is frequently empirically believed that this
nergic sympatholytic mechanisms. is the case in clinical practice.
Leg edema is known to be associated with both
33.3. Safety issues on dopamine agonists ergot and non-ergot dopamine agonists. Reported inci-
dences vary widely and this reaction is more frequent
Adverse drug reactions are defined as any appreciably on agonists than on levodopa. For example, the inci-
harmful or unpleasant reaction resulting from an inter- dence of leg edema reported in levodopa-controlled
vention related to the use of a medicinal product which trials was 14% on ropinirole versus 6% on levodopa
predicts hazard from future administration and war- (Rascol et al., 2000), 42% on pramipexole versus
rants prevention or specific treatment, or alteration of 15% on levodopa (Holloway et al., 2004) and 16%
the dosage regime, or withdrawal of the product on cabergoline versus 3% on levodopa (Bracco et al.,
(Edwards and Aronson, 2000). They can be classified 2004). The mechanism of this side-effect is unknown.
as type A and type B reactions (Rawlins and Thomson, This adverse drug reaction is sometimes reported as
1977). Type A reactions are generally expected and dose-dependent but can also be idiosyncratic. It is
predictable, since they are frequently related to the reversible with discontinuation of the agonist and the
known pharmacological properties of the drug. Their use of a diuretic therapy should be discouraged because
prevalence is relatively high. They are usually non- it is not efficacious and can induce or aggravate other
serious, non-lethal, dose-dependent and can be managed problems like orthostatic hypotension.
with dose adjustment. Conversely, type B reactions are Subcutaneous nodules at the injection site of apo-
unexpected, unpredictable, rare, often serious reactions morphine are another type A adverse reaction. This
and most of the time they lead to drug withdrawal. adverse reaction seems unrelated to the dopaminergic
effects of the drug but rather to its intrinsic physical
properties, since problems of local toxicity have been
33.3.1. Dopamine agonist type A adverse reactions
reported at any site of administration (subcutaneous,
sublingual, intranasal). Such subcutaneous nodules
Such adverse reactions are usually classified as periph-
are frequent, sometimes painful and may become
eral (gastrointestinal reactions such as nausea and
infected, especially when the drug is delivered by
vomiting, cardiovascular reactions such as orthostatic
hypotension and leg edema) or central ones (psychia- subcutaneous continuous pump. Changing the site of
injection every day and careful local hygiene reduce
tric reactions and sedative reactions). These type A
the incidence and severity of such a reaction (Poewe
reactions are shared by other active dopamine-mimetic
et al., 1993; Bowron, 2004).
medications and are generally believed to be the con-
sequence of the stimulation of dopamine receptors
out of the basal ganglia. There is no convincing evi- 33.3.1.2. Central type A adverse reactions
dence that any agonist is associated with a lower risk
Psychiatric symptoms are among the most disabling
than another one for such type A reactions and this is
true for peripheral as well as central effects. dopamine agonist-related adverse drug reactions
(Factor et al., 1995). This is true for any efficacious
dopaminergic medications. The prevalence of these
33.3.1.1. Peripheral type A adverse reactions symptoms on agonists varies according to the reports
Gastrointestinal and cardiovascular adverse reactions and usually ranges between 5 and 20%. In most
are common at the onset of therapy with any dopamine levodopa-controlled double-blind trials, psychiatric
agonist and usually tolerate out over time. They are adverse reactions are reported to be more common
explained by the agonistic effect of the drugs on dopa- on agonists than on levodopa (Rascol et al., 2000;
mine receptors in the gut and in the area postrema Parkinson Study Group, 2000) and this is also empiri-
(gastrointestinal effects) and at the presynaptic level cally observed in clinical practice. Such psychotic
of the orthosympathetic system (orthostatic hypoten- symptoms can present as delusions, usually paranoid,
sion). They can be managed by slow titration. Anti- or hallucinations, predominantly visual, though audi-
emetics like domperidone, a dopamine-blocker agent tory component is common. Abnormal behaviors such
that does not cross the bloodbrain barrier, help to as hypersexuality, pathological gambling and other
treat nausea in countries where this drug is available. related syndromes have also been reported. Such psy-
In large levodopa-controlled double-blind RCTs, gas- chiatric reactions generally develop after a period of
trointestinal and cardiovascular adverse drug reactions treatment and are dose-related. Predisposing factors
are not necessarily reported to be more frequent on such as age, individual psychopathological back-
agonists than on levodopa (Rascol et al., 2000), ground, cognitive impairment and polytherapy are
DOPAMINE AGONISTS 83
sometimes reported. These adverse reactions are a fre- unintended sleep episodes, irresistible sleep
quent reason for withdrawing an agonist after some episodes, abnormal daytime sleepiness, excessive
years of therapy. The development of such psychiatric daytime sleepiness), different populations (non-
reactions might indeed represent an early indicator of demented parkinsonians, parkinsonians on agonists
cognitive decline in such a patient, although hallucina- only, driving parkinsonians, normal population) and
tions and delusion can occur in cognitively normal different methods of assessment (questionnaires,
subjects. The usual management of such psychiatric scales like the Epworth scale, electrophysiological
adverse reactions is to reduce the dose or even stop the techniques like the Multiple Sleep Latency Test)
agonist. It may take weeks before the effect clears. according to the studies. Initially, it has been difficult
If downtitration or withdrawal of the agonist leads to an to demonstrate the exact responsibility of the agonists
unacceptable worsening of motor function, the atypical in causing such symptoms because multiple factors
neuroleptic clozapine proved to improve hallucinations can induce daytime somnolence in patients with Par-
without worsening parkinsonism in two short-term pla- kinsons disease. These factors are mainly related to
cebo-controlled RCTs (French Clozapine Parkinson the disease itself, comorbidities and comedications.
Study Group, 1999; Parkinson Study Group, 1999). In placebo-controlled RCTs conducted in healthy
However, due to safety hematological problems, cloza- volunteers, an agonist like ropinirole induced somnolence
pine is not easy to use and other atypical neuroleptics, (Ferreira et al., 2002). This was also true for levodopa
like quetiapine, for example, are sometimes empirically (Andreu et al., 1999). These experiments strongly
proposed, although convincing RCTs are not available document the link between the drugs and the sedative
to provide a good level of evidence to support this prac- symptoms, regardless of the disease itself. Risk fac-
tice (Matheson and Lamb, 2000). tors are poorly known and may be related to the
Abnormal daytime somnolence was not listed patient (age, gender, genetic susceptibility), the dis-
among common type A adverse reactions of antiparkin- ease (severity, duration, associated symptoms such as
sonian medications like dopamine agonists until the dysautonomia, dementia, depression) or the drug
publication in 1999 of cases of sleep attacks at the (dose, duration of exposure, comedications). It must
wheel in patients with Parkinsons disease treated with now be recognized that somnolence is a frequent
ropinirole or pramipexole (Frucht et al., 1999). It was dopaminergic type A adverse reaction, shared by
thereafter realized that such problems are common with most, if not all, dopaminergic medications (Homann
any dopamine agonist, including ergot and non-ergot et al., 2002), although dopamine agonists appear to
ones. This is based on postmarketing surveillance case be at greater risk than levodopa (Etminan et al.,
reports with apomorphine (Homann et al., 2000), 2001). Patients should be informed of this potential
bromocriptine (Ferreira et al., 2000), cabergoline effect and be advised not to drive if it is present.
(Ebersbach et al., 2000), lisuride (Ferreira et al., The practical management of agonist-induced day-
2000), pergolide (Ferreira et al., 2000; Schapira, time inappropriate sleepiness is poorly known and
2000), piribedil (Ferreira et al., 2000; Tan, 2003), remains empirical. In some instances, dose reduction
pramipexole (Frucht et al., 1999; Ryan et al., 2000), is efficacious. Switching from one agonist to another
ropinirole (Frucht et al., 1999; Paladini, 2000) and can be proposed, but the same reaction can occur
other antiparkinsonian medications than agonists, like with the other agonist. There are no convincing pla-
entacapone (Ebersbach et al., 2000) and levodopa cebo-controlled RCTs on which to base the prescrip-
(Ferreira et al., 2001). Pharmacoepidemiological tion of specific therapies, such as modafinil for
surveys confirmed that all agonists share the risk of example (Montastruc and Rascol, 2001).
daytime somnolence (Hobson et al., 2002; Paus
et al., 2003; Ferreira et al., 2006). In levodopa-con- 33.3.2. Dopamine agonist type B adverse reactions
trolled double-blind RCTs, the agonists usually
induce greater somnolence than levodopa (Etminan Examples of rare but potentially severe type B reac-
et al., 2001). The prevalence of abnormal daytime tions on dopamine agonists are numerous.
somnolence varies greatly according to the studies Fibrosis has been detected in postmarketing surveil-
(6% according to Paus et al., 2003; 16%: Tandberg lance studies with all ergot derivatives, including bromo-
et al., 1999; 18%: Razmy et al., 2004; 27%: Ferreira criptine, lisuride, cabergoline and pergolide. Classically,
et al., 2006; 30%: Montastruc et al., 2001b; 32%: such fibrotic reactions have been reported as pleuropul-
Manni et al., 2004; 34%: Schlesinger and Ravin, monary, pericardiac and/or retroperitoneal syndromes
2003; 43%: Korner et al., 2004; 76%: Brodsky (Todman et al., 1990; Bhatt et al., 1991; Ling et al.,
et al., 2003). This is probably mainly due to different 1999; Shaunak et al., 1999; Ben-Noun, 2000; Mondal
definitions (sleep attacks, sudden onset of sleep, and Suri, 2000). These adverse reactions are usually
84 O. RASCOL ET AL.
considered as rare events, although their exact preva- and surgical treatment options that can be offered to
lence is unknown. Their mechanism is possibly related patients. There are different ways to introduce and
to the dose and duration of exposure to the ergot combine antiparkinsonian medications, according to
compounds. Erythrocyte sedimentation and protein C- various therapeutic strategies. Regardless of these dif-
reactive inflammatory markers are usually increased ferences, the global usefulness of dopamine agonists is
and may be helpful for an early diagnosis. The risk of well established and there is a good level of evidence
developing such a fibrosis on non-ergot agonists is much to document that this class of drugs, as a whole, pro-
lower, if it exists. These reactions are sometimes vides substantial help in the management of patients
life-threatening, although they can be at least partially with Parkinsons disease. Nevertheless, the exact place
reversible after withdrawal of the drug. In general, of the dopamine agonists, as a class, versus other
when a patient develops this kind of adverse reaction medications and the best choice among the different
on an ergot agonist, the strategy is to switch to a agonists within this class (except subcutaneous
non-ergot one. injections of apomorphine) often remains a matter
Recently, several cases of another type of fibrotic of empirical preferences that differ according to
adverse reaction, severe multivalvular heart disease, individual opinions.
have been reported on pergolide and other ergot deri- In de novo patients, the main question is to decide
vative agonists, namely bromocriptine and cabergoline if and when to start levodopa. Should levodopa be
(Vergeret et al., 1984; Agarwal et al., 2004; Horvath used as soon as possible or as a second step, after an
et al., 2004). For the moment, the risk is better docu- initial dopamine agonist early therapy? For each
mented with pergolide (Van Camp et al., 2004). For patient, the choice is based on a subtle combination
this reason, this drug is now generally only used as a of subjective and objective factors, including concerns
second-line alternative option, when other agonists, related to the drug (efficacy, safety, practicality, cost),
especially the non-ergot ones, have not provided a the patient (symptoms, age, needs, expectations,
satisfying response. Operational definitions (clinical experience, comorbidity, socioeconomic level) and
syndrome versus echocardiographic detection), sever- his/her environment (variability of drugs availability
ity grading, prevalence, mechanism of action (related according to national markets, variability in econom-
for some authors to 5-HT2B effects), management ical and health care systems). There is thus not a single
and reversibility after drug withdrawal of such valvu- and unequivocal algorithm applicable to all patients
lar heart reactions remain a matter of debate and it is worldwide (Clarke and Guttman, 2002; Ahlskog,
too early at this stage to conclude definitely on their 2003; Schwarz, 2003).
true impact on the benefit/risk ratio of the ergot Starting a patient on one of the available orally
agonists (Rascol et al., 2004a, b). active dopamine agonists is now considered as a useful
Another example of type B adverse reaction is strategy by a growing number of specialists, because
Coombs-positive hemolytic anemia that has been agonists are effective antiparkinsonian medications
reported with apomorphine, especially in patients and are associated with a lower risk of motor compli-
receiving continuous subcutaneous infusions. This is a cations than levodopa. The benefit of this strategy
rare adverse reaction and the usefulness of blood count must be balanced however by the fact that: (1) the ben-
monitoring for this purpose remains uncertain (Poewe efit of the early use of an agonist on motor complica-
et al., 1993; Pinter et al., 1998). Other examples of type tions has been assessed only up to 5 years and
B reactions due to agonists are alopecia, which has been remains questionable on longer follow-up, especially
reported with bromocriptine (Fabre et al., 1993), visual in terms of quality of life; (2) agonists have a smaller
cortical disturbances on bromocriptine (Lane and symptomatic effect than levodopa on UPDRS scores
Routledge, 1983), loss of vision color on pramipexole according to RCTs (but the difference is only marginal
(Muller et al., 2003) and hypersensitivity (allergic and probably clinically not relevant when small doses
reactions) to many agonists (Martindale, 2002). These of levodopa are allowed as a supplement as soon as
adverse reactions are rare, but their real prevalence the agonist monotherapy becomes insufficient);
and mechanisms often remain unknown. (3) agonists are more expensive than levodopa; and
(4) there is a greater incidence of psychiatric adverse
33.4. Dopamine agonists in clinical practice reactions, somnolence and edema on agonists than on
and their place in the management of patients levodopa. Patients must be informed of these risks,
with Parkinsons disease especially somnolence for drivers. As older patients
(70 years of age and above) are less prone to develop
Treatment of Parkinsons disease has become increas- motor complications on levodopa and more sensitive
ingly complex, due to the growing number of medical to cognitive and psychotic adverse reactions, the
DOPAMINE AGONISTS 85
early use of levodopa rather than that of an agonist is proposed to treat off episodes (Factor, 2004). The clin-
often preferred as first-line therapy in this population. ical effect of a single apomorphine injection usually
Conversely, as younger patients (< 70 years) are at develops within 10 minutes and lasts 45 minutes on
greater risk for motor complications, this is the popula- average, because of the short elimination half-life of
tion where initial treatment with an agonist is the drug. The concomitant use of domperidone, where
especially considered. In most patients, agonist mono- available, reduces the risk of gastrointestinal and
therapy cannot control parkinsonian symptoms for hypotensive adverse reactions. Apomorphine penjet
more than a few years, even when increasing the injections are recommended in patients with severe
dose over time, and must thereafter be complemen- fluctuations resistant to other medical therapies, on
ted with levodopa. In this case, it is recommended top of the regular oral treatment (including an oral
to continue agonist therapy and to supplement it with agonist). When the need for apomorphine injections
the lowest possible dose of levodopa rather than to exceeds 810 injections per day, or when dyskinesias
switch to levodopa monotherapy in order to maintain are too difficult to manage, continuous subcutaneous
as much as possible the long-term benefit on motor infusion with apomorphine pumps is then considered.
complications. This requires a clinical team with sufficient expertise
In more advanced levodopa-treated patients, the in order to train the patient and the care-giver correctly
usefulness of dopamine agonists is established to treat to a relatively complex procedure. A proportion of dif-
onoff problems. Clinical empirical experience and ficult patients who failed on oral medications will
some comparative studies suggest that the adjunction profit from this treatment, with a reduction of time
of an orally active dopamine agonist to levodopa spent off, a reduction in dyskinesia and a reduction
reduces the time spent off in a comparable way than of the daily dose of oral therapies, including levodopa
other pharmacological options such as COMT or (Hagell and Odin, 2001). In the centers where deep
MAO-B inhibitors. On average, there is a 12-hour brain (subthalamic stimulation) surgery is available,
reduction in time spent off, at the cost of some worsen- these patients are nowadays frequently offered a surgi-
ing of dyskinesias that can be managed by adjusting cal approach and apomorphine pumps are rather pro-
concomitantly the dose of levodopa. Interestingly, the posed as a second-line alternative option, in cases of
efficacy of the three different types of adjunct medica- surgical contraindication. However, the benefit/risk
tions to levodopa (agonists, MAO-B and COMT inhi- and cost/effectiveness ratios of both techniques have
bitors) does not seem to be mutually exclusive and never been carefully compared in parallel and this
rather appears to be complementary. Therefore, many should be assessed objectively in the future.
patients with advanced Parkinsons disease will hap- Another important matter of interest for clinical
pen to receive a combination of these drugs after some practice is to compare the advantages and disadvan-
years of management. Starting with one of them first, tages of the different orally active agonists (Tan and
rather than with one of the others, remains a matter Jankovic, 2001). Four aspects can help in comparing
of personal experience and preference. MAO-B inhibi- drugs: efficacy, safety, practicality and costs. Overall,
tors have the advantage of practicality (no titration, excluding apomorphine, all orally active dopamine
once-daily dose and only one dosage for all patients). agonists share the same efficacy profile (global class
COMT inhibitors do not require titration either and effect) (Bonuccelli, 2003; Inzelberg et al., 2003).
are used at a unique dosage. Conversely, dopamine However, the level of evidence differs from one drug
agonists must be titrated but their dosage can then be to the other, because the effect of some agonists has
tailored to individual need. Safety profiles are also dif- been poorly or never assessed in several indications
ferent between the three therapeutic classes of comple- (Tables 33.2 and 33.3). The best level of evidence is
mentary drugs to levodopa. A special place must be clearly offered by the two most recently marketed ago-
allocated to apomorphine in the management of nists, pramipexole and ropinirole. There is however lit-
advanced patients with Parkinsons disease (Poewe tle evidence that an older agonist, like bromocriptine,
and Wenning, 2000; Deleu et al., 2004). This is so is less useful than the newer ones. The few trials iden-
because of the effect size of apomorphine on parkinso- tified with older medications were conducted in times
nian symptoms. It is strong, similar to that of levo- when technical solutions to plan such trials had not yet
dopa, and this is unique for a dopamine agonist. The been developed. This historical factor explains why
group of Lees and coworkers first reported the benefi- such trials are less convincing. Unfortunately, there is
cial effects of apomorphine in patients with advanced no present interest in studying these old drugs again,
Parkinsons disease (Stibe et al., 1988). As already using modern designs, since their patents have expired
mentioned, this drug can only be used via subcuta- and these molecules thus do not offer any more suffi-
neous route. Intermittent penjet injections can thus be cient economical retrieval. As pointed out above, the
86 O. RASCOL ET AL.
evidence to document a substantial superiority of the same family frequently increases the risk of adverse
new agonists over bromocriptine turns out to be rather reactions.
weak and poorly convincing. In several countries, bro-
mocriptine is much cheaper than the newer agonists, 33.5. Conclusions
especially now that generics are available, and this
deserves to be considered. Safety is another important In summary, dopamine agonists are useful medications
matter. Overall, all agonists expose the patients to the for the management of patients with Parkinsons dis-
same risk of peripheral and central type A adverse ease in the early stages as well as in more advanced
reactions. There is no evidence that one of them is stages of the disorder. Although their symptomatic
safer than another one on these aspects. Conversely, effect is well established on the cardinal signs of par-
fibrotic type B reactions appear to be more frequent kinsonism and on motor fluctuations, their clinical
on ergot than non-ergot agonists and this is why pergo- impact on disease progression and on non-motor signs
lide, for example, is no longer considered as a first-line such as depression remains to be documented. All
choice when starting a treatment with a dopamine ago- orally active dopamine agonists have globally quite a
nist. From a practicality perspective, all orally active comparable benefit/risk profile, although: (1) cabergo-
agonists are usually administered t.i.d. except cabergo- line can be used once daily whereas others are usually
line, which is administrated once daily. All must be prescribed t.i.d.; (2) apomorhine is used subcuta-
titrated over weeks up to the minimal dose that is suf- neously in patients with severe refractory motor fluc-
ficient to control symptoms. The impracticality of apo- tuation; and (3) ergot compounds are at greater risk
morphine subcutaneous injections precludes the use of than the non-ergot compounds for rare but potentially
this drug in patients with early Parkinsons disease and severe fibrotic complications. Future developments
limits its indications to patients with severe levodopa- for dopamine agonists in the treatment of Parkinsons
induced motor complications. disease might come from new compounds having
Switching from an agonist to another one for effi- novel pharmacokinetic profiles, such as the transder-
cacy or safety reasons is sometimes considered in clin- mal patch of rotigotine (Rascol, 2005) and the slow-
ical practice, although global efficacy and safety release formulations of already marketed agonists like
profiles (except fibrosis) are similar among the drugs. ropinirole, or from new agonists with novel pharmaco-
Most of the available data is based on open-label dynamic profiles (more selective on dopamine D1-like
non-randomized trials with an overnight switch from or D2-like receptor subtypes) and partial agonists such
bromocriptine to the other agonist. An empirical as SLV 308 (Wolf, 2003). In the next few years,
conversion chart of dose equivalence is sometimes the clinical use of dopamine agonists will extend to
proposed with 10 mg bromocriptine 1 mg pergolide novel indications in disorders with greater prevalence
1 mg pramipexole 2 mg cabergoline 5 mg ropi- than Parkinsons disease, such as the restless-legs
nirole (Reichmann et al., 2003; Grosset et al., 2004; syndrome, for example.
Stewart et al., 2004). Using such equivalent daily
doses, some patients appear to enjoy or not switch- References
ing from bromocriptine to another agonist. This might
be related to the placebo or nocebo effect, but this
Adler CH, Sethi KD, Hauser RA et al. (1997). for the Ropi-
strategy is sometimes interesting in the practical man- nirole Study Group. Ropinirole for the treatment of early
agement of a given patient. Combining two dopamine Parkinsons disease. Neurology 49: 393399.
agonists is another issue that is sometimes considered Agarwal P, Fahn S, Frucht SJ (2004). The diagnosis and
in clinical practice. There is no real convincing phar- management of pergolide-induced fibrosis. Mov Disord
macological rationale to combine in the same subject 19: 699704.
two different orally active dopamine agonists, which Ahlskog JE (2003). Parkinsons disease: is the initial treat-
are supposed to act on the same D2-receptors. How- ment established? Curr Neurol Neurosci Rep 3: 289295.
ever, some movement disorder specialists believe that Albanese A, Colosimo C (2003). Dihydroergocriptine in Par-
such a combination regimen (for example, an agonist kinsons disease: clinical efficacy and comparison with other
dopamine agonists. Acta Neurol Scand 107: 349355.
with a long elimination half-life, like cabergoline a
Allain H, Destee A, Petit H et al. (2000), and the French
non-ergot agonist with a shorter elimination half-life,
Lisuride Study Group. Five-year follow-up of early lisur-
like ropinirole or pramipexole) is useful in some ide and levodopa combination therapy versus levodopa
patients. Such a practice warrants more studies and monotherapy in de novo Parkinosns disease. Eur Neurol
a more objective evaluation before any conclusions 44: 2230.
can be drawn. It is common pharmacological observa- Andreu N, Chale JJ, Senard JM et al. (1999), L-dopa-induced
tion that combining two compounds belonging to the sedation: a double-blind cross-over controlled study versus
DOPAMINE AGONISTS 87

triazolam and placebo in healthy volunteers. Clin Neuro- Calne DB, Teychenne PF, Leigh PN et al. (1974). Treatment
pharmacol 22: 1523. of Parkinsons disease with bromocriptine. Lancet 2:
Arai N, Isaji M, Miyata H et al. (1995). Differential effects 13551356.
of three dopamine receptor agonists in MPTP-treated Chase TN (1998). The significance of continuous dopami-
monkeys. J Neural Transm 10: 5562. nergic stimulation in the treatment of Parkinsons disease.
Barone P, Bravi D, Bermejo-Pareja F et al. (1999), and the Drug 55: S1S9.
Pergolide Monotherapy Study Group. Pergolide monother- Clarke CE, Deane KH (2001a). Ropinirole for levodopa-
apy in the treatment of early Parkinsons disease: a rando- induced complications in Parkinsons disease. Cochrane
mized, controlled study. Neurology 53: 573579. Database Syst Rev 1 CD001516.
Bedard PJ, Di Paolo T, Falardeau P et al. (1986). Chronic Clarke CE, Deane KH (2001b). Ropinirole versus bromo-
treatment with L-dopa but not bromocriptine induces dys- criptine for levodopa-induced complications Parkinsons
kinesia in MPTP-parkinsonian monkeys. Correlation with disease. Cochrane Database Syst Rev 1: CD001517.
[3H]spiperone binding. Brain Res 379: 294299. Clarke CE, Guttman M (2002). Dopamine agonist early
Bennett JP, Landrow ER, Dietrich S et al. (1994). Suppres- therapy in Parkinsons disease. Lancet 360: 17671769.
sion of dyskinesias in advanced Parkinsons disease: mod- Clarke CE, Speller JM (2000a). Pergolide for levodopa-
erate daily clozapione doses provide long-term dyskinesia induced complications in Parkinsons disease. Cochrane
reduction. Mov Disord 9: 404414. Database Syst Rev 2: CD000235.
Ben-Noun L (2000). Drug-induced respiratory disorders: inci- Clarke CE, Speller JM (2000b). Pergolide versus bromocrip-
dence, prevention and management. Drug Saf 23: 143164. tine for levodopa-induced motor complications in Parkin-
Bergamasco B, Frattola L, Muratorio A et al. (2000). sons disease. Cochrane Database Syst Rev 2: CD000236.
Alpha-dihydroergocryptine in the treatment of de novo
Clarke CE, Speller JM, Clarke JA (2000a). Pramipexole for
parkinsonian patients: results of a multicentre, rando-
levodopa-induced complications in Parkinsons disease.
mised, double-blind, placebo-controlled study. Acta
Cochrane Database Syst Rev 3: CD002261.
Neurol Scand 101: 372380.
Clarke CE, Speller JM, Clarke JA (2000b). Pramipexole
Bhatt MH, Keenan SP, Fleetham JA et al. (1991). Pleuropul-
versus bromocriptine for levodopa-induced complications
monary disease associated with dopamine agonist therapy.
in Parkinsons disease. Cochrane Database Syst Rev 3:
Ann Neurol 30: 613616.
CD002259.
Biglan KM, Holloway RG (2002). A review of pramipexole
and its clinical utility in Parkinsons disease. Expert Opin Colzi A, Turner K, Lees AJ (1998). Continuous subcuta-
Pharmacother 3: 197210. neous waking day apomorphine in the long term treatment
Birkmayer W, Hornykiewicz O (1970). Der L-3,4 dioxyphe- of levodopa induced interdose dyskinesia in Parkinsons
nylalanin DOPA: effect bie der Parkinson akinese. disease. J Neurol Neurosurg Psychiatry 64: 573576.
Wien Klin Wochenschr 74: 787788 [Engl transl Parkin- Corrigan MH, Denahan AQ, Wright CE et al. (2000). Com-
sonism Rel Dis 1998; 4(2): 6162]. parison of pramipexole, fluoxetine, and placebo in patients
Blin O (2003). The pharmacokinetics of pergolide in Parkin- with major depression. Depress Anxiety 11: 5865.
sons disease. Curr Opin Neurol 16: S9S12. Cristina S, Zangaglia R, Mancini F et al. (2003). High-dose
Boas J, Worm-Petersen J, Dupont E et al. (1996). The levo- ropinirole in advanced Parkinsons disease with severe
dopa dose-sparing capacity of pergolide compared with dyskinesias. Clin Neuropharmacol 26: 146150.
that of bromocriptine in an open-label, cross-over study. Curran MP, Perry CM (2004). Cabergoline. A review of its
Eur J Neurol 3: 4449. use in the treatment of Parkinsons disease. Drugs 64:
Bonuccelli U (2003). Comparing dopamine agonists in 21252141.
Parkinsons disease. Curr Opin Neurol 16: S13S19. Deleu D, Northway MG, Hanssens Y (2002). Clinical phar-
Bowron A (2004). Practical considerations in the use of macokinetic and pharmacodynamic properties of drugs
apomorphine injectable. Neurology 62: S32S36. used in the treatment of Parkinsons disease. Clin Pharma-
Bracco F, Battaglia A, Chouza C et al. (2004). PKD009 cokinet 41: 261309.
Study Group. The long-acting dopamine receptor agonist Deleu D, Hanssens Y, Northway MG (2004). Subcutaneous
cabergoline in early Parkinsons disease: final results of apomorphine: an evidence-based review of its use in
a 5-year, double-blind, levodopa-controlled study. CNS Parkinsons disease. Drugs Aging 21: 687709.
Drugs 18: 733746. Dewey RB, Hutton JT, LeWitt PA et al. (2001). A rando-
Brodsky MA, Godbold J, Roth T et al. (2003). Sleepiness in mised double-blind placebo-controlled trial of subcuta-
Parkinsons disease: a controlled study. Mov Disord 18: neously injected apomorphine for parkinsonian off-stat
668672. events. Arch Neurol 58: 13851392.
Brunt ER, Brooks DJ, Korczyn AD et al. (2002). on behalf of Di Marco A, Appiah-Kubi LS, Chaudhuri KR (2002). Use of
the 043 Study Group. A six-month multicentre, double- the dopamine agonist cabergoline in the treatment of
blind, bromocriptine-controlled study of the safety and movement disorders. Expert Opin Pharmacother 3:
efficacy of ropinirole in the treatment of patients with Par- 14811487.
kinsons disease not optimally controlled by L-dopa. Domino EF, Ni L, Zang H et al. (1998). Effects of prami-
J Neural Transm 109: 489502. pexole on contraversive rotation and functional motor
88 O. RASCOL ET AL.

impairments in l-methyl-4-phenyl-1,2,3,6-tetrahydro- Frucht S, Rogers JD, Greene PE et al. (1999). Falling asleep
pyridine-induced chronic hemiparkinsonian monkeys. at the wheel: motor vehicule mishaps in persons taking
J Pharmacol Exp Ther 287: 983987. pramipexole and ropinirole. Neurology 52: 19081910.
Durif F, Debilly B, Galitzky M et al. (2004). Clozapine Goetz CG, Koller WC, Poewe W et al. (2002). Management
improves dyskinesias in Parkinson disease: a double-blind, of Parkinsons disease: an evidence-based review. Mov
placebo-controlled study. Neurology 62: 381388. Disord 17: S1S166.
Ebersbach G, Norden J, Tracik F (2000). Sleep attacks in Goetz CG, Poewe W, Rascol O et al. (2005). Evidence-based
Parkinsons disease: polysomnographic recordings. Mov medical review update: pharmacological and surgical
Disord 15 (Suppl 3): P504. treatments of Parkinsons disease: 2001 to 2004. Mov
Eden RJ, Costall B, Domeney AM et al. (1991). Preclinical Disord 20: 395411.
pharmacology of ropinirole (SK-&-F-101468-A) a novel Gomez-Mancilla B, Bedard PJ (1992). Effect of chronic
dopamine-D2 agonist. Pharmacol Biochem Behav 38: treatment with ()-PHNO, a D2 agonist in MPTP-treated
147154. monkeys. Exp Neurol 117: 185188.
Edwards IR, Aronson JK (2000). Adverse drug reactions: Grondin R, Goulet M, Di Paolo T et al. (1996). Cabergoline
definitions, diagnosis, and management. Lancet 356: a long-acting dopamine D2-like receptor agonist, produces
12551259. a sustained antiparkinsonian effect with transient dyskine-
Etminan M, Samii A, Takkouche B et al. (2001). Increased sias in parkinsonian drug-naive primates. Brain Res 735:
risk of somnolence with the new dopamine agonsits in 298306.
patients with Parkinsons disease: a meta-analysis of rando- Grondin R, Bedard PJ, Britton DR et al. (1997). Potential
mised controlled trials. Drug Saf 24: 863888. therapeutic use of the selective dopamine D1 receptor
Fabre N, Montastruc JL, Rascol O (1993). Alopecia: an agonist, A-86929: an acute study in parkinsonian levo-
adverse effect of bromocriptine. Clin Neuropharmacol dopa-primed monkeys. Neurology 49: 421426.
16: 266268. Grosset K, Needleman F, Macphee G et al. (2004). Switching
Facca A, Sanchez-Ramos J (1996). High-dose pergolide from ergot to nonergot dopamine agonists in Parkinsons
monotherapy in the treatment of severe levodopa-induced disease: a clinical series and five-drug dose conversion
dyskinesias. Mov Disord 11: 327329. table. Mov Disord 19: 13701374.
Factor SA (2004). Literature review: intermittent subcuta- Guttman M (1997). and the International Pramipexole-
neous apomorphine therapy in Parkinsons disease. Neu- Bromocriptine Study Group. Double-blind comparison of
rology 62: S12S17. pramipexole and bromocriptine treatment with placebo in
Factor SA, Molho ES, Podskalny GD et al. (1995). Parkin- advanced Parkinsons disease. Neurology 49: 10601065.
sons disease: drug-induced psychiatric states. Adv Neurol Hagell P, Odin P (2001). Apomorphine in the treatment of
65: 115138. Parkinsons disease. J Neurosci Nurs 33: 21343738.
Fahn S, Elton R (1987), and the members of the UPDRS Hall ED, Andrus PK, Oostveen JA et al. (1996). Neuroprotec-
Development Committee. Unified Parkoinsons disease rat- tive effects of the dopamine D2/D3 agonist pramipexole
ing scale. In: S Fahn, CD Marsden, DB Calne, M Goldstein against postischemic or metamphetamine induced degen-
(Eds.), Vol. 2, Macmillan Publishing Co Inc, New-York, eration of nigrostriatal neurons. Brain Res 742: 8088.
pp. 293304. Happe S, Trenkwalder C (2004). Role of dopamine receptor
Fariello RG (1998). Pharmacodynamic and pharmacokinetic agonists in the treatment of restless legs syndrome. CNS
features of cabergoline. Rationale for use in Parkinsons Drugs 18: 2736.
disease. Drugs 55: 1016. Hausser RA, Friedlander J, Zesiewicz TA et al. (2000).
Ferreira JJ, Galitzky M, Montastruc JL et al. (2000). Sleep A home diary to assess functional status in patients with
attacks and Parkinsons disease treatment. Lancet 355: Parkinsons disease with motor fluctuations and dyskine-
13331334. sia. Clin Neuropharmacol 23: 7581.
Ferreira JJ, Thalamas C, Montastruc JL et al. (2001). Levodo- Hely MA, Morris JGL (1999). The Sydney multicentre study
pa monotherapy can induce sleep attacks in Parkinsons of Parkinsons disease: progression and mortality at 10
disease patients. J Neurol 248: 426427. years. J Neurol Neurosurg Psychiatry 67: 300307.
Ferreira JJ, Galitzky M, Thalamas C et al. (2002). Effect of Hely MA, Morris JGL, Reid WGJ (1994). The Sydney multi-
ropinirole on sleep onset: a randomized, placebo-controlled centre study of Parkinsons disease: a randomized, prospec-
sutdy in healthy volunteers. Neurology 58: 460462. tive five year study comparing low dose bromocriptine
Ferreira JJ, Desboeuf K, Galitzky M et al. (2006). Sleep disrup- with low dose levodopa-carbidopa. J Neurol Neurosurg
tion, daytime somnolence and sleep attacks in Parkinsons Psychiatry 57: 903910.
disease: a clinical survey in Parkinsons disease patients and Hening WA, Allen RP, Earley CJ et al. (2004). Restless Legs
age-matched healthy volunteers. Eur J Neurol 13: 209214. Syndrome Task Force of the Standards of Practice Com-
French Clozapine Parkinson Study Group (1999). Clozapine mittee of the American Academy of Sci Medicine. An
in drug-induced psychosis in Parkinsons disease. The update on the dopaminergic treatment of restless legs syn-
French Clozapine Parkinson Study Group. Lancet 353: drome a periodic limb movement disorder. Sleep 27:
20412042. 560583.
DOPAMINE AGONISTS 89

Hobson DE, Lang AE, Martin WR et al. (2002). Excessive Korczyn AD, Brunt ER, Larsen JP et al. (1999). A 3-year
daytime sleepniness and sudden-onset sleep in Parkinson randomized trial of ropinirole and bromocriptine in early
disease: a survey by the Canadian Movement Disorders Parkinsons disease. The 053 Study Group. Neurology
Group. JAMA 287: 455463. 53: 364370.
Hoehn MM, Elton RL (1985). Low dosages of bromocriptine Korner Y, Meindorfner C, Moller JC et al. (2004). Predictors
added to levodopa in Parkinsons disease. Neurology 35: of sudden onset of sleep in Parkinsons disease. Mov Dis-
199206. ord 19: 129812305.
Holloway RG, Shoulson I, Fahn S et al. (2004). Parkinson Laihinen A, Rinne UK, Suchy I (1992). Comparison of lisur-
Study Group. Pramipexole vs levodopa as initial treatment ide and bromocriptine in the treatment of advanced Par-
for Parkinson disease: a 4-year randomized controlled kinsons disease. Acta Neurol Scand 86: 593595.
trial. Arch Neurol 61: 10441053. Lane RJ, Routledge PA (1983). Bromocriptine has been
Homann CN, Wenzel K, Suppan K et al. (2000). Sleep reported to cause visual cortical disturbances. Drug-
attacks after acute administration of apomorphine. Mov induced neurological disorders. Drugs 26: 124147.
Disord 15 (Suppl 3): P585. Lattanzi L, DellOsso L, Cassano P et al. (2002). Pramipexole
Homann CN, Wenzel K, Suppan K et al. (2002). Sleep in treatment-resistant depression: a 16-week naturalistic
attacks in patients taking dopamine agonists: review. study. Bipolar Disord 4: 307314.
BMJ 324: 14831487. Le WD, Jankovic J (2001). Are dopamine receptor agonists
Horvath J, Fross RD, Kleiner-Fisman G et al. (2004). Severe neuroprotective in Parkinsons disease? Drugs Aging 18:
multivalvular heart disease: a new complication of the ergot 389396.
derivative dopamine agonists. Mov Disord 19: 656661. Lees AJ, Katzenschlager R, Head J et al. (2001), on behalf of
Hutton JT, Koller WC, Ahlskog JE et al. (1996). Multicenter, the Parkinsons Disease Research Group on the United King-
placebo-controlled trial of cabergoline taken once daily in dom. Ten-year follow-up of three different initial treatments in
the treatment of Parkinsons disease. Neurology 46: de-novo PD. A randomized trial. Neurology 57: 16871694.
10621065. LeWitt PA (2004). Subcutaneously administered apomor-
Iida M, Miyazaki I, Tanaka KI et al. (1999). Dopamine D2 phine: pharmacokinetics and metabolism. Neurology 62:
receptor-mediated antioxidant and neuroprotective effects S8S11.
of ropinirole, a dopamine agonist. Brain Res 838: 5159. LeWitt PA, Gopinathan G, Ward CD et al. (1982). Lisuride
Inzelberg R, Nisipeanu P, Rabey JM et al. (1996). Double-blind versus bromocriptine treatment in Parkinson disease: a
comparison of cabergoline and bromocriptine in Parkinsons double-blind study. Neurology 32: 6972.
disease patients with motor fluctuations. Neurology 47: LeWitt PA, Ward CD, Larsen TA et al. (1983). Comparison
785788. of pergolide and bromocriptine therapy in parkinsonism.
Inzelberg R, Schechtman E, Nisipeanu P (2003). Cabergoline, Neurology 33: 1091014.
pramipexole and ropinirole used as immunotherapy in Lieberman A, Olanow CW, Sethi K et al. (1998), and the
early Parkinsons disease. Drugs Aging 20: 847855. Ropinirole Study Group. A multicenter trial of ropinirole
Izumi T, Inoue T, Kitagawa N et al. (2000). Open pergolide adjunct treatment for Parkinsons disease. Neurology 51:
treatment of tricyclic and heterocyclic antidepressant- 10571062.
resistant depression. J Affect Disord 61: 127132. Ling LH, Ahlskog JE, Munger TM et al. (1999). Constrictive
Jenner P (2005). A novel dopamine agonist for the transder- pericarditis and pleuropulmonary disease linked to ergot
mal treatment of Parkinsons disease. Neurology 65: dopamine agonist therapy (cabergoline) for Parkinsons
S3S5. disease. Mayo Clin Proc 74: 371375.
Jenner P, Olanow CW (1996). Oxidative stress and the patho- Luquin RM, Laguna J, Obeso JA (1992). Selective D2 recep-
genesis of Parkinsons disease. Neurology 47: S161S170. tor stimulation induces dyskinesia in parkinsonian mon-
Katzenschlager R, Hughes A, Evans A et al. (2005). Contin- keys. Ann Neurol 31: 551554.
uous subcutaneous apomorphine therapy improves dyski- Maj J, Rogoz Z, Skuza G et al. (1997). Antidepressant
nesias in Parkinsons disease: a prospective study using effects of pramipexole, a novel dopamine receptor agonist.
single-dose challenges. Mov Disord 20: 151157. J Neural Transm 104: 525533.
Kaye CM, Nicholls B (2000). Clinical pharmacokinetics of Manni R, Terzaghi M, Sartori I et al. (2004). Dopamine ago-
ropinirole. Clin Pharmacokinet 39: 243254. nists and sleepiness in PD: review of the literature and per-
Koller W, Lees A, Doder M et al. (2001). Tolcapone/Pergo- sonal findings. Sleep Med 5: 189193.
lide Study Group. Randomized trial of tolcapone versus Manson AJ, Turner K, Lees AJ (2002). Apomorphine mono-
pergolide as add-on to levodopa therapy in Parkinsons therapy in the treatment of refractory motor complications
disease patients with motor fluctuations. Mov Disord 16: of Parkinsons disease: long-term follow-up study of 64
858866. patients. Mov Disord 17: 12351241.
Korczyn AD, Brooks DJ, Brunt ER et al. (1998), on behalf of Marsden CD, Parkes JD, Quinn N (1987). Fluctuations of
the 053 Study Group. Ropinirole versus bromocriptine in disability in Parkinsons disease: pathophysiological
the treatment of early Parkinsons disease: 6-month aspects. In: CD Marsden, S Fahn (Eds.), Movement Dis-
interim report of a 3-year study. Mov Disord 13: 4651. orders. Butterworth, London, pp. 96122.
90 O. RASCOL ET AL.

Martignoni E, Pacchetti C, Sibilla L et al. (1991). Dihy- Nutt JG (1990). Levodopa-induced dyskinesia. Neurology
droergocryptine in the treatment of Parkinsons disease: 40: 340345.
a six months double-blind clinical trial. Clin Neurophar- Ogawa N, Miyazaki I, Tanaka K et al. (1999). Dopamine D2
macol 14: 7883. receptor mediated antioxidant and neuroprotective effects
Martindale (2002). Edited by Sean C. Sweetman. The Com- of ropinirole. Parkinsonism Relat Disord 5: S1S81.
plete Drug Reference 33rd edition, Pharmacological Press Olanow CW (1990). Oxidation reactions in Parkinsons dis-
London, Chicago, pp. 11601179. ease. Neurology 40: S32S39.
Matheson AJ, Lamb HM (2000). Quetiapine: a review of its Olanow CW, Fahn S, Muenter M et al. (1994). A multicenter
clinical potential in the management of psychotis symp- double-blind placebo-controlled trial of pergolide as an
toms in Parkinsons disease. CNS Drugs 14: 157172. adjunct to Sinemet in Parkinsons disease. Mov Disord
Matheson AJ, Spencer CM (2000). Ropinirole. A review of 9: 4047.
its use in the management of Parkinsons disease. Drugs Olanow CW, Hauser RA, Gauger L et al. (1995). The effect
60:115137. of deprenyl and levodopa on the progression of Parkin-
Mizuno Y, Kondo T, Narabayashi H (1995). Pergolide in the sons disease. Ann Neurol 38: 771777.
treatment of Parkinsons disease. Neurology 45: S13S21. Olanow CW, Jenner P, Brooks D (1998). Dopamine agonists
Mizuno Y, Yanagisawa N, Kuno S et al. (2003). and the and neuroprotection in Parkinsons disease. Ann Neurol
Pramipexole Study Group. Randomized, double-blind 44: 167174.
study of pramipexole with placebo and bromocriptine Olanow CW, Schapira AHV, Rascol O (2000). Continuous
in advanced Parkinsons disease. Mov Disord 18: dopamine-receptor stimulation in early Parkinsons dis-
11491156. ease. Trends Neurosci 23: S117S126.
Mondal BK, Suri S (2000). Pergolide-induced retroperitoneal
Olanow CW, Agid Y, Mizuno Y et al. (2004). Levodopa in
fibrosis. Int J Clin Pract 54: 403.
the treatment of Parkinsons disease: current controver-
Montastruc JL, Rascol A (1984). Treatment of Parkinsons
sies. Mov Disord 19: 9971005.
disease with high doses of bromocriptine. Possible interac-
Ostergaard L, Werdelin L, Odin P et al. (1995). Pen injected
tion with josamycin. Presse Med 13: 22672268.
apomorphine against off phenomena in late Parkinsons
Montastruc JL, Rascol O (2001). Modafinil and pramipex-
disease: a double blind, placebo controlled study. J Neurol
ole-associated somnolence. Mov Disord 15: 783784.
Neurosurg Psychiatry 58: 681687.
Montastruc JL, Rascol O, Senard JM (1993). Current status
of dopamine agonists in Parkinsons disease management. Paladini D (2000). Sleep attacks in two Parkinsons disease
Drugs 46: 384393. patients taking ropinirole. Mov Disord 15 (Suppl 3): P671.
Montastruc JL, Rascol O, Senard JM et al. (1994). A rando- Parkinson Study Group (1989). Effect of deprenyl on the
mised controlled study comparing bromocriptine to which progression of disability in early Parkinsons disease.
levodopa was later added, with levodopa alone in previously N Engl J Med 321: 13641371.
untreated patients with Parkinsons disease: a five year Parkinson Study Group (1999). Low-dose clozapine for the
follow-up. J Neurol Neurosurg Psychiatry 57: 10341038. treatment of drug-induced psychosis in Parkinsons dis-
Montastruc JL, Desboeuf K, Lapeyre-Mestre M et al. ease. N Engl J Med 340: 757763.
(2001a). Long-term mortality results on the randomized Parkinson Study Group (2000). Pramipexole versus levodopa
controlled study comparing bromocriptine to which levo- as initial treatment for Parkinsons disease: a randomized
dopa was later added with levodopa alone in previously controlled trial. JAMA 284: 19311938.
untreated patients with Parkinsons disease. Mov Disord Parkinson Study Group (2002a). Dopamine transporter brain
16: 511514. imaging to assess the effects of pramipexole vs. levodopa
Montastruc JL, Brefel-Courbon C, Senard JM et al. on Parkinsons disease progression. JAMA 287: 16531661.
(2001b). Sleep attacks and antiparkinsonian drugs: a pilot Parkinson Study Group (2002b). A controlled trial of rasagi-
prospective pharmacoepidemiologic study. Clin Neuro- line in early Parkinson disease: the TEMPO study. Arch
pharmacol 24: 181183. Neurol 59: 19371943.
Mucke HAM (2003). Rotigotine Schwarz Pharma. Drugs 6: Parkinsons Disease Research Group in the United Kingdom
894899. (1993). Comparisons of therapeutic effects of levodopa,
Muller T, Przuntek H, Kuhlmann A (2003). Loss of color levodopa and selegiline, and bromocriptine in patients
vision during long-term treatment with pramipexole. with early, mild Parkinsons disease: three year interim
J Neurol 250: 101102. report. BMJ 307: 469472.
Navan P, Findley LJ, Undy MB et al. (2005). A randomly Paus S, Brecht HM, Koster J et al. (2003). Sleep atatcks,
assigned double-blind cross-over study examining the daytime sleepiness, and dopamine agonists in Parkinsons
relative anti-parkinsonian tremor effects of pramipexole disease. Mov Disord 18: 659667.
and pergolide. Eur J Neurol 12: 18. Pearce RKB, Banerji T, Jenner P et al. (1998). De novo
Neef C, van Laar T (1999). Pharmacokinetic-pharmacody- administration of ropinirole and bromocriptine induces
namic relationship of apomorphine in patients with Par- less dyskinesia than L-dopa in the MPTP-treated marmo-
kinsons disease. Clin Pharmacokinet 37: 257271. set. Mov Disord 13: 234241.
DOPAMINE AGONISTS 91

Periti P, Mazzei T, Mini E et al. (1992). Pharmacokinetic Rascol O, Brooks DJ, Brunt ER et al. (1998), on behalf of
drug interactions of macrolides. Clin Pharmacokinet 23: the 056 Study Group. Ropinirole in the treatment of early
106131. Parkinsons disease: a 6-month interim report of a 5-year
Perugi G, Toni C, Ruffolo G et al. (2001). Adjunctive dopa- levodopa-controlled study. Mov Disord 13: 3945.
mine agonists in treatment-resistant bipolar II depression: Rascol O, Brooks DJ, Korczyn AD et al. (2000), for the 056
an open case series. Pharmacopsychiatry 34: 137141. Study Group. A five-year study of the incidence of dyski-
Pezzoli G, Martignoni E, Pachetti C et al. (1994). Pergolide nesia in patients with early Parkinsons disease who were
compared with bromocriptine in Parkinsons disease: a treated with ropinirole or levodopa. N Engl J Med 342:
multicenter, cross-over, controlled study. Mov Disord 9: 14841491.
431436. Rascol O, Nutt JG, Blin O et al. (2001). Induction by dopa-
Pfeiffer RE (1996). Antiparkinsonian agents: drug interac- mine D1 receptor agonist ABT-431 of dyskinesia similar
tions of clinical significance. Drug Saf 14: 343354. to levodopa in patients with Parkinsons disease. Arch
Piercey MF (1998). Pharmacology of pramipexole, a dopa- Neurol 58: 249254.
mine D3-preferring agonist useful in treating Parkinsons Rascol O, Goetz C, Koller W et al. (2002). Treatment inter-
disease. Clin Neuropharmacol 21: 141151. ventions for Parkinsons disease: an evidence based
Piercey MF, Hoffmann WE, Smith HW et al. (1996). Inhibi- assessment. Lancet 359: 15891598.
tion of dopamine neuron firing by pramipexole, a D3-pre- Rascol O, Payoux P, Ory F et al. (2003). Limitations of current
fering dopamine agonist: comparison to other dopamine Parkinsons disease therapy. Ann Neurol 53 (Suppl 3),
agonists. Eur J Pharmacol 312: 3544. S3S12, discussion S12S15.
Pinter MM, Helscher RJ, Mundsperger N et al. (1998). Transi- Rascol O, Pathak A, Bagheri H et al. (2004a). New concerns
ent increase of pancreatic enzymes evoked by apomorphine about old drugs: valvular heart disease on ergot derivative
in Parkinsons disease. J Neural Transm 105: 12371244. dopamine agonists as an exemplary situation on pharma-
Pinter MM, Pogarell O, Oertel WH (1999). Efficacy, safety covigilance. Mov Disord 19: 611613.
and tolerance of the non-ergoline dopamine agonist prami- Rascol O, Pathak A, Bagheri H et al. (2004b). Dopaminago-
pexole in the treatment of advanced Parkinsons disease: nists and fibrotic valvular heart disease: further considera-
double-blind, placebo-controlled, randomized, multicenter tions. Mov Disord 19: 15241525.
study. J Neurol Neurosurg Psychiatry 66: 436441. Rawlins MD, Thomson JW (1977). Pathogenesis of adverse
Poewe W, Lussi F (2005). Clinical studies with transdermal drug reactions. In: DM Daines, (Ed.), Text Book of
rotigotine in early Parkinsons disease. Neurology 65: Adverse Drug Reactions. Oxford University Press, p. 44.
S11S14.
Razmy A, Lang AE, Shapiro CM (2004). Predictors of
Poewe W, Wenning GK (2000). Apomorphine: an underuti-
impaired daytime sleep and wakefulness in patients with
lized therapy for Parkinsons disease. Mov Disord 15:
Parkinson disease treated with older (ergot) vs newer
789794.
(nonergot) dopamine agonists. Arch Neurol 61: 97102.
Poewe WP, Kleedorfer B, Wagner M et al. (1993). Continu-
Reichmann H, Herting B, Muller A et al. (2003). Switching
ous subcutaneous apomorphine infusions for fluctuating
and combining dopamine agonists. J Neural Transm 110:
Parkinsons disease. Long-term follow-up in 18 patients.
13931400.
Adv Neurol 60: 656659.
Rektorova I, Rektor I, Bares M et al. (2003). Pramipexole
Pogarell O, Gasser T, Van Hilten JJ et al. (2002). Pramipex-
and pergolide in the treatment of depression in Parkinsons
ole in patients with Parkinsons disease and marked drug
disease: a national multicentre prospective randomized
resistant tremor: a randomized, double-blind, placebo-con-
study. Eur J Neurol 10: 399406.
trolled multicentre trial. J Neurol Neurosurg Psychiatry
72: 713720. Rinne U (1989). Lisuride a dopamine agonist in the treatment
Przuntek H, Welzel D, Gerlach M et al. (1996). Early institu- of early Parkinsons disease. Neurology 39: 336339.
tion of bromocriptine in Parkinsons disease inhibits the Rinne UK, Bracco F, Chouza C (1997), on behalf of the
emergence of levodopa-associated motor side effects: PKD009 Collaborative Study Group. Carbergoline in the
long-term results of the results of the PRADO study. treatment of early Parkinsons disease: results of the first
J Neural Transm 103: 699715. year of treatment in a double-blind comparison of caber-
Rascol O (1997). Ropinirole: clinical profile. In: CW goline and levodopa. Neurology 48: 363368.
Olanow, JA Obeso (Eds.), Beyond the Decade of the Rinne UK, Bracco F, Chouza C et al. (1998), and the
Brain: Dopamine Agonists in Early Parkinsons disease. PKDS009 Study Group. Early treatment of Parkinsons
Wells Medical Ltd, Royal Tunbridge Wells, pp. 163176. disease with cabergoline delays the onset of motor compli-
Rascol O (2005). Transdermal delivery of dopaminergic cations. Drugs 55: 2330.
agents. Neurology 65: S1S2. Riopelle RJ (1987). Bromocriptine and the clinical spectrum
Rascol O, Lees AJ, Senard JM et al. (1996). Ropinirole of Parkinsons disease. Can J Neurol Sci 14: 455459.
in the treatment of levodopa-induced motor fluctuations Robertson HA (1992). Dopamine receptor interactions: some
in patients with Parkinsons disease. Clin Neuropharmacol implications for the treatment of Parkinsons disease.
19: 234245. Trends Neurosci 15: 201206.
92 O. RASCOL ET AL.

Rondot P, Ziegler M (1992). Activity and acceptability of Todman DH, Oliver WA, Edwards RL (1990). Pleuropul-
piribedil in Parkinsons disease: a multicentre study. monary fibrosis due to bromocriptine treatment for Parki-
J Neurol 239 (Suppl 1): 2834. nosns disease. Clin Exp Neurol 27: 7982.
Ryan M, Slevin JT, Wells A (2000). Non-ergot dopamine Tolcapone Study Group (1999). Efficacy and tolerability of
agonist-induced sleep attacks. Pharmacotherapy 20: tolcapone compared with bromocriptine in levodopa-trea-
724726. ted parkinsonian patients. Mov Disord 14: 3844.
Schapira AH (2000). Sleep attacks (sleep episodes) with Toyokura Y, Mizuno Y, Kase M et al. (1985). Effects of bro-
pergolide. Lancet 355: 13321333. mocriptine on parkinsonism. A nation-wide collaborative
Schapira AH (2002). Neuroprotection and dopamine double-blind study. Acta Neurol Scand 72: 157170.
agonists. Neurology 58: S9S18. Tulloch IF (1997). Pharmacologic profile of ropinirole: a
Schapira AH, Olanow CW (2003). Rationale for the use of nonergoline dopamine agonist. Neurology 49: S58S62.
dopamine agonists as neuroprotective agents in Parkin- Uitti RJ, Ahlskog JE (1996). Comparative review of dopa-
sons disease. Ann Neurol 53: S149S159. mine receptor agonist in Parkinsons disease. CNS Drugs
Schapira AHV, Olanow CW (2004). Neuroprotection in Par- 5: 369388.
kinsons disease. Mysteries, Myths, and Misconceptions. Van Camp G, Flamez A, Cosyns B et al. (2004). Treatment
JAMA 291: 358364. of Parkinsons disease with pergolide and relation to
Schlesinger I, Ravin PD (2003). Dopamine agonist induce epi- restrictive valvular heart disease. Lancet 363: 11791183.
sode of irresistible daytime sleepiness. Eur Neurol 49: 3033. Vergeret J, Barat M, Taytard A et al. (1984). Pleuropulmon-
Schrag A, Keens J, Warner J (2002). Ropinirole Study ary fibrosis and bromocriptine. Sem Hop 60: 741744.
Group. Ropinirole for the treatment of tremor in early Par- Whone AL, Watts RL, Stoessl AJ et al. (2003). REAL-PET
kinsons disease. Eur J Neurol 9: 253257. Study Group. Slower progression of Parkinsons disease
Schwarz J (2003). Rationale for dopamine agonist use as with ropinirole versus levodopa: the REAL-PET study.
immunotherapy in Parkinsons disease. Curr Opin Neurol Ann Neurol 54: 93101.
16: S27S33. Wise RA, Rompre PP (1989). Brain dopamine and reward.
Sethy VH, Wu H, Oostveen JA et al. (1997). Neuroprotective Annu Rev Psychol 40: 191225.
effects of the dopamine agonists pramipexole and bromo- Wolf WA (2003). SLV-308. Solvay. Curr Opin Investig
criptine in 3-acetylpyridine-treated rats. Brain Res 754: Drugs 4: 878882.
181186. Yoshikawa T, Minamiyama Y, Naito Y et al. (1994). Antiox-
Shannon KM, Bennett JP, Friedman JH (1997). for the Pra- idant properties of bromocriptine, a dopamine agonist.
mipexole Study Group. Efficacy of pramipexole, a novel J Neurochem 62: 10341038.
dopamine agonist, as monotherapy in mild to moderate Zarate CA, Payne JL, Singh J et al. (2004). Pramipexole for
Parkinsons disease. Neurology 49: 724728. bipolar II depression: a placebo-controlled proof of con-
Shaunak S, Wilkins A, Pilling JB et al. (1999). Pericardial, cept study. Biol Psychiatry 56: 5460.
retroperitoneal, and pleural fibrosis induced by pergolide. Ziegler M, Castro-Caldas A, Del Signore S et al. (2003).
J Neurol Neurosurg Psychiatry 66: 7981. Efficacy of piribedil as early combination to levodopa
Stewart D, Morgan E, Burn D et al. (2004). Dopamine ago- in patients with stable Parkinsons disease: a 6-month,
nist switching in Parkinsons disease. Hosp Med 65: randomized placebo-controlled study. Mov Disord 18:
215219. 418425.
Stibe CM, Lees AJ, Kempster PA et al. (1988). Subcutaneous
apomorphine in parkinsonian on-off oscillations. Lancet 1:
Further Reading
403406.
Tan EK (2003). Piribedil-induced sleep attacks in Parkin- Etminan M, Gill S, Samii A (2003). Comparison of the risk
sons disease. Fundam Clin Pharmacol 17: 117119. of adverse events with pramipexole and ropinirole in
Tan EK, Jankovic J (2001). Choosing dopamine agonists in patients with Parkinsons disease: a meta-analysis. Drug
Parkinsons disease. Clin Neuropharmacol 24: 247253. Saf 26: 439444.
Tandberg E, Larsen JP, Karlsen K (1999). Excessive daytime Hunziker T, Bruppacher R, Kuenzi UP et al. (2002). Classifi-
sleepiness and sleep benefit in Parkinsons disease: a com- cation of ADRs: a proposal for harmonization and differ-
munity-based study. Mov Disord 14: 922927. entiation based on the experience of the Comprehensive
Tintner R, Jankovic J (2003). Dopamine agonists in Parkin- Hospital Drug Monitoring Bern/St. Gallen, 19741993.
sons disease. Expert Opin Investig Drugs 12: 18031820. Pharmacoepidemiol Drug Saf 11: 159163.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 34

Monoamine oxidase A and B inhibitors in Parkinsons disease

MOUSSA B. H. YOUDIM1* AND PETER F. RIEDERER2

1
Department of Pharmacology, Technion-Bruce Rappaport Faculty of Medicine, Eve Topf and NPF Neurodegenerative
Diseases Centers, Rappaport Family Research Institute, Haifa, Israel
2
Clinical Neurochemistry, Department of Psychiatry and Psychotherapy, National Parkinson Foundation (USA) Center of
Excellence Research Laboratories, University of Wurzburg, Wurzburg, Germany

34.1. Introduction indirectly acting sympathomimetic amines, present in


food (cheeses and many diverse foodstuffs) and drinks
Monoamine oxidase (MAO), which catalyzes primary, (beer and wine), especially those that are fermented.
secondary and tertiary amines (Fig. 34.1), was discov- Most such amines are metabolized by MAO. Tyramine,
ered in 1928 by Hare Bernhiem and later identified by a substrate of MAO, if present in ingested food cannot
Balschko as one of the most important enzymes in be metabolized as a consequence of MAO inhibition.
neurotransmitter metabolism (Youdim et al., 2005a, It can gain access to the circulatory system and induce
2006). Its physiological roles and inhibitors have a significant release of norepinephrine from peripheral
played major roles in our understanding of the func- adrenergic neurons (Fig. 34.2). The consequence of this
tional roles of dopamine (DA), norepinephrine and ser- can be severe cardiovascular response, resulting in
otonin (5-HT) neurotransmission in the central nervous hypertensive reaction, and which can be fatal in some
system (CNS). One of the first psychotropic drugs to be cases. The search for an understanding of the mechan-
identified was iproniazid, the first MAO inhibitor to ism of the cheese reaction by MAO inhibitors and
be discovered by serendipity and introduced into clinic development of inhibitors devoid of this fatal side-effect
as an antidepressant. Although this drug and other non- continued with the identification of multiple forms of
selective MAO inhibitors demonstrated remarkable MAO with different substrate specificity and inhibitor
antidepressant action in the late 1950s and 1960s sensitivity (Youdim and Weinstock, 2001).
(Table 34.1), their usefulness as antidepressants was
seriously challenged in earlier clinical studies, mainly 34.2. Multiple forms of brain monoamine
because of limited dosage employed. In addition some oxidase
drugs, such as iproniazid, caused liver toxicity and
others, such as tranylcypromine, induced what became The demonstration that MAO was not a single enzyme
known as the cheese reaction. These side-effects ser- but existed in several forms in the liver and brain
iously hampered the development and assessment of tissues of rats (Youdim and Sourkes, 1965) was
other MAO inhibitors as antidepressants and anti- strengthened by the observation that the MAO inhibitor
parkinson drugs. Although the problem of hepatotoxi- clorgyline (Johnston, 1968) could differentiate between
city, which was associated with hydrazine-derived two forms of MAO in brain and most tissues of animals
drugs, was resolved with the development of other and humans. Johnston termed these enzymes type A and
non-hydrazine MAO inhibitors, the cheese reaction type B and where clorgyline (Table 34.1, Fig. 34.3)
remained the major source of problems. was a selective irreversible inhibitor of MAO-A, an
What became known as the cheese reaction is enzyme responsible for oxidative deamination of
associated with the presence of tyramine and other norepinephrine and 5-HT. By contrast, MAO-B was

*Correspondence to: Professor Moussa B. H. Youdim, Centers of Excellence for Neurodegenerative Diseases Research,
Technion-Rappaport Family Faculty of Medicine and Institute, Efron St., PO Box 9697, Haifa 31096, Israel. Email:
youdim@tx.technion.ac.il, Tel: 972-4-8295-290, Fax: 972-4-8513-145.
94 M. B. H. YOUDIM AND P. F. RIEDERER

H2O2 O2 + H+

FAD FADH2
H
R2 + R2 OH2 C + R2
R N R N R O + HN
MAO
R1 R1 R1
Fig. 34.1. Reaction pathway of monoamine oxidase (MAO) catalyzes oxidative deamination of monoamine neurotransmitters.

(Youdim et al., 1988a). Nevertheless, the consistent


Table 34.1 cheese reaction observed with this drug in pharmacolo-
Monoamine oxidase (MAO) inhibitors with application gical and clinical studies led to its abandonment.
in depression and Parkinsons disease The reports by Youdim and colleagues (Collins
et al., 1970; Youdim et al., 1972) for the presence of
MAO type MAO-A and B in postmortem human brains led to
the suggestion that by mapping human brain MAO
AB A B
and distribution according to its isoenzymes, it would
Irreversible be possible to develop effective selective MAO inhibi-
Iproniazid AD tors directed at each form for the treatment of depres-
Phenelzine AD sion and other neuropsychiatric diseases, yet without
Isocarboxazid AD the occasional dangerous side-effects (cheese reaction)
Tranylcypromine AD inherent with older MAO inhibitors (Youdim et al.,
APD? 1972). L-Deprenyl (selegiline), the selective irreversi-
Nialamide AD
ble MAO inhibitor was the first such drug to be shown
Pargyline*
Clorgyline AD
to have MAO inhibitory activities opposite to that of
Deprenyl APD clorgyline, namely being a MAO-B inhibitor that
(selegiline) inhibited the metabolism of phenylethylamine, but
Rasagiline AD, APD? APD not 5-HT and norepinephrine, and to be devoid of the
TV3326 (brain- cheese reaction in isolated pharmacological preparations
selective, and in vivo animal studies (Knoll and Magyar, 1972).
phase II The second such inhibitor was AGN1135 and its
clinical) R-optical isomer, rasagiline (Finberg et al., 1981; Kalir
Reversible et al., 1981; Youdim et al., 2001a). Since then several
Moclobemide AD, other MAO-A and B inhibitors, such as irreversible
APD
(rasagiline) and reversible (moclobemide, lazabemide,
Brofaromine AD
Caroxazone
milacemide and safinamide) inhibitors have also been
Toloxatone AD shown not to induce a cheese reaction in animal and
BW 137OU87 human studies when used at their selective MAO-B-inhi-
Befloxatone AD bitory dosage (Table 34.1, Fig. 34.3). Yet selective irre-
Lazabemide APD versible inhibition of MAO-A is associated with the
Milacemide cheese reaction and studies of Finberg et al. (Finberg
Safinamide APD et al., 1981; Finberg and Tenne, 1982; Youdim et al.,
1988a, b) have clearly illuminated and established that
*Shows slight degree of selectivity for MAO-B. the cheese reaction is the consequence of irreversible
AD, antidepressant; APD, antiparkinsonian.
MAO-A inhibition by at least 80%, an enzyme predomi-
nantly present in the gut, portal system and peripheral
resistant to inhibition by clorgyline and metabolized and central aminergic neurons (Fig. 34.2). However,
benzylamine and, as shown later, phenylethylamine. selective irreversible MAO-B inhibitors, when used at
Tyramine and DA were equally well metabolized by concentrations that lose their selectivity for MAO-B
both forms of the enzyme (Table 34.2) (Youdim et al., and inhibit MAO-A, can also initiate such reactions. In
1988a, 2005; Cesura and Pletscher, 1992). Clorgyline vivo inhibition of MAO-A with irreversible non-selec-
was shown to increase brain levels of norepinephrine tive or irreversible selective MAO-A allows the uptake
and 5-HT and possess antidepressant activity in a series of unmetabolized tyramine into the circulation, which
of double-blind clinical trials in depressed patients eventually gains access to peripheral adrenergic neurons,
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 95

Fig. 34.2. For full color figure, see plate section. The mechanism of potentiation of cardiovascular effects of tyramine the
cheese reaction. The prevention of metabolism of tyamine by irreversible monoamine oxidase (MAO)-AB or MAO-A inhibi-
tors results in the uptake of dietary tyramine in circulation by cardiovascular adrenergic neurons, which initiates the release of
norepinephrine (NA), a substrate for inhibitied MAO-A. L-DOPA, levodopa; COMT, catechol-O-methyltransferase.

thereby releasing norepinephrine (Fig. 34.2). These stu- cheese reaction, even in excess tyramine ingestion,
dies were the impetus for the development of reversible which is far beyond doses that may be present in food
MAO-A inhibitor antidepressants lacking the ability to (Bieck et al., 1988; Da Prada et al., 1988). The lack of
cause a cheese reaction. The rationale was that in the cheese reaction by these reversible MAO-A inhibitors
case of MAO-A inhibition and tyramine ingestion, the (Table 34.1) relies on the concept that, during reversible
latter would displace the reversible inhibitor from the MAO-A inhibition, the presence of tyramine near the
enzyme active site and thus be metabolized itself by the active site of the enzyme results in the displacement
MAO-A (Fig. 34.4). However, reversible MAO-A of the inhibitor, which leads to metabolism of tyramine
(moclobemide) and B (lazabemide, milacemide and safi- by this enzyme in the body and, since tyramine does not
namide) inhibitors are devoid of the cheese reaction by cross the bloodbrain barrier, central MAO-A will
the virtue of the ability of tyramine to displace the inhibi- continue to be inhibited, thus inducing increased brain
tor from its binding site on MAO-A and to be metabo- levels of MAO-A substrates 5-HT and norepinephrine.
lized by it. Indeed, these reversible inhibitors have very The consequence of this is long-term adaptation at post-
high specificity for the enzymes they inhibit and there is synaptic sites, leading to alleviation of depression, since
little cross-over for inactivation of the other enzyme. these neurotransmitters have been implicated in the
Moclobemide, a reversible MAO-A inhibitor, was pathophysiology of depressive illness and drugs that
the first such drug to be developed and introduced into increase their functional activity induce antidepressant
clinics as an antidepressant (Da Prada et al., 1981; activity.
Korn et al., 1988; Youdim et al., 1988a, b; Haefely
et al., 1992, 1993; Priest, 1992; Paykel and Youdim, 34.3. Distribution of MAO-A and MAO-B
1993; Angst et al., 1995; Youdim, 1995). Several other in systemic organs and brain
similar drugs, such as brofaromine and befloxatone
(Table 34.1), have undergone preclinical and clinical MAO is a ubiquitous enzyme and is found in all tissues
studies. Their preclinical and clinical pharmacology in different concentrations and forms. MAO-B is more
has shown similar activity to that of moclobemide abundant, accounting for about 80% of total MAO
(Paykel and Youdim, 1993; Youdim, 1995). These activity in the human basal ganglia, which is in contrast
drugs, including moclobemide, have a limited cardio- to rat brain, where the ratio is roughly 1:1 (Table 34.3).
vascular tyramine potentiation and do not cause a In the human brain the distribution of MAO-A and
96 M. B. H. YOUDIM AND P. F. RIEDERER
CH3 CH3
O N N
CH3 H
CI CI Clorgyline Selegiline

CH3 H
N N
H3C N H H
CH CH3 H CH3 H

Methamphetamine Amphetamine

N O
N N
Rasagiline H Ladostigil H
(Agilect)

HO
NH2 NH2
N
Aminoindane Hydroxyaminoindane

N
OH NH2
NH
M30 F
Brain selective O O
MAO-AB
inhibitor
Safinamide C17H18N2O2F NW=301
Fig. 34.3. Structures of monoamine oxidase (MAO)-A, B and MAO-AB inhibitors in clinical use or under development.
Unlike selegiline, rasagiline is not metabolized to amphetamine or methamphetamine but rather to aminoindane. Ladostigil,
a cholinesterase brain-selective MAO-AB inhibitor derivative of rasagiline, is metabolized to hydroxyaminoindane, a conse-
quence of carbamate oxidation. Both ladostigil and M30 are brain-selective MAO-AB inhibitors that do not potentiate the
cardiovascular effect of tyramine (cheese effect). Safinamide is a selective reversible MAO-B inhibitor with other pharmaco-
logical attributes, including its interaction with K channel.

MAO-B differs in various regions (Collins et al., disease is detected (Adolfsson et al., 1980). Trans-
1970; OCarroll et al., 1983). Dopaminergic neurons mitter analyses of three different age groups (09.9
of the substantia nigra reveal extremely low staining years, 1059.9 years and 60 years and older) defined
of MAO. This is in contrast to noradrenergic neurons according to previous studies on ontogenesis and senes-
of the locus ceruleus, which stain positively with the cence in human brain showed a decrease in the 5-hy-
MAO-A substrate 5-HT and serotoninergic neurons droxyindole acetic acid (5-HIAA)/5-HT ratio after the
of the raphe nuclei which stain for MAO-B. Glial first decade of life. Changes in the dopaminergic system
cells stain predominantly for MAO-B, whereas astro- were seen in senescene with decreasing DA levels and
cytes contain both enzyme forms (Konradi et al., an increase in the homovanillic acid/DA ratio, whereas
1989). Nevertheless, non-selective inhibition of the dihydroxyphenylacetic acid/DA ratio was unaf-
MAO results in a highly significant increase of DA fected. Norepinephrine was not changed over the life-
in human and rodent brains regions. Furthermore, time (Konradi et al., 1992). In rodents and humans,
both reversible and irreversible MAO-A inhibitors MAO-A is present in the extraneuronal compartment
increase brain levels of 5-HT and norepinephrine, and within the dopaminergic terminals, where it is
indicating that, even though the activity of MAO in involved in the metabolism of intraneuronal and
these neurons may be low, the enzyme has a meta- released DA respectively (Fig. 34.5). Its role is to main-
bolic regulatory role. tain the neurotransmitter concentration low within the
An increase in MAO-B with age and in both pla- neuron (Youdim et al., 1988a, b; Cesura and Pletscher,
telets (Murphy et al., 1976; Bridge et al., 1985) and 1992). However, it should be remembered that DA is
human brains with Parkinsons and Alzheimers equally well metabolized by both MAO-A and B.
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 97
Table 34.2 Table 34.3
Some natural and synthetic substrates of monoamine Distribution of monoamine oxidase (MAO)-A and MAO-B
oxidase (MAO) in humans and in the brains of selected species

MAO type Total activity (%)

AB A B Tissue MAO-A MAO-B

Neurotransmitters Brain
and metabolites Human (striatum: 20 80
Epinephrine all regions)
Norepinephrine Monkey 25 75
Dopamine Guinea pig 20 80
Serotonin (5-HT) Cat 25 75
Metanephrine Pig 40 60
Normetanephrine Rat 55 45
Tele-N-methylhistamine Mice 50 50
1-methyl-4- Aminergic neurons 100 0
phenyl-4-propionoxy Glia and astrocytes 50 50
-piperidine (MPTP) Other tissues
Milacemide Liver 55 45
Trace amines, dietary (human, rat, rabbit)
amines, drugs Small intestine 75 25
Tyramine Kidney (rat) 25 75
Octopamine Lings (rat) 55 45
Phenylethylamine Human platelet 5 95
Tryptamine Chromaffin cell 5 95
Phenylethanolamine Pheochromocytoma 95 5
Synephrine PC-12 cells 95 5
Phenylephrine

Note: The above selectivity for MAO subtypes is based on determi-


nation in cell-free preparations.

FAD SRS FAD


SRS

MAO-A RIMA MAO-A--RIMA

SRS FAD
SRS FAD

Tyramine Tyramine displacement of RIMA

FAD FAD RIMA and


SRS SRS
tyramine
metabolites

Fig. 34.4. The mechanism of interaction of reversible monoamine oxidase (MAO)-A inhibitor (RIMA, moclobemide) with
monoamine oxidase, its displacement by tyramine and their eventual metabolism by MAO and drug-metabolizing enzyme
respectively.
98 M. B. H. YOUDIM AND P. F. RIEDERER

Dopamine
Glia receptor D2
Entacapone
agonists
MAO-A & B

3OMD
Selegiline
Rasagiline

COMT Bloodbrain barrier Ladostigil, M30


D2
DT
SV
SV
Tyrosine DDC
L-dopa Dopamine
L-DOPA DA
SV
MAO A AR
DDC
D1

Dopamine

MAO-A & B
Benzerezide Moclobarmide
carbidopa
Astrocyte Postsynaptic terminal
Presynaptic terminal
substantia nigra origin Striatum

Fig. 34.5. The pathway of dopamine (DA) synthesis from tyrosine via hydroxylation by tyrosine hydroxylase to levodopa
(L-dopa) and subsequent decarboxylation by dopa decarboxylase (DDC) to DA and its metabolism by interaneuronal monoa-
mine oxidase (MAO)-A and by glia and asotrocyte MAO-A and B extraneuronally. The selective inhibition of MAO by various
selective MAO-A (moclobemide) or MAO-B (selegiline, rasagiline) does not alter the steady state of striatal dopamine. On the
other hand, non-selective MAO-AB (ladostigil and M30) inhibitors induce highly significant increases in striatal dopamine and
in other brain regions. D1 and D2, dopamine receptors; DAT, dopamine transporter; AR, amine reuptake; SV, synaptic vesicle;
COMT, catechol-O-methyltransferase; 3OMD, O-methyldopa; DT, dopamine transporter; SV, vesicle.

34.4. MAO-B inhibitors in Parkinsons disease as antiparkinsonian drugs, had systemic toxicology.
However a recent addition is the antiparkinsonian
Selegiline (L-deprenyl), a selective irreversible inhibi- drug rasagiline, a selective potent irreversible MAO-B
tor of MAO-B, without the ability to cause a cheese with more than 1015-fold the potency of selegiline
reaction at its selective dosage, was identified for the (Youdim et al., 2001a; Finberg and Youdim, 2002;
treatment of PD because of the latter property and pro- Parkinson Study Group, 2002, 2004, 2005; Rascol
minence of MAO-B in extrapyramidal brain regions of et al., 2005) which, unlike selegiline, does not give rise
human brain (Birkmayer et al., 1975, 1977; Lees et al., to methamphetamine metabolites, nor does it have
1977). It has been widely used in the treatment of Par- the sympathomimetic activity of selegiline (Finberg
kinsons disease and has been shown to postpone the et al., 1981; Finberg and Youdim, 2002) or cause
need for levodopa in early Parkinsons disease and to cardiovascular complications (Youdim, and Riederer,
be useful in the management of end-of-dose akinesia 1993a; Finberg et al., 1999; Abassi et al., 2004) or
in fully developed disease (Birkmayer et al., 1975, orthostatic hypotension observed with selegiline. The
1977; Lees et al., 1977; Parkinson Study Group, orthostatic hypotension of selegiline has now been
1993; Szeleny, 1993). Since then several other reversi- shown to be related to the methamphetamine metabolite
ble and irreversible MAO-B inhibitors have been (Abassi et al., 2004). Rasagiline increases the production
developed. The reversible MAO-B inhibitors lazabe- of DA following levodopa treatment in monkeys, as mea-
mide and milacemide, which showed great promise sured by microdialysis (Finberg et al., 1998a, b) and on
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 99
chronic treatment in rats it induced increased release of metabolism in the human striatum. This is mainly
DA (Lammensdorf et al., 1996). because the effect of L-selegiline or clorgyline on DA
Safinamide is a recently developed reversible elevation in parkinsonian brains and brains obtained at
selective MAO-B inhibitor with iron channel property autopsy from depressed subjects on MAO inhibitors
that does not induce a cheese reaction (Marzo et al., (Youdim et al., 1972) is not as profound as the increase
2004). This drug has anticonvulsant activity. It im- in 5-HT and norepinephrine or the selective MAO-B
proves motor function in early Parkinsons disease substrate, phenylethylamine, observed respectively
(Stocchi et al., 2004). A median safinamide dose of with these inhibitors. These data have clearly indicated
70 mg/day (range 4090 mg/day) has been shown to that when one form of the enzyme is fully inhibited in
increase the percentage of parkinsonian patients human basal ganglia, the other MAO can equally meta-
improving their motor scores by 30% from baseline bolize DA and thus its level will not change drastically
(responders). In a subgroup of 101 patients under in the striatum (Youdim and Riederer, 1993a).
stable treatment with a single DA agonist, the addition The major reason why MAO-A inhibitors have not
of safinamide potentiated the response (47.1% respon- received attention as antiparkinsonian drugs is because
ders, mean 4.7-point motor score decrease; P  0.05). its irreversible inhibitors cause the cheese reaction.
These results suggest that doses of safinamide exerting However, the reversible MAO-A inhibitor antidepres-
ion channel block and glutamate release inhibition sant (RIMA) does not possess this side-effect
add to its symptomatic effect and warrant further (Da Prada et al., 1988; Youdim et al., 1988a, b). The
exploration of this drug in Parkinsons disease. antiparkinsonian effects of a RIMA, moclobemide,
In parkinsonian brains obtained at autopsy from when added to the standard therapy with levodopa
2 weeks or 4 years of L-selegiline-treated subjects, a and dopaminergic agonists, on motor performance of
highly significant (>3000%) and moderate (4070%) patients with idiopathic PD has been studied (Sier-
increase in phenylethylamine and DA was respectively adzan et al., 1995). The influence of the drug on the
observed in substantia nigra, caudate nucleus, putamen mood and cognitive function of non-depressed parkin-
and globus pallidus, without any changes in 5-HT, sonians has been evaluated. This was also considered
norepinephrine or their metabolite contents (Riederer valid, since 2060% of parkinsonian patients suffer
et al., 1986; Riederer and Youdim, 1986; Youdim from depression and many factors could account for
and Riederer, 1993a). By contrast, clorgyline (an this, including the reported degeneration of raphe
MAO-A inhibitor) increases both norepinephrine and nucleus 5-HT and locus ceruleus noradrenergic neu-
5-HT highly significantly with a slight increase of DA rons. Nevertheless, depression has a biological basis
in human basal ganglia and animal striatum (Youdim and in PD the main monoaminergic systems in the
et al., 1972). However, tranylcypromine, a non-selec- brain are disturbed. Indeed, besides the deficit in stria-
tive MAO inhibitor, increases the three neurotransmit- tal DA, significant reductions in locus ceruleus, nore-
ters in human and rat brain. Indeed, in rat brain pinephrine and raphe nucleus 5-HT have consistently
clorgyline, selegline and rasagiline are unable to in- been reported. The involvement of the latter two
crease striatal DA or alter its metabolism (Finberg and monoamines in the pathogenesis of depression would
Youdim, 2002). Only when both enzymes are inhibited suggest that PD may be a useful model for the study
does striatal DA increase, resulting in DA functional of depression. Recent molecular genetic studies
activity potentiation (Green et al., 1977). demonstrate that polymorphism in the 5-HT transpor-
ter, as observed in endogenous depression, is identical
34.5. MAO-A inhibitors in Parkinsons disease to that in parkinsonian patients with severe and fre-
quent episods of depression. It could be shown that
Most investigators forget that DA is equally well the 5-HT transporter gene-linked polymorphic region,
metabolized by MAO-B and -A and that the latter but not the MAO-A gene promoter-associated poly-
enzyme is located intraneuronally and regulates the morphism, may be a risk factor for depression in par-
cytoplasmic concentration of DA with neurons and kinsonian patients, whereas neither polymorphism
the extend DA is released. Nevertheless, little attention increases the risk for development of Parkinsons dis-
has been paid to MAO-A inhibition by MAO-A inhibi- ease itself (Mossner et al., 2001).
tors in the treatment of PD, even though it has clearly However, moclobemide was well tolerated by
been established that DA is equally well metabolized healthy volunteers when administered with levodopa
by MAO-A and MAO-B in the striatum (Collins and benserazide. Its safety and tolerance in parkinso-
et al., 1970; OCarroll et al., 1983). There is no indica- nians treated with levodopa and a peripheral decarbox-
tion as to what is the contribution of MAO-A or MAO- ylase inhibitor were evaluated (Sieradzan et al., 1995).
B inhibition or MAO-A compartmentalization to DA In these studies moclobemide was used as 450 mg
100 M. B. H. YOUDIM AND P. F. RIEDERER
daily dosage in parkinsonian subjects (Sieradzan et al., long-term memory. There have also been reports on
1995). More recent studies indicate that the dosage of improvement in memory and choice reaction time in the
moclobemide can be increased to 900 mg, without elderly subjects. In other studies in non-depressed elderly
unwanted side-effects, with a better clinical response people and in healthy young volunteers, moclobemide
in depressed subjects and greater efficacy for MAO-A had no significant effects on cognitive functions. Most
inhibition, and presumably the same could be true for importantly, it does not produce hypertensive reactions
PD subjects. The low dosage, as used in moclobemide- due to interaction with dietary tyramine (cheese reac-
treated PD studies (Sieradzan et al., 1995), may also tion). Numerous animal (rats and mice) studies have
explain why a relatively mild symptomatic antiparkin- shown that inhibition of MAO-A with either reversible
sonian response was obtained. Such a problem does (moclobemide) or irreversible (clorgyline) inhibitors of
not occur with L-selegiline or rasagiline, since they this enzyme induce little change in DA, suggesting that
bind covalently to the enzyme and fully inhibit at first glance DA metabolism does not appear to be
MAO-B. In this context, treatment with moclobemide affected by this inhibitor. Nevertheless, striatal microdia-
and selegiline or rasagiline should be avoided, without lysis studies in rats have shown that, after moclobemide
application of tyramine restriction. Nevertheless, sev- or clorgyline or rasagiline, the release of DA occurs,
eral studies with such combinations have not shown and this is highly significant (Haefely et al., 1992, 1993;
unwanted side-effects, whereas moclobemide can be Lamensdorf et al., 1996; Finberg et al., 1998a). This would
safely prescribed to patients on levodopa/decarboxy- indicate that, although MAO-A or B inhibition does
lase inhibitor therapy. A combination of reversible affect brain DA steady state, it affects its release. This
MAO-A (e.g. moclobemide) and reversible MAO-B may explain the antiparkinsonian effects of such drugs.
(e.g. lazabemide) inhibitor may be worth considering Thus, the increased release of DA during MAO-A inhibi-
as a therapeutic means. However, caution may need to tion may account for the prolonged duration of motor
be taken with regard to a combination of MAO inhibitors response to single levodopa challenge in response to
(even selective ones) with classical antidepressants and moclobemide (Szeleny, 1993; Sieradzan et al., 1995;
especially serotonin selective reuptake inhibitors Sternic et al., 1998). The relatively mild symptomatic
(SSRIs). Such combined treatment strategies may effect of moclobemide in PD may be associated with
provoke the serotonin syndrome, which is a serious the pharmacokinetic properties of this inhibitor. Moclo-
adverse reaction in human brains. In the treatment of bemide, being a reversible inhibitor, could be displaced
therapy-resistant depression, MAO inhibitors are an from its binding site on MAO-A present intraneuronally
important therapeutic strategy. The combined treatment and extraneuronally by DA formed from levodopa. Thus,
of MAO inhibitors with tricyclic antidepressants or disinhibition of the enzyme could occur. Haefely et al.
SSRIs should be avoided (Bijl, 2004; Izumi et al., (1992, 1993), examining the effect of moclobemide on
2006; Nieuwstraten et al., 2006) due to the possibility DA metabolism in microdialysis studies, have indeed
of initiating the deleterious serotonin syndrome. shown that DA can displace the inhibitor from its
The mild antiparkinsonian action of moclobemide MAO-A binding site. Under such conditions, namely
(Sieradzan et al., 1995) is matched by its true antide- lack of MAO-A inhibition, reductions in DA accumula-
pressant activity in several double-blind studies as tion and release could occur. Thus, it is evident that
compared with other antidepressants such as fluoxe- special attention needs to be paid to the pharmacoki-
tine, imipramine and desimipramine (Bieck et al., netics of the ratio of daily dosage of moclobemide
1988; Priest, 1992; Paykel and Youdim, 1993; Angst or other reversible MAO-A inhibitors to levodopa
et al., 1995; Youdim, 1995). Recent studies on moclo- for a more effective action of moclobemide on DA
bemides antidepressant action in depressive parkinso- metabolism and its antiparkinsonian activity.
nian subjects has shown that the drug can be a useful The studies so far done with the MAO-A inhibitor
additional antidepressant and is well tolerated with moclobemide have shown it to be well tolerated and
standard therapy. Thus, the RIMA antidepressant safe in levodopa-treated parkinsonians (Sieradzan
moclobemide could influence the function of the ami- et al., 1995). The observed shortening of latency and
nergic neurotransmitters, as well as depression in Par- prolonged duration of motor response to single-dose
kinsons disease. Moclobemide, originally developed levodopa challenge may be explained by the known
as an antidepressant, is effective in the treatment of effects of moclobemide on the metabolism of exogen-
depression. A number of studies have assessed its ous levodopa to DA and oxidative deamination by
effects on cognition, but the differences in subject MAO-A. Moclobemide seems to have a mild sympto-
samples and experimental designs render their results matic benefit in PD (Sieradzan et al., 1995; Sternic
inconclusive. In major depression, moclobemide was et al., 1998). In particular, the baseline motor condi-
found to improve vigilance, psychomotor speed and tions after overnight withdrawal of medication
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 101
improved during moclobemide treatment. The results Thus, although DA may not be directly involved in
suggest that bradykinesia may be more amenable to the pathophysiology of depression or the mechanism
moclobemide than other parkinsonian disabilities. In of the action of MAO inhibitors as antidepressants, it
non-depression parkinsonians, moclobemide does not may modulate serotonergic activity (Green and Gra-
significantly influence cognitive measures of mood. hame-Smith, 1975, Green and Youdim, 1975; Youdim
Further controlled studies are required to evaluate the et al., 1976). Thus, this may be one reason why it has
place of moclobemide and other reversible selective been suggested that a non-selective reversible MAO-
MAO-A inhibitors such as brofaromine and befloxa- A-B inhibitor that blocks DA metabolism might be
tone, which are more powerful inhibitors with greater more effective as an antidepressant than a selective
affinity for MAO-A. reversible MAO-A inhibitor, such as moclobemide,
which is thought to be a milder antidepressant and bet-
34.6. Selective MAO-B inhibitors in ter tolerated by older patients (Sternic et al., 1998;
Parkinsons disease with depressive illness Amrein et al., 1999; Erfurth and Back, 1999; Jansen
and Ballering, 1999).
Selegiline was originally developed as a psychoenergi- Depression has a biological basis of its own, but
zer antidepressant. However, earlier uncontrolled open particularly in Parkinsons disease, where the main
studies with large doses (100 mg) and later control monoaminergic systems are degenerating. It is now
studies with higher doses (> 15 mg) established a sig- well recognized that some 4060% of parkinsonian
nificant antidepressant action. However, in monother- subjects suffer from endogenous depression (Bosboom
apy and in combination with the 5-HT precursor 5- and Wolters, 2004; Lauterbach, 2004; Leentjens, 2004;
hydroxytryptophan (5-HTP), it had significant antide- Rihmer et al., 2004). Most often it is forgotten that, in
pressant activity in controlled studies (Mendlewicz addition to the deficit in striatal DA, highly significant
and Youdim, 1978, 1979, 1983; Mann and Gershon, reductions in locus ceruleus norepinephrine and raphe
1980; Mann, 1989). An antidepressant activity with a nucleus 5-HT have consistently been reported in par-
selegiline patch at high doses is reported in recent kinsonian brains. The association of diminished nora-
studies. It is most likely that at high doses, selegiline drenergic and serotoninergic neurotransmission in the
loses its MAO-B inhibitory selectivity and also inhi- pathogenesis of depressive illness would suggest that:
bits MAO-A, as has been regularly observed in animal (1) Parkinsons disease may be an ideal condition for
studies, but it bypasses system inhibition of MAO-A. the study of depression and its treatment; and (2) a
However, consideration should be given to a com- drug such as moclobemide and other RIMAs could
bination of selegiline or rasagiline with 5-HTP as anti- exhibit dual pharmacological activities as antiparkin-
depressant since three double-blind studies have sonian drug and antidepressant in the same patients
shown that the selegiline5-HTP combination is sig- suffering from the two diseases. Several studies have
nificantly more effective than 5-HTP alone in bipolar shown that in depressed parkinsonian patients moclo-
depressed subjects (Mendlewicz and Youdim, 1978, bemide is a useful antidepressant and it is well toler-
1979, 1983) and little evidence was obtained for the ated with standard therapy. The fact that RIMA can
efficacy of selegiline monotherapy at its selective be safely prescribed to parkinsonian patients on levo-
MAO-B-inhibitory concentrations as antidepressant dopa/decarboxylase inhibitor therapy may be an ideal
(Mendlewicz and Youdim, 1978). These studies situation for the symptomatic treatment of parkinso-
require confirmation. The highly significant antide- nian subjects with episodes of depression. RIMA are
pressant action of the combination of 5-HTP plus considered to be not as effective antidepressants as the
L-deprenyl (selegiline) has been attributed to its action non-selective MAO inhibitors, even though selective
in human brain on DA metabolism, where selegiline inhibition of MAO-A results in increased norepinephr-
significantly increases DA and phenylethylamine (a ine and 5-HT, with little change in DA. However,
releaser of DA) in various regions. It has been shown non-selective MAO inhibitors induce a much greater
that the functional activity of 5-HT (i.e behavioral increase in brain levels of these neurotransmitters
response in rats to increased levels of brain 5-HT) (Green and Youdim, 1975; Green et al., 1977). This
formed from tryptophan or 5-HTP is DA-dependent has been attributed to the observation that, when
(Green and Grahame-Smith, 1975; Green and Youdim, MAO-A is selectively inhibited, the intact MAO-B
1975; Youdim et al., 1976). This may explain why can continue to metabolize these amines, albeit at a
alpha-methyl-p-tyrosine, an inhibitor of tyrosine slower rate, when their elevated concentrations satisfy
hydroxylase, is able to block 5-HT functional activity the Km (Michaelis constant) of MAO-B for these
in response to the 5-HT precursors tryptophan and amines (Green and Youdim, 1975; Green et al., 1977).
5-HTP in rats (Green and Grahame-Smith, 1975). Since selective MAO-B inhibitors do not increase brain
102 M. B. H. YOUDIM AND P. F. RIEDERER
levels of the three neurotransmitters, they have limited 2003). Compartive cognitive studies with other choli-
effect in their full functional neurotransmission. Thus, nesterase inhibitors (galantamine, rivastigmine) in
if we are to understand the contribution of MAO-A and monkey have shown that it is superior to these inhibi-
B inhibition to DA metabolism and neurotransmission tors. This may be accounted for by its ability to increase
in PD and the contribution of DA neurotransmission to both acetylcholine and DA and cognitive enhancement
increased functional activity of 5-HT in the prevention by the latter neurotransmitter. Being weaker than rivas-
of depression in Parkinsons disease, there is a need for tigmine and galantamine, its toxicity is limited as com-
either non-selective reversible or brain-selective irrever- pared to the latter cholinesterase inhibitors. It retains the
sible MAO-A and B inhibitors as antidepressant antipar- neuroprotective antiapoptotic activity of rasagiline
kinsonian agents, devoid of tyramine potentiation, (Weinstock et al., 2001; Youdim and Weinstock,
namely the cheese reaction. 2001; Maruyama et al., 2003).
Ladostigil does not exhibit either MAO-A or B
34.7. Novel brain-selective irreversible inhibition in vitro or in acute in vivo studies. However,
MAO-AB inhibitors with limited tyramine on chronic treatment in mice, rats, rabbits and mon-
potentiation in the treatment of Parkinsons keys, it shows remarkable selectivity for the inhibition
disease comorbid with dementia and of both enzymes in the brain, with little effects
depressive illness on MAO-A and B in the liver and small intestine
(Weinstock et al., 2000, 2002a, b, 2003). Its in vivo
There are no profound clinical studies for the combined MAO-AB inhibitory activity is explained by the obser-
treatment with selective MAO inhibitors and acetylcho- vation that ladostigil acts as a prodrug. The oxidation
line esterase inhibitors in patients with (Alzheimers) of its carbamate cholinesterase inhibitory moiety gives
dementia or parkinsonian patients developing dementia rise to hydroxyl rasagiline that inhibits both MAO and
(Lewy body disease). However, recent studies have B and may be selectively accumulated in the aminer-
shown that patients with Lewy body disease on levo- gic neurons. This novel property explains its limited
dopa therapy for their extrapyramidal disorder respond tyramine potentiation in rats and rabbits, as compared
cognitively to anti-Alzheimer acetylcholinesterase inhi- to tranylcypromine, clorgyline and moclobemide
bitors (Benecke, 2003; Mosimann and McKeith, 2003; (Weinstock et al., 2002b). It induces highly significant
Wild et al., 2003) without loss of response to dopami- increases in norepinephrine, 5-HT and DA in the hip-
nergic antiparkinsonian drugs. A significant number of pocampus and striatum of rats and mice (Weinstock
these patients, as well as subjects with Alzheimers dis- et al., 2000; Sagi et al., 2003) and, in the forced-swim
ease and Parkinsons disease, have a predisposition to test model for antidepressant activity, it is equivalent
depression (Chan-Palay, 1992). to amitryptiline, clorgyline, tranylcypromine and
To overcome this problem, a series of neuroprotective moclobemide (Weinstock et al., 2002a). Furthermore,
bifunctional cholinesterase brain-selective MAO-AB being an MAO-AB inhibitor, similar to MAO-B inhi-
inhibitors (Weinstock et al., 2000; Youdim and Bucca- bitors (selegiline, rasagiline and lazabemide), it pre-
fesco, 2005a, b) from the pharmacophore of antiparkin- vents the striatal dopaminergic neuron degeneration
sonian drugs selegiline and rasagiline (Sterling et al., and DA depletion induced by the parkinsonism neuro-
2002) by introducing a carbamate cholinesterase toxin, N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
inhibitory moiety in such compounds to increase choli- (MPTP), in the mouse model of Parkinsons disease
nergic activity (Fig. 34.3). This was done to retain the (Sagi et al., 2003). And, unlike selegiline and rasagi-
neuroprotective and antiparkinsonian activities of these line, it highly significantly increases brain DA in
compounds. Among the compounds identified, ladostigil MPTP-treated mice above the level of control. The
([TV3326 (N-propargyl)-(3R)-aminoindan-5-yl]-ethyl fact that it inhibits both forms of MAO is a bonus for
methyl carbamate) (Fig. 34.3) (Weinstock et al., 2000; preventing metabolism of DA by either form of MAO.
Sterling et al., 2002), a derivative of rasagiline, is show-
ing remarkable pharmacological activities that may 34.8. Combined treatment of MAO inhibitors
make this compound ideal for the treatment of Parkin- with COMT inhibitors
sons disease comorbid with depressive illness and
dementia. Ladostigil is a butryl and acetylcholinesterase The combined treatment of MAO inhibitors and the
inhibitor and possesses the anti-Alzheimer drug proper- peripherally and centrally acting catechol-O-methyl-
ties of other cholinesterase inhibitors, such as rivastig- transferase (COMT) inhibitors entacapone and tolca-
mine and galantamine, by attenuating the deficit in pone has not been sufficiently evaluated in clinical
spatial learning in rats and monkeys in response to sco- studies with patients suffering from Parkinsons dis-
polamine (Weinstock et al., 2000; Buccafusco et al., ease. However, preclinical data let us assume that such
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 103
combined treatment would not be harmful because ism of 6-OHDA is believed to be associated with oxida-
catecholamines could still be metabolized in the CNS tive stress, possibly mediated by release of bound iron
by COMT activity. (II). A neuroprotective effect of high-dose selegiline
and rasagiline is suggested by the normalization of 6-
34.9. Neuroprotection OHDA-induced release of acetylcholine from striatal
slices and DA neurons (Knoll, 1987).
Selegiline and rasagiline increase neuronal survival in Selegiline, pargyline and 2-hexyl-N-methylpropargy-
a variety of in vitro and in vivo models of neural lamine (Gibson, 1987; Finnegan et al., 1990; Yu et al.,
insult, often independently of MAO-B inhibition 1994; Zhang et al., 1996) but not mofegiline (Magyar,
(for review, see Tatton and Chalmers-Redman, 1996; 1994) or the MAO-A inhibitor clorgyline (Knoll,
Foley et al., 2000; Youdim et al., 2005, 2006). These 1987) protect noradrenergic neurons in rodent hippo-
results have provoked excitement with respect to their campus and locus ceruleus against the toxin N-(2-chlor-
potential as disease-modifying agents. The neurotoxins oethyl)-N-ethyl-2-bromobenzylamine (DSP-4; toxic
MPTP and 6-hydroxydopamine (6-OHDA) selectively effect due to its derivative, the aziridinium ion), and
damage dopaminergic neurons and MPTP elicits par- selegiline reversibly inhibits DSP-4-stimulated norepi-
kinsonian syndromes in mice, goldfish and primates, nephrine release in rat hippocampus (Magyar et al.,
including humans. MPTP is both a substrate and a 1998). It is not clear whether this capacity depends on
weak, partially reversible inhibitor of MAO-B, and inhibition of MAO-B, and inhibition of monoamine
a reasonably good substrate and potent, reversible transport appears not to be involved (Hallman and Jons-
inhibitor of MAO-A. The neurotoxic mechanism of son, 1984; Yu et al., 1994). However, Magyar (1993)
MPTP is its conversion by astrocytic MAO-B to the attributed the protective effects of selegiline and its
toxin, 1-methyl-4-phenylpyridinium (MPP), a rever- p-fluoro-derivative to the prevention of aziridinium
sible inhibitor of both MAO-A and MAO-B (Singer ion uptake. Selegiline and mofegiline are also effective
and Ramsay, 1991), which enters the neuron via the against 3,4-methyenedioxymethamphetamine (MDMA)-
DA transporter (for review, see Gerlach et al., 1991). induced serotonergic toxicity in rats (Mytilineou et al.,
There are also a range of endogenous substances, such 1997a, b). Both selegiline and rasagiline, but not pargy-
as the b-carbolines and tetrahydroisoquinolines, that line, protect cultured mesencephalic dopaminergic
are metabolized to neurotoxic compounds by MAO-B neurons against glutamate receptor-mediated toxicity
(Glover and Sandler, 1993). All MAO-B inhibitors (Sprague and Nichols, 1995; Mytilineou et al., 1997b;
protect DA neurons against MPTP and such toxins Finberg et al., 1996, 1999a, b). For a review on the neu-
by inhibiting their activation and, in some cases, neu- roprotective activity of rasagiline and selegiline in other
ronal uptake. This has been demonstrated in animal in vivo models, see Youdim et al., (2003, 2005, 2006).
models for selegiline, pargyline and rasagiline at the
cellular, neurochemical and behavioral levels (for 34.9.1. Neurorescuing effects
review, see Glover and Sandler, 1993; Tatton and
Chalmers-Redman, 1994). A neuroprotective effect It is recognized that lesions of the CNS activate a
against intrastriatally injected MPP has been demon- chain of responses which result in greater damage than
strated (Wu et al., 1995, 1996). Selegiline antagonizes might be expected from the magnitude of the initial
MPP-induced apoptosis in a dopaminergic hybrid insult. These processes are only incompletely under-
cell line, and this protection is independent of MAO- stood, but it is hoped that it might be possible to slow,
B inhibition; higher concentrations (1 mM), however, halt or even reverse such processes after their initia-
induced apoptosis (Le et al., 1997), although this could tion. Such intervention is the major hope for the more
not be confirmed by Vaglini et al. (1996). Sautter effective management of central neurodegenerative
et al. (1994) reported that both rasagiline and selegi- disorders, including Parkinsons and Alzheimers dis-
line protected marmosets against the effects of MPTP. ease.
In contrast to MPTP, 6-OHDA must be applied Selegiline has been reported to protect dopaminergic
directly to the nigrostriatal system in order to elicit a neurons from MPTP-induced cell death, even when
toxic effect and its neurotoxicity does not require meta- applied 3 days after exposure to the toxin, and prevent
bolism by MAO-B for its activation. However, rasagi- the neurodegeneration of tyrosine hydroxylase-reactive
line has been shown to protect against this neurotoxin cells in the rat substantia nigra by subchronic administra-
(Blandini et al., 2004), against spontaneous central adre- tion of selegiline (Tatton and Wadia, 1991). Tatton
nergic degeneration in spontaneously hypertensive SHR coined the term neurorescuing to describe this phenom-
rats (Eliash et al., 2005) and in cerebellar granule cells enon, because at the time of selegiline administration,
(Bonneh-Barkay et al., 2005). The neurotoxic mechan- the MAO-B-catalyzed generation of MPP is completed
104 M. B. H. YOUDIM AND P. F. RIEDERER
within 36 hours of exposure to MPTP and neurodegen- (Tatton et al., 1994). Their mechanism has been attri-
eration had already commenced. The same laboratory butable to neurotrophic and antiapoptotic properties
was unable to observe this effect with other MAO-A of selegiline and rasagiline (Tatton et al., 1996; Pater-
and B inhibitors (with the possible exception of pargy- son and Tatton, 1998; Paterson et al., 1998; Maruyama
line and rasagiline (Tatton et al., 1996; Youdim et al., et al., 2004; Youdim et al., 2005). They noted that the
2006). Both selegiline and rasagiline are able to observed neuroprotective action was accompanied by
increase the survival rate of motor neurons following reduced intramitochondrial Ca2 levels and reduced
axotomy of the rat facial nerve (Salo and Tatton, cytoplasmic radical concentrations, as well as by a
1992) or subjection to spinal cord ischemia (Ravi- decrease in the DNA fragmentation characteristic of
kumar et al., 1998) of rat retinal ganglion cells follow- apoptotic cell death. The authors also recorded the
ing damage to the optic nerve (Buys et al., 1995), of altered expression of a number of cellular mRNAs
cultured mouse mesencephalic cells following with- and proteins (discussed below), and suggested that
drawal of growth factors and serum (Roy and Bdard, the ultimate result of these changes is the prevention
1993) and of cultured mesencephalic neurons from of mitochondrial swelling, a stabilization of the mito-
rodents (Koutsilieri et al., 1996) and dogs (Schmidt chondrial membrane potential (cM), thus inhibiting
et al., 1997) treated with MPP. one of the processes thought to be among the earliest
These effects are seen at concentrations where events in apoptosis (Figs. 34.6 and 34.7) (Tatton and
MAO-B inhibitors do not inhibit the enzyme and Chalmers-Redman, 1996; Akao et al., 2002a, b). In a
PC12 and SHSY-5Y cells contain only MAO-A, and further investigation, they demonstrated that a signifi-
which can be blocked by protein synthesis inhibitors cant decline in cM could be measured in nerve growth

Fig. 34.6. The mechanism of neurotoxin-induced neuronal death and its prevention by propargylamines. Mitochondria are responsi-
ble for cell survival/death via the regulation of Bcl-2 family antiaptotic (Bcl-2) and proapoptotic (Bad, Bax) proteins. Rasagiline, sele-
giline and propargylamine have been shown to induce cell survival in response to serum withdrawal or neurotoxins in neuronal cell
cultures (SHSY-5Y and PC-12) through the activation of Bcl-2 and Bcl-Xl, and the downregulation of Bad and Bax. These propargy-
lamines produce their neuroprotective activity by interacting with the mitochondrial outer membrane. The propargylamine moiety in
these inhibitors prevents neurotoxin-induced collapse of mitochondrial membrane potential, mitochondria permeability transition and
the opening of the voltage-dependent channel, as a consequence of antiapoptoic Bcl-2 family protein upregulation. The consequence
of this is the prevention of ubiquitin-proteasome inhibition, release of cytochrome c and activation of proapoptotic caspases, resulting
in cell survival. SOD, superoxide dismutase; PKC, protein kinase; AIF, apoptotic inducing factor; PT, permeability transition.
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 105

Fig. 34.7. For full color figure, see plate section. Mitochondria are responsible for cell survival/death via the regulation of BCl-2
family antiapoptotic (Bcl-2) and proapoptotic (Bad, Bax) family proteins. Rasagiline, selegiline and propargylamine have been
shown to induce cell survival in response to serum withdrawal or neurotoxins in neuronal cell cutltures (SHSY-5Y and PC-12)
via activation of Bcl-2 and Bcl-Xl and downregulation of Bad and Bax. These propargylamines produce their neuroprotective
activity by interaction with the outer-mitochondrial out membrane. They prevent neurotoxins inducing the collapse of mito-
chondrial membrane potential, mitochondrial permeability transition and the opening of the voltage-dependent channel
(VDAC) as a consequence of Bcl-2 and Bcl-Xl activation. OM, outer mitochondria; IM, inner mitochondria; GAPDH, glycer-
aldehyde-3-phosphate dehydrogenase; ROS, reactive oxygen species; HK, hexokinase; CK, creatine kinase; ANT, adenosine
nucleotide translocase; CyD, cyclophilin D; CsA, ciclosporin A; PBR, peripheral benzodiazepine receptor.

factor-deprived PC12 cells 3 hours after the withdra- Tatton and Chalmers-Redman (1996) reported that
wal of trophic support; this could be reversed by both desmethylselegiline (DMS), not selegiline, was respon-
nerve growth factor restoration and by selegiline and sible for the antiapoptotic effect in their cell culture
rasagiline (Wadia et al., 1998; Bar-Am et al., 2005). systems. Given the kinetics of selegiline metabolism
It is suggested that selegiline and rasagiline alter the and its oral bioavailibility, however, they considered
relationship between Ca2 levels and cM, perhaps it unlikely that DMS would play a major part in the
by stimulation of protein synthesis, including that of observed clinical benefits of selegiline, as oral delivery
BCL-2 (Wadia et al., 1998) (Figs. 34.6 and 34.7). allows it to exhibit only an estimated 520% of its anti-
Both rasagiline and selegiline prevent the transloca- apoptotic potential. DMS is also an MAO-B inhibitor,
tion of proapoptic glyceraldehyde-3-phosphate dehy- but less potent than selegiline (Borbe et al., 1990). It
drogenase (GAPDH) in neuronal cells from is therefore unlikely that MAO-B inhibition accounts
cytoplasm to the nucleus (Maruyama et al., 2001). for its actions. In contrast, Magyar et al. (1998) found
Paterson et al. (1990) reported that the R-isomers of DMS to be only weakly antiapoptotic in a human mel-
selegiline and 2-heptyl-N-methylpropargylamine anoma cell line. The amphetamine metabolites of sele-
inhibited cytosine arabinoside (ara C)-induced apop- giline, on the other hand, antagonize the antiapoptotic
tosis in a primary cerebellar granule neuronal culture, effects of selegiline and rasagiline (Bar-Am et al.,
but not that elicited by low K levels. It is perhaps 2004a) (Fig. 34.9), and at high concentrations are proa-
significant that ara C-, but not K-induced apoptosis poptotic (Tatton and Chalmers-Redman, 1996). Salo
is associated with impaired mitochondrial function. and Tatton (1992) compared the rescue phenomena
Rasagiline shares all the properties of selegiline in they had observed with those which can be achieved
terms of the molecular mechanisms described for with ciliary neurotrophic factor, and demonstrated
selegiline in its neuroprotection and neurorescue that astrocytic ciliary neurotrophic factor mRNA
activity (Figs. 34.6 and 34.7: see Youdim et al., expression is, in fact, upregulated by selegiline
2005 for review). Recent studies have clearly indi- (Seniuk et al., 1994). Further, Kragten et al. (1998)
cated that the neuroprotective neurorescue activity reported that the selegiline analog CGP 3466 lacks
of these propargylamine resides in the propargyl moi- MAO-B-inhibitory activity but is neuroprotective
ety, since propargylamine itself has a similar mechan- in a human neuroblastoma cell line; this activity
ism of action with almost identical potency (Weinreb was shown to be dependent on specific binding to
et al., 2004; Bar-Am et al., 2005) (Fig. 34.8). the enzyme glyceraldehyde-3-phosphatase. Thiffault
106 M. B. H. YOUDIM AND P. F. RIEDERER

Fig. 34.8. For full color figure, see plate section. The proposed schematic pathway of the neuroprotective-neurorescue activity
of rasagiline and propargylamine and their ability to process amyloid precursor protein (APP) via activation of a-secretase to
release the neuroprotective-neurotrophic soluble amyloid protein-alpha (sAPPa). Both propargylamines activate PKC and ERK
pathways and the inhibitors of PKC (GF109203X and Calpbestin) and ERK1/2 (PD98059 and UO126) prevent
their neuroprotective activity. P-PKC, pan protein kinase C; P-ERK, extracellular signal regulation kinase; BDNF, brain-
derived neurotrophic factor; GMDF, glia-derived neurotrophic factor.

1200

1000
Cell death (% of control)

*
800

** *
600 ** **
**
400

200

0
C SF R R+A R+AI S S+A A+AI A AI
Fig. 34.9. Neurorescue of partially differentiated PC-12 cells in serum-free cultures by rasagiline and selegiline and their pre-
vention by methamphetamine metabolites of selegiline. but not by aminoindan. C, control; R, rasagiline; S, selegiline; A,
methamphetamine; AI, aminoindan; SF, serum free.
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 107
et al. (1997), however, found that no long-term benefit obtained with rasagiline (Maruyama et al., 2002,
(30 days posttreatment) was affected by either selegi- 2004; Naoi et al., 2003; Youdim et al., 2003, 2005).
line or mofegiline with respect to MPTP-induced These changes were selective and did not represent a
nigral cell loss in mice. A recent similar study with general increase in cellular synthetic activity. It was
post-MPTP chronic rasagiline treatment has shown therefore proposed that the propargyl moiety of rasagi-
neurorescue of DA neurons, which has been attributed line initiates a transcriptional program which removes
to its ability to induce protein kinase C (PKC)-depen- the cell from the apoptotic pathway, probably by stabi-
dent-mitogen-activated protein (MAP) kinase path- lizing the mitochondrial membrane and reducing oxi-
way resulting from activation and translocation of dative stress (Tatton and Chalmers-Redman, 1996).
PKCa and b and phosphorylation of ERK1/2 These changes are not attributable to MAO-B inhibi-
(Yogev-Falach et al., 2003; Weinreb et al., 2004; Sagi tion, since propargylamine is a very poor MAO inhibi-
et al., 2006) (Fig. 34.8). tor. Furthermore, the recent studies with the S-optical
Rasagiline has demonstrated neuroprotective prop- isomer of rasagiline, TVP-1022, a drug that is 1000
erties in a variety of models of neuroinsult, including times less potent than MAO inhibitors, have shown
closed-head injury in mice and facial nerve axotomy that MAO inhibition is not a prerequisite for neuropro-
in newborn rats, and in several models of acute drug- tection. TVP 1022 shares a similar molecular mechan-
induced dopaminergic motor and cognition impair- ism of neuroprotection (Youdim et al., 2001b). It has
ment; selegiline was also effective in several of these now been established that the neuroprotective activity
models, but there was no correlation between the resides in the propargyl moiety, since propargylamine
potency of the two inhibitors (Mytilineou et al., has similar activity, with the same potency in neuronal
1997a, b; Speiser et al., 1998a, b). In vitro, rasagiline cell culture studies (Weinreb et al., 2004; Bar-Am
rescued nerve cells and increased the percentage of et al., 2005).
tyrosine hydroxylase-positive cells in serum depriva- Selegiline increases the number of glial fibrillary
tion-induced apoptosis (Finberg et al., 1996) and selegi- acidic protein (GFAP)-immunoreactive astrocytes in
line increased the proportion of tyrosine hydroxylase- rat striatum (Biagini et al., 1993) and subchronic treat-
positive cells without increasing total cell number, ment with selegiline lead to an increase in the number
whereas mofegiline and Ro 166491 were ineffective of GFAP- and basic fibroblast growth factor (bFGF)-
in similar experiments (Tatton et al., 1994). Both rasa- positive astrocytes following a mechanical lesion of
giline and selegiline were found to promote the survival the rat brain (Biagini et al., 1994). The proliferation
of rat E14 mesencephalic dopaminergic cells in culture, of such cells is normally associated with CNS injury,
but not of co-cultured GABAergic cells; rasagiline, and represents the first stage of the neurotrophin-
but not selegiline, also displayed this effect under mediated repair process. Revuelta et al. (1997) treated
serum-free conditions (Finberg et al., 1998a, b). rats with selegiline 3 weeks before and 3 weeks after
The effects of the rasagiline metabolite L-aminoin- the unilateral transection of the medial forebrain bun-
dane, only observed at high doses, were distinct from dle; although the density of GFAP-positive astrocytes
those of rasagiline itself (Speiser et al., 1997). L-Ami- was elevated in the drug-treated animals in both the
noindane has no amphetamine-like properties, but its lesioned and unlesioned substantia nigra (but not stria-
structural similarity to benzylamine suggests that it tum), the treatment did not reduce the axotomy-
may be a reversible MAO-B inhibitor (Fowler et al., induced degeneration of the nigrostriatal system.
1980). Unlike the methamphetamine metabolite of sele- In dopaminergic cell cultures exposed to MPP,
giline, which interferes with neuroprotective and neuror- high-dose selegiline (110 M), administered on the
escue activity of rasagiline and selegiline, such an effect day following toxin exposure, improved cell survival
is not seen with aminoindane and aminoindane may have rate significantly, possibly via stimulation of neurotro-
neuroprotective activity (Bar-Am et al., 2004b). phin synthesis (Koutsilieri et al., 1996). Selegiline is
Tatton and Chalmers-Redman (1996) reported that also reported to stimulate synthesis of interleukins-1
selegiline alters the expression of some 50 or more and -6 in cultured peripheral blood mononuclear cells
glial and neuronal proteins whose synthesis is modu- from healthy volunteers (Wilfried et al., 1997; Muller
lated. Of these, 10 have been identified: the levels of et al., 1998). Elevated cerebrospinal fluid levels of
both forms of superoxide dismutase, glutathione per- these trophins have been reported in untreated de novo
oxidase, tyrosine hydroxylase, BCL-2, BCL-XL and Parkinsons and Alzheimers disease patients (Blum-
c-FOS mitochondrial NADH dehydrogenase were Degen et al., 1996), suggesting that their increased
increased; however the levels of BAX and c-JUN, synthesis may be an early response to the underlying
two proteins associated with the initiation of apoptosis, disease process. Furthermore, selegiline and rasagiline
were decreased. Very similar results have also been elevated the nerve growth factor GDNF and BDNF
108 M. B. H. YOUDIM AND P. F. RIEDERER
concentration in an astrocyte and SHSY-5Y cell cul- symptomatic control of parkinsonism the safety profile
tures (Semokova et al., 1996; Maruyama et al., 2004). of selegiline (510 mg/day) is good and no precaution is
necessary at these dosages. Clinical studies designed to
34.10. Other properties of MAO-B inhibitors identify clinical neuroprotection point to the prevention
of disease progression. This conclusion was drawn from
Selegiline suppresses the degradation of putrescine, a key an evidence-based review (Goetz et al., 2002) of the
molecule in the regulation of polyamine synthesis. Poly- Movement Disorder Society, in which levels of therapeu-
amines, such as spermine and spermidine, are stored and tic measures in Parkinsons disease were analyzed.
synthesized in brain microglia, and are believed to play a Tables 34.434.6 (from Riederer et al., 2004) give a
number of roles in the CNS (for review, see Zappia and closer view on prospective, randomized, double-blind
Pegg, 1988), including the modulation of N-methyl-D- and placebo-controlled studies with 510 mg/day selegi-
aspartate (NMDA) receptor activity (Williams et al., line monotherapy in Parkinsons disease (Table 34.4), of
1991), whose activation plays a central role in excito- selegiline as an adjunct to levodopa (Table 34.5) and sele-
toxic damage to the CNS. The N1-acetylated versions gilines efficacy for clinical neuroprotection in Parkin-
of these polyamines are themselves good MAO-B sons disease (Table 34.6). Forty years of experience
substrates; the suppression of the metabolism of these with selegiline in the treatment of Parkinsons disease
precursors and of putrescine should therefore lead to have indicated that selective inhibition of MAO is a useful
diminished sensitivity of the NMDA receptor, and thus therapeutic concept for both the early and advanced
offer a direct connection between MAO-B inhibition phases of this progressive disorder. In addition, basic
and neuroprotection by selegiline and rasagiline (Youdim research points to the efficacy of selegiline as a possible
and Riederer, 1993b). neuroprotective drug. However, this could not be demon-
strated by the state-of-the-art clinical trials designed at the
34.11. Clinical aspects of MAO-B inhibitors
in Parkinsons disease
Table 34.4
The major clinical indications of selective MAO-A Symptomatic efficacy of selegiline monotherapy
inhibitors are for depressive disorders, anxiety disorders in Parkinsons disease
and, more recently, Parkinsons disease. In addition, the
selective reversible MAO-A inhibitor moclobemide has Author (year) N Results
shown minor indications for attention-deficit hyperac- Parkinson Study 800 Motor UPDRS (1 vs 3 months)
tivity syndrome, smoking cessation and cognitive Group (1989) Placebo 16.8 / 17.5
deficits in dementia. By contrast, selective MAO-B Selegiline 15.7 / 15.8
inhibitors have primarily found use in Parkinsons dis- Myllyla et al. 52 Disability significant less in the
ease and there may be some indications in Alzheimers (1992) selegiline group up to 12
disease. Nevertheless, high dosage of selegiline, which months
loses its selectivity and inhibits MAO, has been Allain et al. 93 Significant better motor UPDRS
reported to possess antidepressant activity. (1993) and depression scores at 3
The clinical applications of MAOIs in depressive dis- months in the selegiline group
Mally et al. 20 Significant change in motor
orders and anxiety disorders have been reviewed in
(1995) behavior and daily activity
detail by Riederer et al. (2004): the focus here is on the (UPDRS) after 3 weeks at 10
clinical use of the new irreversible type B-MAO inhibi- mg selegiline. Total UPDRS
tor rasagiline. Also some comparison has been made to scores and North Western
selegiline, which has been in clinical use since 1975. ratings were changed
significantly after 4 weeks.
34.11.1. Selegiline Greatest changes in walking
and in hypokinesia, rigidity
Selegilines potential as an antiparkinson drug has been were not modified by selegiline
reviewed in numerous studies (Riederer and Lachen- Palhagen et al. 157 Change in total UPDRS at 6
mayer, 2003; Riederer et al., 2004). From these clinical (1998) weeks and 3 months:
Placebo 25.3 / 1.75.4
studies it has been concluded that selegiline is efficacious
Selegiline 0.45.0 / 15.3
as monotherapy and that there is evidence for a sympto-
matic effect as adjunct therapy (Goetz et al., 2002). There Prospective, randomized, double-blind, placebo-controlled studies.
is, however, insufficient evidence for the ability of selegi- UPDRS, Unified Parkinsons Disease Rating Scale.
line to prevent or control motor complications. In the From Riederer et al. (2004).
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 109
Table 34.5
Selegiline as adjunct to levodopa (symptomatic efficacy)

Author (year) N Results

Przuntek and Kuhn 21 Significant improvement in CURS and Schoppe motor performance series in 2/3 of cases
(1987) on adjunct selegiline
Sivertsen et al. (1989) 38 No significant difference in total CURS and scores for rigidity and funcitonal performance
Larsen et al. (1999) 163 Patients treated with levodopa selegiline developed markedly less severe parkinsonism
and required lower doses of levodopa during the 5-year study period
Przuntek et al. (1999) 116 Over 5 years, only a slight rise of levodopa dose in the selegiline group, but marked rise in
the placebo group. Equal therapeutic efficacy in the selegiline group with lower
levodopa dose
Shoulson et al. (2002) 368 During 2-year follow-up:
Selegiline: 34% dyskinesias, 16% freezing,
change in total UPDRS 7.0612.7
Placebo: 19% dyskinesia, 29% freezing,
change in total UPDRS 1.5110.4

Prospective, randomized, double-blind, placebo-controlled studies.


CURS, Columbia University Rating Scale; UPDRS, Unified Parkinsons Disease Rating Scale.
From Riederer et al. (2004).

Table 34.6
Neuroprotection in Parkinsons disease by selegiline

Authors (year) Name of the study N Result

Tetrud and Langston (1990) Pilot study for 54 Endpoint (levodopa)


DATATOP Placebo 312. days
Selegiline 548.9 days
Parkinson Study Group DATATOP 800 Endpoint (levodopa)
(1993a, b, 1989) After 12 months:
Placebo 47%
Selegiline 26%
Myllyla et al. (1992) Finnish trial 47 Endpoint (levodopa)
Placebo 37228 days
Selegiline 54590 days
Allain et al. (1993) French selegiline 93 Endpoint (levodopa)
multicentre trial After 3 months:
Placebo 18.4%
Selegiline 4.5%
Olanow et al. (1995) SINDEPAR 101 Deterioration in UPDRS between
baseline and final visit (14 months)
Placebo 5.81.4 points
Selegiline 0.41.3 points
Przuntek et al. (1999) SELEDO 116 Primary endpoint: need for >50%
increase in levodopa dose
Placebo 2.6 years
Selegiline 4.9 years

UPDRS, Unified Parkinsons Disease Rating Scale.


From Riederer et al. (2004).
110 M. B. H. YOUDIM AND P. F. RIEDERER
time selegiline was being studied (Riederer and Lachen- cardia or other cardiovascular adverse reactions (Marek
mayer, 2003). et al., 1997) and side-effects are no more frequent than
with placebo.
34.11.2. Zydis selegiline Based on these data, a controlled trial of rasagiline in
early Parkinsons disease, the TEMPO study (Parkinson
Zydis selegiline is a new galenic form of selegiline Study Group, 2002) has been performed. The goal of this
HCl, a freeze-dried product which is taken as a melt clinical examination was to demonstrate the efficacy,
tablet (Clarke et al., 2003a). The reason for develop- safety and tolerability of rasagiline in untreated patients
ing this drug derives from the fact that selegiline is a with early Parkinsons disease by conducting a rando-
sympathomimetic agent and it is administered orally. mized, placebo-controlled and double-blind trial over 6
It undergoes a first-pass metabolization in the liver, months. A first report was published by the Parkinson
which leads to 90% breakdown of the parent com- Study Group in 2002. A total of 138 patients entered
pound into its metabolites amphetamine and metam- the study taking placebo, whereas 134 were receiving 1
phetamine. These metabolites and selegiline itself are mg/day rasagiline and another 132 were receiving 2
thought to be responsible for its side-effects cardio- mg/day monotherapy respectively. In the placebo group
vascular disturbances (Churchyard et al., 1997). This 112 patients completed 26 weeks without additional
may not be a selegiline-specific problem according to therapy: this figure was 111 in the group receiving 1
the numerous clinical publications over the last 40 mg/day rasagiline and 105 patients in the group on 2
years (Birkmayer et al., 1985, Gerlach et al., 2003). mg/day rasagiline. The TEMPO study clearly demon-
Nevertheless it is important to avoid amphetamine strated that rasagiline monotherapy was effective in this
and metamphetamine cardiovascular effects (Abassi 26-week clinical trial. Table 34.7 shows this significant
et al., 2004). Zydis selegiline, in a dose of 1.25 mg/ improvement in UPDRS scores and subscales for rasagi-
day, avoids the first-pass breakdown and produces a line at 1 and 2 mg/day versus placebo. Table 34.8 gives
plasma concentration to inhibit irreversibly platelet evidence that there were no adverse events experienced
MAO-B. This inhibition corresponds to the effect of by either treatment group.
10 mg selegiline. Zydis selegiline is well tolerated and In a second phase of that trial (Parkinson Study
reduces Unified Parkinsons Disease Rating Scale Group, 2004) subjects randomized to 1 or 2 mg/day
(UPDRS) scores significantly within 12 weeks of start- of rasagiline continued to receive this dosage, whereas
ing treatment (Clarke et al., 2003b). Double-blind, mul- patients previously taking placebo received rasagiline
ticenter trials show that Zydis selegiline significantly at 2 mg/day. This clinical design is named delayed-
reduces daily off time at 46 weeks of treatment with onset clinical trial and considers symptomatic effi-
1.25 mg dose by 9.9% (P 0.003) and at 1012 weeks cacy of a drug as well as possible disease-modifying
with the 2.5 mg dose by 13.2% (P < 0.001). The total related activity to neuroprotective effect (Leber,
number of off hours could be reduced by 2.2 hours/ 1997). This study demonstrated that rasagiline is effec-
day at week 12 from baseline, whereas this value was tive even 1 year after start of treatment. Subjects treated
only 0.6 hours/day in the placebo group. Dyskinesia- with rasagiline, 2 and 1 mg/day, for 12 months showed
free time increased by 1.8 hours/day at week 12. There less functional decline than subjects whose treatment
was no difference in adverse reactions between the was delayed for 6 months, suggesting its possible dis-
patient groups receiving Zydis selegiline and placebo ease-modifying activity (Table 34.9). Again, there was
(Waters et al., 2004). no evidence for group differences with regard to
adverse events (Parkinson Study Group, 2004).
34.11.3. Rasagiline (Azilect, Agilect) In the PRESTO study (Parkinson Study Group,
2005), safety, tolerability and efficacy of rasagiline
In a phase II evaluation, rasagiline mesylate has been in levodopa-treated parkinsonian patients and motor
administered to de novo parkinsonian patients in a fluctuations have been studied. A total of 472 patients
10-week, placebo-controlled and randomized trial were enrolled with at least 2.5 hours off time each day,
using dosages of up to 4 mg/day. Specific attention despite being on optimized antiparkinsonian therapy.
has been given to cardiovascular parameters, since this In this multicenter, randomized placebo-controlled,
inhibitor, unlike selegiline, has no sympathomimetic double-blind, parallel-group clinical trial, rasagiline
activity and is metabolized to aminoindan rather than was used in doses of 1.0 or 0.5 mg/day in comparison
amphetamine derivatives (Abassi et al., 2004). Rasagi- to placebo. Outcome measures were changed from
line is 1015 times more active than selegiline and was baseline in total daily off time during the 26-week
well tolerated in all doses (0.52 mg/day) used. There treatment, in the percentage of patients completing
was no evidence of side-effects like hypertension, brady- the 26-week trial and in adverse event frequency.
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 111
Table 34.7
Primary analysis of changes between baseline and 26 weeks

Effect size (95% confidence interval)

Characteristic Rasagiline 1 mg/day groups vs placebo Rasagiline 2 mg/day group vs placebo

Total UPDRS score 4.20 (5.66 to 2.73) 3.56 (5.04 to 2.08)


UPDRS motor subscale 2.71 (3.86 to 1.55) 1.68 (2.84 to 0.51)
ADL subscale 1.04 (1.60 to 0.48) 1.22 (1.78 to 0.65)
Mental subscale 0.14 (0.44 to 0.15) 0.26 (0.56 to 0.04)
PIGD subscale 0.15 (0.41 to 0.11) 0.20 (0.46 to 0.06)
Rigidity 0.38 (0.80 to 0.039) 0.39 (0.81 to 0.02)
Tremor 0.63 (1.03 to 0.23) 0.38 (0.78 to 0.02)
Bradykinesia 1.51 (2.19 to 0.82) 0.77 (1.47 to 0.08)
Schwab and England ADL stage 0.77 (0.42 to 1.96) 0.39 (0.81 to 1.58)
Hoehn and Yahr stage 0.04 (0.13 to 0.04) 0.04 (0.13 to 0.04)
PDQUALIF scale 2.91 (5.19 to 0.64) 2.74 (5.02 to 0.45)
Beck Depression Inventory 0.35 (0.86 to 0.16) 0.21 (0.72 to 0.30)
Timed motor score 0.55 (1.19 to 0.08) 0.36 (1.00 to 0.28)

UPDRS, Unified Parkinsons Disease Rating Scale; ADL, activities of daily living; PIGD, postural instability/gait disorder; PDQUALIF,
Parkinsons Disease Quality of Life.
Primary outcome variable adjusted according to the primary model.
Center-treatment interaction included if significant.
Reproduced from Parkinson Study Group (2002), with permission from Archives of Neurology.

Table 34.8
Adverse events by treatment group*

Placebo group Rasagiline 1 mg/day Rasagiline 2 mg/day Combined rasagiline


Adverse events (n 138) group (n 134) group (n 132) groups (n 266)

Any event 110 (79.7) 109 (81.3) 111 (84.1) 220 (82.7)
Any event (moderate or 63 (45.7) 58 (43.3) 60 (45.5) 118 (44.4)
severe intensity)
Infection 22 (15.9) 20 (14.9) 21 (15.9) 41 (15.4)
Headache 14 (10.1) 19 (14.2) 16 (12.1) 35 (13.2)
Accidental injury 14 (10.1) 10 (7.5) 10 (7.6) 20 (7.5)
Dizziness 15 (10.9) 9 (6.7) 10 (7.6) 19 (7.1)
Astheniay 15 (10.9) 6 (4.5) 6 (4.5) 12 (4.5)
Nausea 10 (7.2) 7 (5.2) 9 (6.8) 16 (6.0)
Arthralgia 6 (4.3) 5 (3.7) 14 (10.6) 19 (7.1)
Back pain 7 (5.1) 7 (5.2) 8 (6.1) 15 (5.6)
Pain 8 (5.8) 8 (6.0) 6 (4.5) 14 (5.3)

*Data are presented as the number (percentage) of patients. Between-groups differences were not statistically significant, unless otherwise indicated.
yP 0.03 for the difference between placebo and combined treatment groups; P 0.05, difference between placebo and each of the individual
treatment groups.
Reproduced from Parkinson Study Group (2002), with permission from Archives of Neurology.

In general, the efficacy of rasagiline at 1 mg/day the 1 mg/day patient group. UPDRS subscores were
was greater than within the patient group receiving significant with respect to improvement of activities
0.5 mg/day. However, the primary outcome measure, of daily living during off time, motor performance
reduction of daily off time, could be reached in both during on time, rigidity and tremor in both treatment
treatment groups, although it was much better in groups.
112 M. B. H. YOUDIM AND P. F. RIEDERER
Table 34.9
Change from baseline in efficacy variables between the 371 subjects receiving 6 months and 1 year of treatment*

Rasagiline, 1 mg/day vs delayed rasagiline, Rasagiline, mg/day, vs delayed rasagiline,


Variable 2 mg/day 2 mg/day

UPDRS
Totaly 1.82 (3.64 to 0.01){ 2.29 (4.11 to 0.48)}
Motory 1.06 (2.47 to 0.34) 0.99 (2.39 to 0.41)
ADL 0.48 (1.15 to 0.19) 0.96 (2.39 to 0.29)k
Mentaly 0.16 (0.09 to 0.42) 0.07 (0.33 to 0.19)
Hoehn and Yahr scale score 0.08 (0.01 to 0.17) 0.04 (0.05 to 0.13)
Schwab and England scale score 0.21 (1.47 to 1.04) 0.15 (1.41 to 1.11)

*Data are given as effect size (95% confidence interval). Rasagiline was administered as rasagiline mesylate.
yThe model used to determine effect size includes a treatment  center interaction.
{P 0.05.
}P 0.01.
kP 0.005.
Reproduced from Parkinson Study Group (2004), with permission from Archives of Neurology.

Bradykinesia and dyskinesia improved in only the 1 addition, rasagiline is well tolerated in patients older
mg/day group. With regard to side-effects, there was a than 70 years. There is no evidence for typical side-
significant increase at 1 mg/day rasagiline in weight effects of dopaminergic treatment such as daytime som-
loss, anorexia and vomiting. There were no group dif- nolence, leg edema or nausea. Treatment with rasagiline
ferences with regard to blood pressure or pulse rate. is simple once daily without the need for titration.
The most common serious side-effects in all three Rasagiline is easy to handle as an adjunct to levodopa.
groups were related to accidental injury (n 6), arthri- In conclusion, rasagiline may be a relatively ideal
tis, worsening Parkinsons disease, stroke, melanoma drug once a day for the treatment of Parkinsons dis-
(n 3) and urinary tract infection (n 3). One patient ease both in the early stages as monotherapy and in
was identified as having a melanoma before initiating advanced stages in combination with levodopa. Its
study medication and its occurrence is no more than efficacy is equal to that of entacapone. Rasagiline
is seen with levodopa. reduces significantly off-time periods in patients with
A further clinical trial was set up to demonstrate the motor fluctuations and improves daily on time without
effectiveness of rasagiline as an adjunct to levodopa. troublesome dyskinesias. The drug is well tolerated
The Lasting effect in Adjunct therapy with Rasagiline and does not show adverse events, even after long-
Given Once daily (LARGO) study was an 18-week, term treatment. Rasagiline has the potential to demon-
randomized, placebo-controlled, double-blind, double- strate clinical neuroprotection as assumed from the
dummy, parallel-group multicenter trial. A total of 687 1-year delayed-onset clinical trial.
outpatients were randomly assigned to oral rasagiline:
231 individuals received 1 mg once daily, 227 received 34.12. Conclusion
entacapone, a peripheral COMT inhibitor, 200 mg with
every levodopa dose and 229 individuals were on pla- After more than 30 years of experience with MAO and
cebo. Primary outcomes were change in daily off time, its inhibitors, we are now in a better position to under-
clinical global improvement and UPDRS scores (Rascol stand the importance of this enzyme in brain function
et al., 2005). Table 34.10 shows that rasagiline improves and how MAO inhibitors alter the function of central
daily off time significantly and at the same rate as enta- actions of monoaminergic neurotransmitters norepi-
capone. In addition, daily on time without troublesome nephrine, 5-HT and DA. The experiences gained with
dyskinesia improved significantly in both groups at selegiline (L-deprenyl), rasagiline and moclobemide
similar rates. UPDRS secondary analyses and ancillary as antiparkinsonian drugs and other RIMAs have
study efficacy assessment showed equipotency of rasagi- shown that we can synthesize selective MAO-A or
line and entacapone adjunct to levodopa (unpublished MAO-B inhibitors directed at the active sites of each
data). There were no differences whatsoever between enzyme, and such drugs are devoid of the unwanted
placebo-, rasagiline- and entacapone-treated individuals hazardous side-effects associated with the earlier
with regard to adverse events (Rascol et al., 2005). In MAO inhibitors. The many clinical experiences have
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE
Table 34.10
Primary and associated efficacy assessments

Adjusted mean change from baseline to treatment (SE)

Rasagilin Entacapone Placebo Difference, rasagiline vs Difference, entacapone


(n 222) (n 218) (n 218) placebo (95% CI) P vs placebo (95% CI) P

Daily off time (h) 1.18 (015) 1.20 (015) 0.40 (015) 0.78 (1.18 to 0.39) 0.0001 0.80 (1.20 to 0.41) < 0.0001
Daily on time without troublesome dyskinesia (h) 0.85 (0.17) 0.85 (0.17) 0.03 (017) 0.82 (0.36 to 1.27) 0.0005 0.82 (0.36 to 1.27) 0.0005
Daily on time with troublesome dyskinesia (h) 0.23 (0.13) 0.18 (0.13) 0.14 (0.13) 0.09 (0.28 to 0.46) 0.6209 0.04 (0.32 to 0.41) 0.8157
Responder rate (number [%])* 113 (51%) 99 (45%) 70 (32%) 2.5y (1.62 to 3.85) < 0.0001 2.0y (1.29 to 3.06) 0.0019

Assessments measured by entries in 24-h diaries. Off-time period of poor overall function (i.e. increasing signs of Parkinsons disease). On-time period of good overall function and mobility.
*Responders were defined as patients showing an improvement of 1 h or more in the change from baseline in mean total daily off time.
yOdds ratio.
From Rascol et al. (2005).

113
114 M. B. H. YOUDIM AND P. F. RIEDERER
also educated us how to use these drugs, for an ade-
Adolfsson R, Gottfries CG, Oreland L et al. (1980). Increased
quate period and with the right dosage these were activity of brain and platelet monoamine oxidase in
the features that contained the inherent problems in dementia of Alzheimer type. Life Sci 27: 10291034.
earlier studies with non-selective MAO inhibitors. Akao Y, Maruyama W, Yi H et al. (2002a). An anti-Parkin-
The present experiences with reversible MAO-A inhi- sons disease drug, N-propargyl-1(R)-aminoindan (rasagi-
bitors, especially moclobemide, are demonstrating line), enhances expression of anti-apoptotic bcl-2 in
remarkable activities in different clinical settings. human dopaminergic SH-SY5Y cells. Neurosci Lett
The most prominent aspect of these is that moclobe- 326 (2): 105108.
mide is a true antidepressant with few side-effects, is Akao Y, Maruyama W, Shimizu S et al. (2002b). Mitochon-
well tolerated and shows clinical antidepressant effi- drial permeability transition mediates apoptosis induced
by N-methyl(R)salsolinol, an endogenous neurotoxin,
cacy and has antiparkinsonian activity. Because rever-
and is inhibited by Bcl-2 and rasagiline, N-propargyl-1
sible MAO-A inhibitors such as moclobemide are
(R)-aminoindan. J Neurochem 82 (4): 913923.
showing remarkably safe action and relatively low Allain H, Pollak P, Neukirch HC (1993). Symptomatic effect
incidents of side-effects, it is being considered in the of selegiline in de novo parkinsonian patients. The French
treatment of mental functions in Parkinsons and Alz- Selegiline Multicenter Trial. Mov Disord 8(Suppl 1):
heimers diseases, in which a significant number of 3640.
these subjects (4060%) suffer from endogenous Amrein R, Martin JR, Cameron AM (1999). Moclobemide in
depression. However, RIMAs are considered as mild patients with dementia and depression. Adv Neurol 80:
antidepressants and may be useful for the treatment 509519.
of subtypes of major depressions. The availability of Angst J, Amrein R, Stabl M (1995). Moclobemide and tricyclic
the novel brain-selective MAO-AB inhibitor ladosti- antidepressants in severe depression: meta analysis and
prospective studies. J Clin Psychopharmacol 15: 16S23S.
gil, with limited tyramine potentiation and equivalent
Bar-Am O, Yogev-Falach M, Amit T et al. (2004a). Regulation
or better than RIMA, will indicate whether MAO-AB
of protein kinase C by the anti-Parkinson drug, MAO-B
inhibition would be more effective than antiparkinso- inhibitor, rasagiline and its derivatives, in vivo. J Neurochem
nian and antidepressant drugs than the selective 89 (5): 11191125.
MAO-A or B inhibitors so far examined in PD. Bar-Am O, Amit T, Youdim MB (2004b). Contrasting
Furthermore, recent clinical studies with cholinester- neuroprotective and neurotoxic actions of respective
ase inhibitors have demonstrated that introduction of metabolites of anti-Parkinson drugs rasagiline and selegi-
such drugs in Lewy body disease patients does not line. Neurosci Lett 355 (3): 169172.
interfere with their response to levodopa antiparkinso- Bar-Am O, Weinreb O, Amit T et al. (2005). Regulation of
nian drug therapy. However, the therapeutic effects of Bcl-2 family proteins, neurotrophic factors, and APP pro-
MAO-A or B inhibitors in Lewy body disease have not cessing in the neurorescue activity of propargylamine.
FASEB J 19 (13): 18991901.
been investigated. Thus, therapeutically there may be
Benecke R (2003). Diffuse Lewy body diseasea clinical
a greater advantage with the use of a drug such as
syndrome or a disease entity? J Neurol 250 (Suppl 1):
ladostigil, which has multiple pharmacological I39I42.
actions, namely the ability to inhibit both forms of Biagini G, Zoll M, Fuxe K et al. (1993). L-deprenyl increases
MAO and cholinesterase, related to the comorbidity GFAP immunoreactivity selectively in inactivated astro-
of neurological neurodegenerative diseases with cytes in rat brain. Neuroreport 5: 955958.
depressive illness. Biagini G, Frasoldati A, Fuxe K et al. (1994). The concept of
astrocyte-kinetic drug in the treatment of neurodegenera-
Acknowledgments tive diseases: evidence for Ldeprenyl-induced activation
of reactive astrocytes. Neurochem Int 25: 1722.
This work was supported by National Parkinson Foun- Bijl D (2004). The serotonin syndrome. Neth J Med 62 (9):
dation (Miami, USA), Michael J. Fox foundation (New 309313.
York), Golding Parkinson Research Fund and Eve Birkmayer W, Riederer P, Youdim MB et al. (1975). The
Topf Neurodegenerative Diseases Center (Technion, potentiation of the anti akinetic effect after L-dopa treat-
ment by an inhibitor of MAO-B, Deprenil. J Neural
Haifa). We are grateful for their support.
Transm 36: 303326.
References Birkmayer W, Riederer P, Ambrozi L et al. (1977). Implications
of combined treatment with Madopar and L-deprenil in
Parkinsons disease. A long-term study. Lancet 1: 439443.
Abassi ZA, Binah O, Youdim MB (2004). Cardiovascular
Birkmayer W, Knoll J, Riederer P et al. (1985). Increased
activity of rasagiline, a selective and potent inhibitor of
life expectancy resulting from addition of L-deprenyl to
mitochondrial monoamine oxidase B: comparison with
Madopar treatment in Parkinsons disease: a longterm
selegiline. Br J Pharmacol 143 (3): 371378.
study. J Neural Transm 64 (2): 113127.
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 115

Blandini F, Armentero MT, Fancellu R et al. (2004). Eliash S, Shteter N, Eilam R (2005). Neuroprotective effect
Neuroprotective effect of rasagiline in a rodent model of of rasagiline, a monoamine oxidase-B inhibitor, on sponta-
Parkinsons disease. Exp Neurol 187 (2): 455459. neous cell degeneration in a rat model. J Neural Transm
Blum-Degen D, Muller T, Kuhn W et al. (1996). Interleukin- 112 (8): 9911003.
1 beta and interleukin-6 are elevated in the cerebrospinal Erfurth AB, Back T (1999). Severe therapy refractory
fluid of Alzheimer and de novo Parkinsons disease depression as initial mainstream of Parkinsons disease.
patients. Neurosci Lett 202: 1720. Psychiatr Prax 26: 4647.
Bonneh-Barkay D, Ziv N, Finberg JP (2005). Characteriza- Finberg JP, Tenne M (1982). Relationship between tyramine
tion of the neuroprotective activity of rasagiline in potentiation and selective inhibition of monoamine oxi-
cerebellar granule cells. Neuropharmacology 48 (3): dase types A and B in the rat vas deferens. Br J Pharmacol
406416. 77 (1): 1321.
Borbe HO, Niebch G, Nickel B (1990). Kinetic evaluation of Finberg JP, Youdim MB (2002). Pharmacological properties of
MAO-B-activity following oral administration of selegi- the anti-Parkinson drug rasagiline; modification of endogen-
line and desmethyl-selegiline in rat. J Neural Transm ous brain amines, reserpine reversal, serotonergic and dopa-
Suppl 32: 131137. minergic behaviours. Neuropharmacology 43 (7): 11101118.
Bosboom JL, Wolters ECh (2004). Psychotic symptoms in Finberg JP, Tenne M, Youdim MB (1981). Tyramine antag-
Parkinsons disease: pathophysiology and management. onistic properties of AGN 1135, and irreversible inhibitor
Expert Opin Drug Saf 3 (3): 209220. of monoamine oxidase type B. Br J Pharmacol 73: 6574.
Bridge TP, Soldo BJ, Phelps BH et al. (1985). Platelet mono- Finberg JP, Lamensdorf I, Commissiong JW et al. (1996).
amine oxidase activity: demographic characteristics con- Pharmacology and neuroprotective properties of rasagi-
tribute to enzyme activity variability. J Gerontol 40: line. J Neural Transm Suppl 48: 95101.
2328. Finberg JP, Wang J, Bankiewicz K et al. (1998a). Increased
Buccafusco JJ, Terry AV Jr, Goren AVJr et al. (2003). Potential striatal dopamine production from L-DOPA following
cognitive actions of (n-propargly-(3r)-aminoindan-5-yl)- selective inhibition of monoamine oxidase B by R()-N-
ethyl, methyl carbamate (tv3326), a novel neuroprotective propargyl-1-aminoindan (rasagiline) in the monkey.
agent, as assessed in old rhesus monkeys in their perfor- J Neural Transm Suppl 52: 279285.
mance of versions of a delayed matching task. Neuroscience Finberg JP, Takeshima T, Johnston JM et al. (1998b).
119 (3): 669678. Increased survival of dopaminergic neurons by rasagiline,
Buys YM, Trope GE, Tatton WG (1995). ()-Deprenyl a monoamine oxidase B inhibitor. Neuroreport 9: 703707.
increases the survival of rat retinal ganglion cells after optic Finberg JP, Lamensdorf I, Weinstock M et al. (1999). Pharma-
nerve crush. Curr Eye Res 14: 119126. cology of rasagiline (N-propargyl-1R-aminoindan). Adv
Cesura AM, Pletscher A (1992). The new generation of mono- Neurol 80: 495499.
amine oxidase inhibitors. Prog Drug Res 38: 171297. Finnegan KT, Skratt JJ, Irwin I et al. (1990). Protection against
Chan-Palay V (1992). Depression and senile dementia of DSP-4-induced neurotoxicity by deprenyl is not related to its
Alzheimers type: a role for moclobemide. Psychopharma- inhibition of MAO B. Eur J Pharmacol 184: 119126.
cology 106: S137S139. Foley P, Gerlach M, Youdim MBH et al. (2000). MAO-B
Churchyard A, Mathias CJ, Boonkongchuen P et al. (1997). inhibitors: multiple roles in the therapy of neurodegenera-
Autonomic effects of selegiline: possible cardiovascular toxi- tive disorders? Parkinsonism Rel Disord 6 (1): 2547.
city in Parkinsons disease. J Neurol Neurosurg Psychiatry Fowler CJ, Oreland L, Marcusson J et al. (1980). Titration of
63 (2): 228234. human brain monoamine oxidase -A and -B by clorgyline
Clarke A, Johnson ES, Mallard N et al. (2003a). A new low- and L-deprenil. Naunyn Schmiedebergs Arch Pharmacol
dose formulation of selegiline: clinical efficacy, patient 311: 263272.
preference and selectivity for MAO-B inhibition. J Neural Gerlach M, Riederer R, Przuntek R et al. (1991). MPTP
Transm 110 (11): 12571271. mechanisms of neurotoxicity and their implications for
Clarke A, Brewer F, Johnson ES et al. (2003b). A new formu- Parkinsons disease. Eur J Pharmacol 208: 273286.
lation of selegiline: improved bioavailability and selectivity Gerlach M, Reichmann H, Riederer P (2003). Die Parkinson-
for MAO-B inhibition. J Neural Transm 110 (11): Krankheit Grundlagen, Klinik, Therapie. Unter Mitarbeit
12411255. von W. Gotz und U. Sommer. Dritte, uberarbeitete und
Collins GG, Sandler M, Williams ED et al. (1970). Multiple erweiterte Auflage. XVII, 368 Seiten.
forms of human brain mitochondrial monoamine oxidase. Gibson CJ (1987). Inhibition of MAO-B, but not MAO-A,
Nature 225: 817820. blocks DSP-4 toxicity on central NE neurons. Eur J Pharma-
Da Prada M, Keller H, Keller R et al. (1981). Ro 111163, a col 141: 135138.
specific short acting MAO inhibitor with antidepressant Glover V, Sandler M (1993). Neurotoxins and monoamine
properties. In: Monoamine Oxidase. Basic and Clinical oxidase B inhibitors: possible mechanisms for the neuro-
Frontiers. Excerpta Medica, Amsterdam, pp. 183196. protective effect of ()-deprenyl. In: I Szelenyi, (Ed.),
Da Prada M, Zurcher G, Wuthrich I et al. (1988). On tyra- Inhibitors of Monoamine Oxidase B. Pharmacology and
mine, food beverages and reversible MAO inhibitor moclo- Clinical use in Neurodegenerative Disorders. Birkhauser,
bemide. J Neural Transm Suppl 26: 3156. Basel, pp. 169181.
116 M. B. H. YOUDIM AND P. F. RIEDERER

Goetz CG, Koller WC, Poewe W et al. (2002). Treatment Kragten E, Lalande I, Zimmermann K et al. (1998). Glyceral-
intervention for Parkinsons disease: an evidence based dehyde-3-phosphate dehydrogenase, the putative target of
assessment. Mov Disord 17 (4): 3839. the antiapoptotic compounds CGP 3466 and R-()-depre-
Green AR, Grahame-Smith DG (1975). In: DG Grahame nyl. J Biol Chem 273: 58215828.
Smith (Ed.), Handbook of Psychopharmacology, Vol. 3, Lamensdorf I, Youdim MB, Finberg JP (1996). Effect of
Plenum, New York, pp. 350388. long-term treatment with selective monoamine oxidase
Green AR, Youdim MB (1975). Effects of monoamine oxi- A and B inhibitors on dopamine release from rat striatum
dase inhibition by clorgyline, deprenil or tranylcypromine in vivo. J Neurochem 67 (4): 15321539.
on 5-hydroxytryptamine concentrations in rat brain and Larsen JP, Boas J, Erdal JE (1999). Does selegiline modify
hyperactivity following subsequent tryptophan administra- the progression of early Parkinsons disease? Results from
tion. Br J Pharmacol 55 (3): 415422. a five-year study. The Norwegian-Danish Study Group.
Green AR, Mitchell BD, Tordoff AF et al. (1977). Evidence Eur J Neurol 6: 539547.
for dopamine deamination by both type A and type B Lauterbach EC (2004). The neuropsychiatry of Parkinsons
monoamine oxidase in rat brain in vivo and for the degree disease and related disorders. Psychiatr Clin North Am
of inhibition of enzyme necessary for increased functional 27 (4): 801825.
activity of dopamine and 5-hydroxytryptamine. Br J Phar- Le W, Jankovic J, Xie W et al. (1997). ()-Deprenyl protec-
macol 60 (3): 343349. tion of 1-methyl-4-phenylpyridium ion (MPP)-induced
Haefely W, Burkard WP, Cesura AM et al. (1992). Biochem- apoptosis independent of MAO-B inhibition. Neurosci
istry and pharmacology of moclobemide, a prototype Lett 224: 197200.
RIMA. Psychopharmacology (Berl) 106: S6S14. Leber P (1997). Slowing the progression of Alzheimer dis-
Haefely W, Burkard WP, Cesura A et al. (1993). Pharmacology ease: methodologic issues. Alzheimer Dis Assoc Disord
of moclobemide. Clin Neuropharmacol 16 (Suppl 2): S8S18. 11 (Suppl 5): S10S21; discussion S37S39.
Hallman H, Jonsson G (1984). Pharmacological modifications Leentjens AF (2004). Depression in Parkinsons disease:
of the neurotoxic action of the noradrenaline neurotoxin conceptual issues and clinical challenges. J Geriatr
DSP4 on central noradrenaline neurons. Eur J Pharmacol Psychiatry Neurol 17 (3): 120126.
103 (34): 269278. Lees AJ, Shaw KM, Kohout LL et al. (1977). Deprenyl in
Izumi T, Iwamoto N, Kitaichi Y et al. (2006). Effects of co- Parkinsons disease. Lancet 2 (8042): 791795.
administration of a selective serotonin reuptake inhibitor Magyar K (1993). Pharmacology of monoamine oxidase type
and monoamine oxidase inhibitors on 5-HT-related beha- B inhibitors. In: I Szelenyi, (Ed.), Inhibitors of Monoamine
vior in rats. Eur J Pharmacol 532 (3): 258264. Oxidase B. Pharmacology and Clinical Use in Neurodegen-
Jansen Steur EN, Ballering LA (1999). Combined and selective erative Disorders. Birkhauser, Basel, pp. 125143.
monoamine oxidase inhibition in the treatment of depression Magyar K (1994). Behaviour of ()-deprenyl and its analo-
in Parkinsons disease. Adv Neurol 80: 505509. gues. J Neural Transm Suppl 41: 167175.
Johnston JP (1968). Some observations upon a new inhibitor Magyar KB, Szende J, Lengyel J et al. (1998). The neuropro-
of monoamine oxidase in brain tissue. Biochem Pharma- tective and neuronal rescue effects of ()-deprenyl. J Neural
col 17: 12851297. Transm 52 (Suppl): 109123.
Kalir A, Sabbagh A, Youdim MB (1981). Selective acetylenic Mally J., Kovacs AB., Stone TW. (1995). Delayed develop-
suicide and reversible inhibitors of monoamine oxidase ment of symptomatic improvement by ()-deprenyl in
A and B. Br J Pharmacol 73: 5564. Parkinsons disease. J. Neurol. Sci., 134: 143145.
Knoll J (1987). R-()-deprenyl (Selegiline, Movergan) facil- Mann J, Gershon S (1980). L-deprenyl, a selective monoa-
itates the activity of the nigrostriatal dopaminergic neuron. mine oxidase type-B inhibitor in endogenous depression.
J Neural Transm Suppl 25: 4566. Life Sci 26: 877882.
Knoll J, Magyar K (1972). Some puzzling pharmacological Mann JJ (1989). A controlled study of antidepressant efficacy
effects of monoamine oxidase inhibitors. Adv Biochem and side effects of ()-deprenyl. A selective monoamine
Psychopharmacol 5: 393408. oxidase inhibitor. Arch Gen Psychiatry 46: 4550.
Konradi C, Kornhuber J, Froelich L et al. (1989). Demonstration Marek KL, Friedman J, Hauser R et al. (1997). Phase II
of monoamine oxidase-A and -B in the human brainstem by a evaluation of rasagiline mesylate (TVP-1012), a novel
histochemical technique. Neuroscience 33 (2): 383400. anti-parkinsonian drug, in parkinsonian patients not using
Konradi C, Kornhuber J, Sofic E et al. (1992). Variations of levodopa/carbidopa. Mov Disord 12: 838846.
monoamines and their metabolites in the human brain Maruyama W, Akao Y, Youdim MB et al. (2001). Transfec-
putamen. Brain Res 579 (2): 285290. tion-enforced Bcl-2 overexpression and an anti-Parkinson
Korn A, Da Prada M, Raffesberg W et al. (1988). Tyramine drug, rasagiline, prevent nuclear accumulation of glyceral-
pressor effect in man: studies with moclobemide, a novel, dehyde-3-phosphate dehydrogenase induced by an endo-
reversible monoamine oxidase inhibitor. J Neural Transm genous dopaminergic neurotoxin, N-methyl(R)salsolinol.
Suppl 26: 5771. J Neurochem 78 (4): 727735.
Koutsilieri E, Chen TS, Rausch WD et al. (1996). Selegiline Maruyama W, Akao Y, Carrillo MC et al. (2002). Neuropro-
is neuroprotective in primary brain cultures treated with tection by propargylamines in Parkinsons disease: sup-
1-methyl-4-phenylopyridinium. Eur J Pharmacol 306: pression of apoptosis and induction of prosurvival genes.
181186. Neurotoxicol Teratol 24 (5): 675682.
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 117

Maruyama W, Weinstock M, Youdim MB et al. (2003). OCarroll AM, Fowler CJ, Phillips JP et al. (1983). The dea-
Anti-apoptotic action of anti-Alzheimer drug, TV3326 mination of dopamine by human brain monoamine oxidase.
[(N-propargyl)-(3R)-aminoindan-5-yl]-ethyl methyl carba- Arch Pharmacol 322: 198223.
mate, a novel cholinesterase-monoamine oxidase inhibitor. Olanow C.W. Hauser R.A., Gauger L, Malaparia T, Koller W,
Neurosci Lett 341 (3): 233236. Hubble J, Bushenbark K, Lilienfeld D, Esterlitz J (1995).
Maruyama W, Nitta A, Shamoto-Nagai M et al. (2004). The effect of deprenyl and levodopa on the progression of
N-Propargyl-1(R)-aminoindan, rasagiline, increases glial Parkinsons disease. J. Ann. Neurol, 38: 771777.
cell line-derived neurotrophic factor (GDNF) in neuroblas- Palhagen S, Heinonen EH., Hagglund J, Kaugesaar T., Kontants
toma SH-SY5Y cells through activation of NF-kappaB H, Maki-Ikola O, Palm R., Turunen j. (1998). Selegiline
transcription factor. Neurochem Int 44 (6): 393400. delays the onset of disability in de novo parkinsonian patients.
Marzo A, Dal Bo L, Monti NC et al. (2004). Pharmacokinetics Swedish Parkinson Study Group. J Neurol 51: 520525.
and pharmacodynamics of safinamide, a neuroprotectant Parkinson Study Group (1989). Effect of deprenyl on the
with antiparkinsonian and anticonvulsant activity. Pharma- progression of disability in early Parkinsons disease. The
col Res 50 (1): 7785. Parkinson Study Group. N. Engl. J. Med., 321: 13641371.
Mendlewicz J, Youdim MB (1978). Anti-depressant poten- Parkinson Study Group (1993a). A controlled trial of lazabe-
tiation of 5-hydroxytryptophan by L-deprenyl, an MAO mide (RO196327) in untreated Parkinsons disease. Ann.
type B inhibitor. J Neural Transm 43: 279286. Neurol., 33: 350356.
Mendlewicz J, Youdim MBH (1979). Anti-depressant poten- Parkinson Study Group (1993b). Effects of Tocopherol and
tiation of 5-hydroxytrytophan by l-deprenyl in affective Deprenyl on the Progression of Disability in Early Parkin-
disorder. J Aff Disord 2: 137146. sons Disease. New Engl. J. Med., 328: 176183.
Mendlewicz J, Youdim MBH (1983). L-Deprenil, a selective Parkinson Study Group (2002). A controlled trial of rasagi-
monoamine oxidase type B inhibitor, in the treatment of line in early Parkinson disease: the TEMPO Study. Arch
depression: a double blind evaluation. Br J Psychiatry Neurol 59 (12): 19371943.
142: 508511. Parkinson Study Group (2004). A controlled, randomized,
Mosimann UP, McKeith IG (2003). Dementia with Lewy delayed-start study of rasagiline in early Parkinson dis-
bodiesdiagnosis and treatment. Swiss Med Wkly ease. Arch Neurol 61 (4): 561566.
133 (910): 131142. Parkinson Study Group (2005). A randomized placebo-
Mossner R, Henneberg A, Schmitt A et al. (2001). Allelic controlled trial of rasagiline in levodopa-treated patients
variation of serotonin transporter expression is associated with Parkinson disease and motor fluctuations: the PRE-
with depression in Parkinsons disease. Mol Psychiatry STO study. Arch Neurol 62 (2): 241248.
6 (3): 350352. Paterson IA, Tatton WG (1998). Antiapoptotic actions of mono-
Murphy DL, Wright C, Buchsbaum M et al. (1976). Platelet amine oxidase B inhibitors. Adv Pharmacol 42: 312315.
and plasma amine oxidase activity in 680 normals: sex and Paterson IA, Boulton AA, Juorio AV (1990). Phenylethyla-
age differences and stability over time. Biochem Med 16: mine in the CNS: effects of monoamine oxidase inhibiting
254265. drugs, deuterium substitution and lesions and its role in the
Muller T, Kuhn W, Kruger R et al. (1998). Selegiline as neuromodulation of catecholaminergic neurotransmission.
immunostimulanta novel mechanism of action? J Neural J Neural Transm Suppl 29: 119129.
Transm Suppl 52: 321328. Paterson IA, Zhang D, Warrington RC et al. (1998).
Myllyla VV., Sotaniemie KA., Vuorinen JA., Heinonen EH. R-deprenyl and R-2-heptyl-N-methylpropargylamine pre-
(1992). Neurology, 42: 339343. vent apoptosis in cerebellar granule neurons induced by
Mytilineou C, Radcliffe PM, Olanow CW (1997a). L-()- cytosine arabinoside but not low extracellular potassium.
desmethylselegiline, a metabolite of selegiline [L-()- J Neurochem 70: 515523.
deprenyl], protects mesencephalic dopamine neurons Paykel E, Youdim MBH (Eds.) (1993). Monoamine oxidase
from excitotoxicity in vitro. J Neurochem 68 434436 inhibitors: new developments. Clin Neuropharmacol 16
Clarke A, Brewer F, Johnson ES et al. (1997a). A new (Suppl 2): 96212.
formulation of selegiline: improved bioavailability and Priest RG (1992). Moclobemide: a range of opportunities.
selectivity for MAO-B inhibition. J Neural Transm Psychopharmacology (Berl) 106: S140S141.
110 (11): 12411255. Przuntek H., Kuhn W. (1987). The effect of R-()-deprenyl
Mytilineou C, Radcliffe P, Leonardi EK et al. (1997b). in de novo Parkinson patients on combination therapy with
L-deprenyl protects mesencephalic dopamine neurons levodopa and decarboxylase inhibitor. J. Neural Transm
from glutamate receptor-mediated toxicity in vitro. J Neu- 25: 97104.
rochem 68: 3339. Przuntek H., Conrad B., Dichgans J., Kraus PH., Krauseneck P.,
Naoi M, Maruyama W, Youdim MB et al. (2003). Anti-apoptotic Pergande G., Rinne U., Schimrigk K., Schnitker J., Vogel
function of propargylamine inhibitors of type-B monoamine HP. (1999). SELEDO: a 5-year long-term trial on the effect
oxidase. Inflammopharmacology 11 (2): 175181. of selegiline in early Parkinsonian patients treated with
Nieuwstraten C, Labiris NR, Holbrook A (2006). Systematic levodopa. J. Neurol. 6: 141150.
overview of drug interactions with antidepressant medica- Rascol O, Brooks DJ, Melamed E et al. (2005). LARGO Study
tions. Can J Psychiatry 51 (5): 300316. Group. Rasagiline as an adjunct to levodopa in patients with
118 M. B. H. YOUDIM AND P. F. RIEDERER

Parkinsons disease and motor fluctuations (LARGO, Last- K., Rudolpha A., Parkisnon Study Group. (2002). Impact of
ing effect in Adjunct therapy with Rasagiline Given Once sustained deprenyl (selegiline) in levodopa-treated Parkin-
daily, study): a randomised, double-blind, parallel-group sons disease: a randomized placebo-controlled extension
trial. Lancet 365 (9463): 947954. of the deprenyl and tocopherol antioxidative therapy of par-
Ravikumar R, Lakshmana MK, Rao BS et al. (1998). kinsonism trial. J. Ann. Neurol., 51: 604612.
()-Deprenyl attenuates spinal motor neuron degeneration Sieradzan K, Chanon S, Stern G et al. (1995). The therapeu-
and associated locomotor deficits in rats subjected to spinal tic potential of moclobemide, a reversible selective mono-
cord ischemia. Exp Neurol 149: 123129. amine oxidase A inhibitor in Parkinsons disease. Clin
Revuelta M, Venero JL, Machado A et al. (1997). Deprenyl Psychopharmacol 15: 51S60S.
induces GFAP immunoreactivity in the intact and injured Singer TP, Ramsay RR (1991). The interaction of monoa-
dopaminergic nigrostriatal system but fails to counteract mine oxidases with tertiary amines. Biochem Soc Trans
axotomy-induced degenerative changes. Glia 21: 204216. 19: 211215.
Riederer P, Lachenmayer L (2003). Selegilines neuroprotective Sivertsen B., Dupont E., Mikkelsen B., Mogensen P., Ras-
capacity revisited. J Neural Transm 110 (11): 12731278. mussen C., Boesen F., Heinonen E. (1989). Selegiline
Riederer P, Youdim MBH (1986). Human brain monoamine and levodopa in early of moderately advanced Parkinsons
oxidase activity and monoaminemetabolism in Parkinso- disease: a double-blind controlled short- and longterm
nian patients treated with L-deprenyl. J Neurochem 45: study. E. Acta Neurol. Scand. 126: 147152.
13491356. Speiser Z, Katzir O, Rehavi M et al. (1998a). Sparing by rasagi-
Riederer P, Konradi C, Schay V et al. (1986). Location of line (TVP-1012) of cholinergic functions and behavior in the
MAO-A and MAO-B in human brain, a step in under- postnatal anoxia rat. Pharmacol Biochem Behav 60: 387393.
standing the therapeutic action of L-deprenyl. Adv Neurol Speiser Z, Levy R, Cohen S (1998b). Effects of N-propargyl-
45: 111119. 1-(R)aminoindan (rasagiline) in models of motor and cog-
Riederer P, Lachenmayer L, Laux G (2004). Clinical applica- nition disorders. J Neural Transm Suppl 52: 287300.
tions of MAO-inhibitors. Curr Med Chem 11: 20332043. Sprague JE, Nichols DE (1995). Inhibition of MAO-B
Rihmer Z, Seregi K, Rihmer A (2004). Parkinsons disease protects against MDMA-induced neurotoxicity in the stria-
and depression. Neuropsychopharmacol Hung 6 (2): 8285. tum. Psychopharmacology (Berl) 118: 357364.
Roy E, Bdard PJ (1993). Deprenyl increases survival of rat Sternic N, Kacar A, Filipovic S et al. (1998). The therapeutic
foetal nigral neurones in culture. Neuroreport 4: 11831186. effect of moclobemide, a reversible selective monoamine
Sagi Y, Weinstock M, Youdim MB (2003). Attenuation of oxidase A inhibitor, in Parkinsons disease. Clin Neuro-
MPTP-induced dopaminergic neurotoxicity by TV3326, pharmacol 21: 9396.
a cholinesterase-monoamine oxidase inhibitor. J Neuro- Sterling J, Tamas T, Toth G et al. (2002). Novel dual inhibi-
chem 86 (2): 290297. tors of AChE and MAO derived from hydroxyl aminoindan
Sagi Y, Mandel S, Amit L et al. (2006). Activation of tyro- and phenetylamine as potential treatment for Alzheimers
sine kinase receptor signaling pathway by rasagiline facil- disease. J Med Chem 45 (24): 52605279.
itates neurorescue and restoration of nigrostriatal Stocchi F, Arnold G, Onofrj M et al. (2004). Safinamide
dopamine neurons in post-MPTP-induced parkinsonism. Parkinsons Study Group. Improvement of motor function in
Neurobiol Dis 25 (1): 3544. early Parkinson disease by safinamide. Neurology 63 (4):
Salo PT, Tatton WG (1992). Deprenyl reduces the death of moto- 746748.
neurons caused by axotomy. J Neurosci Res 31: 394400. Szeleny I (Ed.) (1993). Inhibitors of Monoamine Oxidase
Sautter J, Gerlach M, Schwarz J et al. (1994). TVP-1012, an B. Birkhauser, Boston.
irreversible MAO-B-inhibitor protects against MPTP- Tatton WG, Chalmers-Redman RM (1996). Modulation of
neurotoxicity in the monkeycomparison with deprenyl. gene expression rather than monoamine oxidase inhibition:
11th International Symposium on Parkinsons Disease, ()-deprenyl-related compounds in controlling neurodegen-
Rome, March 2630. eration. Neurology 47 (Suppl 3): S171S183.
Schmidt DE, Ebert MH, Lynn JC et al. (1997). Attenuation Tatton WG, Ju WY, Holland DP et al. (1994). ()-Deprenyl
of 1-methyl-4-phenylpyridinium (MPP) neurotoxicity reduces PC12 apoptosis by inducing new protein synth-
by deprenyl in organotypic canine substantia nigra cul- esis. J Neurochem 63: 15721575.
tures. J Neural Transm 104: 875885. Tatton WG, Wadia JS, Ju WY et al. (1996). ()-Deprenyl
Semokova I, Wolz P, Schilling M et al. (1996). Selegiline reduces neuronal apoptosis and facilitates neuronal out-
enhances NGF synthesis and protects central nervous sys- growth by altering protein synthesis without inhibiting
tem (CNS) neurons from excitotoxic and ischemic damage. monoamine oxidase. J Neural Transm 48: 4559.
Eur J Pharmacol 315: 1930. Tetrud JW, Langston JW. (1990). The effect of deprenyl
Seniuk NA, Henderson JT, Tatton WG et al. (1994). Increased (selegiline) on the natural history of Parkinsons disease.
CNTF gene expression in process-bearing astrocytes fol- J. W. Science, 245: 519522.
lowing injury is augmented by R()-deprenyl. J Neurosci Thiffault C, Lamarre-Theroux L, Quirion R et al. (1997).
Res 37: 278286. L-deprenyl and MDL72974 do not improve the recovery
Shoulson I., Oakes D., Fahn S., Lang A., Langston J.W., of dopaminergic cells following systemic administration
LeWitt P., Olanow C.W., Penney J.B., Tanner C., Kieburtz of MPTP in mouse. Mol Brain Res 44: 238244.
MONOAMINE OXIDASE A AND B INHIBITORS IN PARKINSONS DISEASE 119

Vaglini F, Pardini C, Cavalletti M et al. (1996). L-deprenyl amyloid precursor protein processing. FASEB J 17 (15):
fails to protect mesencephalic dopamine neurons and 23252327.
PC12 cells from the neurotoxic effects of 1-methyl-4- Youdim MBH (Ed.) (1995). Reversible and Selective
phenylpyridinium ion. Brain Res 741: 6874. Inhibitors of Monoamine Oxidase: New Findings. Acta
Wadia JS, Chalmers-Redman RM, Ju WJ et al. (1998). Psyuchiat, Scand 91:143.
Mitochondrial membrane potential and nuclear changes Youdim MB, Buccafusco JJ (2005a). Multi-functional drugs
in apoptosis caused by serum and nerve growth factor for various CNS targets in the treatment of neurodegenera-
withdrawal: time course and modification by ()-depre- tive disorders. Trends Pharmacol Sci 26 (1): 2735.
nyl. J Neurosci 18: 932947. Youdim MB, Buccafusco JJ (2005b). CNS Targets for multi-
Waters CH, Sethi KD, Hauser RA et al. (2004). Zydis Sele- functional drugs in the treatment of Alzheimers and Par-
giline Study Group. Zydis selegiline reduces off time in kinsons diseases. J Neural Transm 111: 519537.
Parkinsons disease patients with motor fluctuations: a 3- Youdim MB, Riederer P (1993a). Dopamine metabolism and
month, randomized, placebo-controlled study. Mov Disord neurotransmission in primate brain in relationship to
19: 426432. monoamine oxidase A and B inhibition. J Neural Transm
Weinreb O, Bar-Am O, Amit T et al. (2004). Neuroprotection 91: 181195.
via pro-survival protein kinase C isoforms associated with Youdim MB, Riederer P (1993b). The relevance of glial mono-
Bcl-2 family members. FASEB J 18 (12): 14711473. amine oxidase-B and polyamines to the action of selegiline
Weinstock M, Bejar C, Wang RH et al. (2000). TV 3326, in Parkinsons disease. Mov Disord 8 (Suppl 1): S8S13.
a novel neuroprotective drug with cholinesterase and mono- Youdim MB, Weinstock M (2001). Molecular basis of neuro-
amine oxidase inhibitory activities for the treatment of protective activities of rasagaline and the anti-Alzheimer
Alzheimers disease. J Neural Transm Suppl 60: 157169. drug TV3326 [(N-propargyl-(3R)aminoindan-5-YL)-ethyl
Weinstock M, Kirschbaum-Slager N, Lazarovici P et al. (2001). methyl carbamate.] Cell Mol Neurobiol 21 (6): 555573.
Neuroprotective effects of novel cholinesterase inhibitors Youdim MBH, Sourkes TL (1965). The effect of heat, pH
derived from rasagiline as potential anti-Alzheimer drugs. and riboflavin deficiency on monoamine oxidase activity.
Ann NY Acad Sci 939: 148161. Can J Biochem 43: 13051318.
Weinstock M, Poltyrev T, Bejar C et al. (2002a). Effect of TV Youdim MB, Collins GG, Sandler M et al. (1972). Human
3326, a novel monoamine-oxidase cholinesterase inhibitor, brain monoamine oxidase, multiple forms and selective
in rat models of anxiety and depression. Psychopharmacol- inhibitors. Nature 236: 225228.
ogy (Berl) 160 (3): 318324. Youdim MBH, Green AR, Grahame-Smith DG (1976). The
Weinstock M, Gorodetsky E, Wang RH et al. (2002b). role of 5-hydroxytryptamine, dopamine and MAO in the pro-
Limited potentiation of blood pressure response to oral duction of hyperactivity. In: W Birkmayer, O Hornykiewicz
tyramine by brain-selective monoamine oxidase A-B inhi- (Eds.) Advances in Parkinsonism. Editiones Roche, Base,
bitor, TV-3326 in conscious rabbits. Neuropharmacology pp. l155163.
43 (6): 9991005. Youdim MBH, Finberg JPM, Tipton KF (1988a). Monoamine
Weinstock M, Gorodetsky E, Poltyrev T et al. (2003). A novel oxidase. In: U Trendelengurg , U Weiner (Eds.), Catechola-
cholinesterase and brain-selective monoamine oxidase inhi- mine II. Springer-Verlag, Berlin, pp. 119192.
bitor for the treatment of dementia comorbid with depres- Youdim MBH, Da Prada M, Amrein R (Eds.) (1988b). The
sion and Parkinsons disease. Prog Neuropsychopharmacol cheese effect and new reversible MAO-A inhibitors.
Biol Psychiatry 27 (4): 555561. J Neural Transm (Suppl 26).
Wild R, Pettit T, Burns A (2003). Cholinesterase inhibitors Youdim MB, Gross A, Finberg JP (2001a). Rasagiline [N-pro-
for dementia with Lewy bodies. Cochrane Database Syst pargyl-1R()-aminoindan], a selective and potent inhibitor
Rev 3: CD003672. of mitochondrial monoamine oxidase B. Br J Pharmacol
Wilfried K, Muller T, Kruger R et al. (1997). Selegiline 132 (2): 500506.
stimulates biosynthesis of cytokines interleukin-1 beta Youdim MB, Wadia A, Tatton W et al. (2001b). The
and interleukin-6. Neuroreport 7: 28472848. anti-Parkinson drug rasagiline and its cholinesterase
Williams K, Romano C, Dichter MA et al. (1991). Modula- inhibitor derivatives exert neuroprotection unrelated to
tion of the NMDA receptor by polyamines. Life Sci 48: MAO inhibition cell culture and in vivo. Ann NY Acad
469498. Sci 939: 450458.
Wu RM, Murphy DL, Chiueh CC (1995). Neuronal protec- Youdim MB, Amit T, Falach-Yogev M (2003). The essenti-
tive and rescue effects of deprenyl against MPPdopami- ality of Bcl-2, PKC and proteasome-ubiquitin complex
nergic toxicity. J Neural Transm 100: 5361. activations in the neuroprotective-antiapoptotic action of
Wu RM, Murphy DL and Chiueh CC (1996). Suppression of the anti-Parkinson drug, rasagiline. Biochem Pharmacol
hydroxyl radical formation and protection of nigral neu- 66 (8): 16351641.
rons by l-deprenyl (selegiline). Ann NY Acad Sci 786: Youdim MB, Maruyama W, Naoi M (2005). Neurophar-
379390. macological, neuroprotective and amyloid precursor
Yogev-Falach M, Amit T, Bar-Am O et al. (2003). The processing properties of selective MAO-B inhibitor anti-
importance of propargylamine moiety in the anti-Parkinson parkinsonian drug, rasagiline. Drugs Today (Barc)
drug rasagiline and its derivatives in MAPK-dependent 41 (6): 369391.
120 M. B. H. YOUDIM AND P. F. RIEDERER

Youdim MBH, Edmondson D, Tipton KF (2006). Monoa- mine oxidases A and B in the brain of the common
mine oxidases. Nat Rev Neurosci 7: 295309. marmoset. J Neural Transm Suppl 52: 271278.
Yu PH, Davis BA, Fang J et al. (1994). Neuroprotective Green AR, Youdim MBH (1976). Use of animal models to
effects of some monoamine oxidase-B inhibitors against study the action of monoamine oxidase inhibitors. In:
DSP-4-induced noradrenaline depletion in the mouse Monoamine Oxidase and Its Inhibition, Ciba Foundation
hippocampus. J Neurochem 63: 18201828. Symposium No. 39 Elsevier, Amsterdam, pp. 231246.
Zappia V, Pegg AE (Eds.) (1998b). Progress in Polyamine Lai CT, Yu PH (1997). R()-deprenyl potentiates dopamine-
Research. Plenum Press, New York. induced cytotoxicity toward catecholaminergic neuro-
Zhang X, Zuo DM, Davis BA et al. (1996). Immuno- blastoma SH-SY5Y cells. Toxicol Appl Pharmacol 142:
histochemical evidence of neuroprotection by R(-)-deprenyl 186191.
and N-(2-hexyl)-N-methylpropargylamine on DSP-4-induced Larsen JP, Boas J (1997). The effects of early selegiline ther-
degeneration of rat brain noradrenergic axons and terminals. apy on long-term levodopa treatment and parkinsonian
J Neurosci Res 43: 482489. disability: an interim analysis of a NorwegianDanish
5-year study. Norwegian-Danish Study Group. J. Move-
ment Disord 12: 175182.
Further Reading
Schmauss M (2002). Kontrolluntersuchungen. In: Riederer P,
Bieck PR, Antonin KH, Schmidt E (1993). Clinical pharma- Laux, G, Poldinger (Hrsg.). Neuro-Psychopharmaka,
cology of reversible monoamine oxidase inhibitors. Clin Band 3: Antidepressiva, Phasenpophylaktika und Stim-
Neuropharmacol 16 (Suppl 2): S34S41. mungsstabilisierer, 2. neubearbeitete Auflage. Springer,
Fuller RW, Hemrick-Luecke SK (1985). Inhibition of types Wien, pp. 538539.
A and B monoamine oxidase A and B by 1-methyl- Tatton WG, Greenwood CE (1991). Rescue of dying neurons:
4-phenyl-1,2,3,6-dihydropyridine. J Pharmacol Exp Ther a new action for deprenyl in MPTP parkinsonism. J Neurosci
232: 696701. Res 30: 666672.
Gerlach M, Riederer P (1996). Animal models of Parkinsons Zreik MJ, Fozard MJR, Dudley MW et al. (1989). MDL
disease: an empirical comparison with the phenomenology 72.974A: a potent and selective enzyme activated irreversi-
of the disease in man. J Neural Transm 103: 9871041. ble inhibitor of monoamine oxidase type B with potential
Gotz ME, Breithaupt W, Sautter J et al. (1998). Chronic for use in Parkinsons disease (Parkinsons disease and
TVP-1012 (rasagiline) doseactivity response of monoa- dementia section). J Neural Transm 1: 243254.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 35

Anticholinergic medications

YAROSLAU COMPTA AND EDUARDO TOLOSA*

Neurology Service, Hospital Clinic, University of Barcelona, Barcelona, Spain

35.1. Introduction an antiparkinsonian effect, confirming the aforemen-


tioned Feldburgs (1945) hypothesis.
Ordenstein, following the observations of his professor,
Jean-Martin Charcot, first described the beneficial 35.2. Mechanism of action and clinical trials
effect of the belladonna alkaloids (mainly containing
atropine as active component) on tremor and other Anticholinergics appear to improve symptoms of
Parkinson disease (PD) symptoms (Ordenstein, 1867). PD through a central anticholinergic effect exerted in
Subsequent investigators, such as Gowers (1888) at the striatum. Duvoisin showed that cholinesterase inhi-
the end of the 19th century, agreed that other substances bitors, which can penetrate the brain, increase the
like Indian hemp (containing Cannabis sativa), scopol- severity of PD symptoms (Barbeau, 1962; Coyle and
amine (hyoscine) and hyoscyamine (duboisine) were Snyder, 1969), an effect that can be reversed by anti-
effective in mitigating tremor and muscular rigidity. cholinergics, such as benzotropine. This observation
Ordenstein suggested that parkinsonian symptoms provides a rationale for the use of anticholinergics
resulted from peripheral parasympathetic overactivity, in PD and supports the notion that a state of striatal
after the vagolyitic effect exerted by belladonna alka- cholinergic preponderance exists in PD secondary
loids. It was not until 1945 that Feldburg discovered to the striatal dopamine deficiency (Barbeau, 1962).
that acetylcholine was also a central neurotransmitter It is thought that the antimuscarinic properties of the
which was abundant in the striatum at the synaptic vesi- anticholinergics mediate their antiparkinsonian pro-
cles of nerve terminals, and suggested a central effect perties. In two studies with direct mesencephalic and
for anticholinergic drugs. A few years later, Barbeau pallidal administration of drugs, atropine was found
(1962) proposed that an altered dopaminergiccholi- to have an antiparkinsonian action (Nashold, 1959;
nergic balance exists in the striatum in PD with the Velasco et al., 1982).
primary deficit in dopamine leading to a secondary Centrally active anticholinergics (muscarinic recep-
relative overactivity of acetylcholine. Normalization tor antagonists) exert a modest improvement on PD
of this striatal imbalance would explain the beneficial symptoms, but the percentage of patients reported to
effect of anticholinergics in PD. Another proposed improve with these drugs has varied greatly, from 40
mechanism of action for anticholinergics in PD is the to 77% in open trials (Corbin, 1949; Strang, 1965)
inhibition of dopamine reuptake in the striatum (Coyle and from 20 to 40% in double-blind studies (Parkes
and Snyder, 1969). et al., 1974).
For over half a century the belladonna alkaloids It is frequently stated that anticholinergics are more
formed the mainstay of the medical management of effective in alleviating resting tremor and rigidity than
the Parkinson syndrome. These natural products akinesia and that the major benefit derived from their
were substituted in the 1960s by a series of synthetic use is precisely from their tremorolytic effects (Doshay
anticholinergics and antihistaminics (Strang, 1965). and Constable, 1957; Burns et al., 1964; Strang, 1965;
Synthetic anticholinergic drugs with selective periph- Ebling, 1971; Obeso and Martnez-Lage, 1987). Such
eral effects (Duvoisin, 1966, 1967) proved not to have affirmation has its origin in the observation that the

*Correspondence to: Professor Eduardo Tolosa, Neurology Service, Hospital Clinic, Esc 8, 4 planta, Villarroel 170, 08036
Barcelona, Spain. E-mail: ETOLOSA@clinic.ub.es, Tel: 0034-93-227-5785, Cell: 0034-227-5783.
122 Y. COMPTA AND E. TOLOSA
injection of the cholinomimetic drug tremorine elicited 35.3. Adverse effects
tremor and rigidity in animals (Everett et al., 1956),
symptoms that improved after the administration of Peripheral adverse effects of these agents include
atropine or scopolamine. Reports suggesting that antic- tachycardia, constipation (rarely leading to paralytic
holinergics do not have a specific antitremor effect, ileus), urinary retention, blurred vision and dry mouth
however, abound in the literature. Thus, Marshall (Duvoisin, 1965; Ebling, 1971). Gingivitis and caries,
(1968) was of the opinion that anticholinergics had rarely leading to loss of teeth, may occur (Lang and
little or no effect upon the tremor of PD, and Yahr Blair, 1989) and reduced sweating may interfere with
and Duvoisin (1968), in a review on the subject, body temperature regulation. These effects are all
concluded that the modest improvement achieved reversible when diminishing or with discontinuation
with anticholinergics is derived from a greater effect of the drug, and can even show some tolerance after
in relieving muscular rigidity and akinesia rather than prolonged exposure. Rarely, some of these side-effects
tremor. can be beneficial at times, as is the case for dry mouth,
Still, although a specific tremolytic effect of the which can be advantageous in patients with prominent
anticholinergics in PD has not been well documented, drooling. Caution must be exercised in elder male
most investigators report improvement in tremor with patients with comorbid prostate hypertrophy, due to a
anticholinergic monotherapy. A double-blind study high risk for urinary retention. Blurred vision is a com-
using objective evaluation of tremor with accelero- mon side-effect, attributed to reduced accommodation
metry (Koller, 1986) clearly showed a reduction in tre- due to parasympathetic blockade. Extremely rare is
mor amplitude of about 60% from baseline in a group the occurrence of acute narrow-angle glaucoma, which
of 10 de novo patients with early parkinsonism, after can be precipitated in predisposed patients.
treatment with trihexyphenidyl. However, other stu- The usefulness of anticholinergics is also limited by
dies using quantitative measures of tremor differed central side-effects. These include sedation, confusion
in their results, showing both improvement (Agate and psychiatric disturbances, such as hallucina-
et al., 1956) and lack of tremolytic effect (Norris and tions and psychosis (Porteous and Ross, 1956; Koller,
Vas, 1967). 1984). Impaired mental function (mainly immediate
The differences described in the effect of anticholi- memory and memory acquisition) is a well-documented
nergics on tremor and other symptoms of the PD have central side-effect that resolves after drug withdrawal
been attributed to differences in patient selection, drug (van Herwaarden et al., 1993). Anticholinergics can
dosages, specific drugs studied and on methods of lead to an exacerbation of frontal lobe dysfunction in
assessment. Whether patients with early mild disease PD patients (Syndulko et al., 1981; Sadeh et al., 1982;
respond better or worse than those with more advanced Dubois et al., 1987). An impairment of higher cortical
parkinsonism has not been clarified, and it appears that functions has been found in non-demented PD subjects
the use of higher doses of anticholinergics does not with an acute subclinical dose of scopolamine (Bedard
yield better results than lower doses. Marsden (1969), et al., 1998) and impaired neuropsychiatric function
in a placebo-controlled study, compared different has been demonstrated even in patients without cog-
doses of two anticholinergics and found no evidence nitive impairment (Syndulko et al., 1981; Sadeh et al.,
of increased benefit from higher doses with either 1982). These central effects are more likely to occur
of the two, a failure that did not seem to be related with advanced age and in patients with dementia (De
to the appearance of side-effects. Burns et al. (1964) Smet et al., 1982). The marked involvement of the
also found no evidence of increasing benefit with cholinergic system (i.e. nucleus basalis of Meynert) in
increasing dosage of trihexyphenidyl in a carefully PD (Forster and Lewy, 1912; Braak and Braak, 2000)
controlled clinical trial. Therapeutic differences among and in dementia with Lewy bodies (Tiraboschi et al.,
the various synthetic anticholinergics are probably 2002) pathology is probably the basis of the cognitive
minor, but some patients may tolerate one better than changes induced by anticholinergics.
the other.
Studies of trihexyphenidyl (Martin et al., 1974), ben- 35.4. Anticholinergic use in clinical practice
zotropine (Tourtellotte et al., 1982) and bornaprine
(Cantello et al., 1986) in levodopa-treated patients, and The anticholinergics currently in use are listed in
two reviews indicate that adjunctive anticholinergics Table 35.1. One of the first introduced synthetic drugs
have only a minor effect on PD symptoms in patients is a piperidine compound, trihexyphenidyl. It was initi-
on levodopa therapy (Goetz et al., 2002; Katzenschlager ally developed as a gastrointestinal antispasmodic
et al., 2003). agent, shown in trials in the late 1940s to be as effective
ANTICHOLINERGIC MEDICATIONS 123
Table 35.1 et al., 1974; Tourtellotte et al., 1982; Yahr et al.,
1982; Koller, 1986).
Anticholinergic drugs commonly used to treat tremor in
Development of tolerance to the effects of benefi-
Parkinsons disease
cial effects of anticholinergics is said to occur fre-
Generic name Daily dose (mg) quently. Such loss of therapeutic benefit is indeed a
common clinical observation after months of treat-
Trihexyphenidyl 120 ment, but should be attributed, at least in part, to
Benztropine 0.56 disease progression. Withdrawal of anticholinergics,
Ethopropazine 100800 even in patients in whom it is thought that the drugs
Procyclidine 7.520 are no longer effective, invariably results in worsening
Diphenhydramine 25200 of the parkinsonian symptoms, at times to a level
Bornaprine 88.25 worse than the patients baseline state (Hughes et al.,
1971; Horrocks et al., 1973).
In PD anticholinergic drugs have been reported to
as belladonna alkaloids in the management of PD, with alleviate dystonic spasms resulting from chronic levo-
one-half of patients achieving an average of 20% dopa administration, as is the case in early-morning
improvement (Rix and Fisher, 1972). The most repre- dystonia (Poewe et al., 1987). In another study by
sentative drugs of the antihistamines are diphenhydra- Poewe et al. (1988), challenge with procyclidine in 9
mine (Montuschi, 1949) and orphenadrine (Strang, PD patients with foot dystonia resulted in abolition
1965); orphenadrine is thought to be more potent of dystonia in 6, amelioration in 1 and no effect in
because of its more potent anticholinergic activity. the remaining 2 patients.
Another widely used anticholinergic drug is benztro-
pine, a combination of the atropine molecule and the 35.5. Summary
benzhydryl group of the diphenhydramine molecule
(Doshay et al., 1952). Developed in the 1950s, this drug The anticholinergics have been shown to improve the
is more potent than trihexyphenidyl and less sedative main symptoms of PD modestly. They are used as
than antihistaminics. Other less commonly used antic- initial treatment in the early stages of the disease. Less
holinergics are phenothiazine derivatives (i.e. ethopro- commonly they are administered as adjuncts to levo-
pazine), which, despite being neuroleptics, show dopa or other therapeutic agents in more advanced
prominent anticholinergic activity and, hence, some stages. Levodopa-induced dystonia is another situation
degree of antiparkinsonian effects (Young, 1972). where anticholinergics could be helpful.
Nowadays, due to the propensity to induce adverse In recent years the use of anticholinergics for the
effects, the anticholinergics have been progressively symptomatic treatment of PD has decreased signifi-
substituted as first-line treatment of the early stages cantly, mostly due to the successful introduction into
of PD (Yahr, 1990) by other drugs such as the mono- therapy of a variety of new dopamine agonists and
amine oxidase-B inhibitors or the modern dopamine enzyme inhibitors that help to control symptoms and
agonists, which have better tolerability and have have been shown to be efficacious in delaying the intro-
been shown to be associated with fewer motor com- duction of levodopa or minimizing motor complica-
plications when compared with levodopa (Rascol tions of chronic levodopa administration. Such studies
et al., 2002). However, no trials have directly com- have not been performed with the anticholinergics.
pared the effects of dopamine agonists with those of Because of their side-effect profile, with both periph-
the anticholinergics in PD. eral and central adverse effects, the anticholinergics
Despite a lack of definite data to confirm a special should be always started at low doses and upward titra-
role in the management of tremor, anticholinergics tion of doses done gradually. Due to their negative
are also recommended in patients in whom other thera- effects on memory and cognition, their use should be
pies such as the agonists amantadine or levodopa have avoided in elderly patients. Therapeutic differences
failed to control tremor sufficiently. among the various synthetic anticholinergics are prob-
In patients on long-term levodopa therapy and ably minor, but some patients may tolerate one better
more advanced disease, e.g. patients with associated than another.
motor complications such as fluctuations or dyskine-
sias, the beneficial effect from adding anticholinergics References
has been questioned (Martin et al., 1974), but some
authors believe that in some instances the addition of Agate FJ, Dosahy LJ, Curtis FK (1956). Quantitative measure-
anticholinergics may convey some benefit (Parkes ment of therapy in paralysis agitans. JAMA 160: 353354.
124 Y. COMPTA AND E. TOLOSA

Barbeau A (1962). The pathogenesis of Parkinsons disease: Hughes RC, Polgar JG, Weightman D et al. (1971). Levodopa
a new hypothesis. Can Med Assoc J 87: 802807. in Parkinsonism: the effects of withdrawal of anticholinergic
Bedard MA, Lemay S, Gagnon JF et al. (1998). Induction of drugs. Br Med J 2 (760): 487491.
a transient dysexecutive syndrome in Parkinsons disease Katzenschlager R, Sampaio C, Costa J et al. (2003). Antic-
using a subclinical dose of scopolamine. Behav Neurol holinergics for symptomatic management of Parkinsons
11: 187195. disease. Cochrane Database Syst Rev. (2): CD003735.
Braak H, Braak E (2000). Pathoanatomy of Parkinsons Koller WC (1984). Disturbance of recent memory functions
disease. J Neurol 247 (Suppl 2): II310. in parkinsonian patients on anticholinergic therapy. Cortex
Burns D, DeJong D, Solis-Quiroga OH (1964). Effects of 20: 307311.
trihexyphenidyl hydrochloride (Artane) on Parkinsons Koller WC (1986). Pharmacologic treatment of parkinsonian
disease. Neurology 14: 1323. tremor. Arch Neurol 43: 126127.
Cantello R, Riccio A, Gilli M (1986). Bornaprine vs placebo Lang AE, Blair RDG (1989). Anticholinergic drugs and
in Parkinson disease: double-blind controlled cross-over amantadine in the treatment of Parkinsons Disease. In:
trial in 30 patients. Ital J Neurol Sci 7 (1): 139143. DB Calne, (Ed.), Handbook of Experimental Pharmacology,
Corbin KB (1949). Trihexyphenidyl: evaluation of a new Vol. 88: Drugs for the Treatment of Parkinsons Disease.
agent in treatment of parkinsonism. JAMA 141: 377382. Springer Verlag, Berlin, Heidelberg.
Coyle JT, Snyder SH (1969). Antiparkinsonian drugs: inhibi- Marsden CD (1969). Extending the use of anticholinergic
tion of dopamine uptake in the corpus striatum as a possi- drugs in Parkinsons disease. In: FJ Gillingham, IM
ble mechanism of action. Science 166: 899901. Donaldson (Eds.), Third Symposium on Parkinsons
De Smet Y, Ruberg M, Serdaru M et al. (1982). Confusion, Disease. E & S Livingstone, Edinburgh, pp. 185192.
dementia, and anticholinergics in Parkinsons disease. J Marshall J (1968). Tremor. In: PJ Vinken, GW Bruyn (Eds.),
Neurol Neurosurg Psychiatry 45: 11611164. Handbook of Clinical Neurology: Diseases of Basal Ganglia.
Doshay LJ, Constable K (1957). Treatment of paralysis Elsevier North-Holland, Amsterdam, pp. 809825.
agitans with orphenadrine (Disipal) hydrochloride. JAMA Martin WE, Lowenson RB, Resch JA et al. (1974). A con-
163: 13521357. trolled study comparing trihexyphenidyl hydrochloride
Doshay LJ, Constable K, Fromer S (1952). Preliminary study plus levodopa with placebo plus levodopa in patients with
of a new antiparkinsonian. Neurology 2: 233243. Parkinsons disease. Neurology 24: 912919.
Dubois B, Danze F, Pillon B et al. (1987). Cholinergic- Montuschi E (1949). Benadryl in parkinsonism. The Lancet
dependent cognitive deficits in Parkinsons disease. Ann 1: 546.
Neurol 22: 2630. Nashold BS (1959). Cholinergic stimulation of globus
Duvoisin RC (1965). A review of drug therapy in parkinson- pallidus in man. Proc Soc Exp Biol Med 101: 6869.
ism. Bull NY Acad Med 41: 898910. Norris JW, Vas CJ (1967). Mehixene hydrochloride and
Duvoisin RC (1966). The mutual antagonism of cholinergic parkinsonian tremor. Acta Neurol Scand 43: 535538.
and anticholinergic agents in parkinsonism. Trans Am Obeso JA, Martnez-Lage M (1987). Anticholinergics and
Neurol Assoc 91: 7379. amantadine. In: W Koller (Ed.), Handbook of Parkinsons
Duvoisin RC (1967). Cholinergic-anticholinergic antagonism Disease. Marcel Dekker, New York, pp. 309316.
in parkinsonism. Arch Neurol 17: 124136. Ordenstein L (1867). Sur la paralysie et la sclerose en plaque
Ebling P (1971). The medical management of Parkinsons generalise. Martinet, Paris.
disease before the introduction of L-dopa. Aust N Z J Parkes JD, Baxter RC, Marsden CD et al. (1974). Com-
Med 1 (Suppl): 3538. parative trial of benzhexol, amantadine and levodopa in
Everett GM, Blockus LE, Shepperd IM (1956). Tremor induced the treatment of Parkinson disease. J Neurol Neurosurg
by tremorine and its antagonism by anti-parkinsonian drugs. Psychiatry 37: 422426.
Science 124: 79. Poewe W, Lees AJ, Steiger D et al. (1987). Foot dystonia in
Feldburg W (1945). Present views on the mode of action Parkinsons disease: clinical phenomenology and neuro-
of acetylcholine in the central nervous system. Physiol pharmacology. Adv Neurol 45: 357360.
Rev 25: 596642. Poewe W, Lees AJ, Stern GM (1988). Dystonia in Parkinsons
Forster E, Lewy FH (1912). Paralysis agitans. In: M Lewan- disease: clinical and pharmacological features. Ann Neurol
dowsky (Ed.), Pathologische Anantomie. Handbuch der 235: 7378.
Neurologie. Springer Verlag, Berlin, pp. 920933. Porteous HB, Ross DDN (1956). Mental symptoms in
Goetz CG, Koller WC, Poewe W et al. (2002). Management parkinsonism following benzhexol hydrochloride therapy.
of Parkinsons disease: an evidence-based review. Mov Br Med J 2: 138140.
Disord 17 (Suppl 4): S1S166. Rascol O, Goetz C, Koller W et al. (2002). Treatment inter-
Gowers WR (1888). A Manual of Disease of the Nervous ventions for Parkinsons disease: an evidence based
System. P. Blakistons Sons and Company, Philadelphia. assessment. Lancet 359 (9317): 15891598.
Horrocks PM, Vicary DJ, Rees JE et al. (1973). Anticholiner- Rix A, Fisher RG (1972). Comparison of trihexyphinidyl and
gic withdrawal and benzhexol treatment in Parkinsons dihydromorphanthridine derivative in control of tremor of
disease. J Neurol Neurosurg Psychiatry 36: 936941. parkinsonism. South Med J 65: 13851389.
ANTICHOLINERGIC MEDICATIONS 125

Sadeh M, Braham J, Modan M (1982). Effects of anticholi- long-term anticholinergic therapy. Clin Neuropharmacol
nergic drugs on memory in Parkinsons disease. Arch 16 (5): 438443.
Neurol 39: 666667. Velasco F, Velasco M, Romo R (1982). Effect of carbachol
Strang RR (1965). Orphenadrine (Disipal) in the treatment and atropine perfusions in the mesencephalic tegmentum
of parkinsonism: a two year study of 150 patients. Med J and caudate nucleus of experimental tremor in monkeys.
Aust 2 (11): 448450. Exp Neurol 78: 450460.
Syndulko K, Gilden ER, Hansch EC et al. (1981). Decreased Yahr M (1990). Principles of medical treatment. In: G Stern
verbal memory associated with anticholinergic treatment (Ed.), Parkinsons disease. Chapman and Hall, Baltimore,
in Parkinsons disease patients. Int J Neurosci 14: 6166. pp. 495508.
Tiraboschi P, Hansen LA, Alford M et al. (2002). Early and Yahr M, Duvoisin RC (1968). Medical therapy of parkinson-
widespread cholinergic losses differentiate dementia with ism. In: PJ Vinken, GW Bruyn (Eds.), Handbook of Clin-
Lewy bodies from Alzheimer disease. Arch Gen Psy- ical Neurology: Diseases of Basal Ganglia. Elsevier
chiatry 59 (10): 946951. North-Holland, Amsterdam, pp. 283300.
Tourtellotte WW, Potvin AR, Syndulko K et al. (1982). Parkin- Yahr MD, Clough CG, Bergman KJ (1982). Cholinergic
sons disease: Congentin with Sinemet, a better response. and dopaminergic mechanisms in Parkinsons disease after
Prog Neuropsychopharmacol Biol Psychiatry 6: 5155. long term levodopa administration. Lancet 1: 709710.
van Herwaarden G, Berger HJ, Horstink MW (1993). Short- Young RR (1972). Treatment of parkinsonism. N Engl J Med
term memory in Parkinsons disease after withdrawal of 287: 10471048.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 36

Antiglutamatergic drugs in the treatment of Parkinsons disease

MARIA GRACIELA CERSOSIMO* AND FEDERICO EDUARDO MICHELI

Program of Parkinsons Disease and Other Movement Disorders, Hospital de Clnicas, University of
Buenos Aires, Buenos Aires, Argentina

36.1. Introduction associated with an increase in the phosphorylation of


serine and tyrosine residues of the striatal NMDA
In the last two decades drugs targeting glutamatergic receptors (Chase et al., 2000).
pathways have been a matter of growing interest Glutamate is such a powerful excitatory transmitter
for the treatment of a wide variety of neurologic dis- that stimulation of the NMDA receptors for prolonged
orders, including Parkinsons disease (PD) (Lipton periods of time or in excessive amounts leads to neuro-
and Rosenberg, 1994). In the vertebrate brain gluta- nal cell death as a result of a massive calcium influx,
mate is the main excitatory neurotransmitter and med- overproduction of free radicals, mitochondrial dys-
iates neurotransmission of most excitatory synapses function and apoptosis (Olney, 1969; Bonfoco et al.,
(Miller, 1998; Kemp and Mc Kernan, 2002). 1995). In addition, excitotoxicity can take place in
Hyperactivity of glutamatergic transmission app- the presence of normal levels of glutamate when the
ears to be involved in different aspects of PD. First, NMDA receptor activity is pathologically increased
glutamate-mediated excitotoxicity is one of the mech- (Zeevalk and Nicklas, 1992).
anisms proposed in the cascade of events leading to On the other hand, glutamate-mediated synaptic trans-
neuronal cell death (Olney, 1969; Olney and Ho, mission is essential for the normal functioning of the cen-
1970); therefore, agents capable of avoiding this could tral nervous system. In fact, compounds with a very strong
be good candidates for neuroprotection (Lange and Rie- antiglutamatergic action can also block the normal neuro-
derer, 1994). Second, glutamatergic pathways from the nal function resulting in unacceptable side-effects
subthalamic nucleus to the globus pallidus pars interna (Schmidt and Bubser, 1989; Kaur and Starr, 1997). Unfor-
are enhanced in the parkinsonian state, which might tunately, the clinical use of potent glutamate antagonists,
contribute in the development of PD symptoms (Nash such as MK-801 which has remarkable excitotoxicity-
and Brotchie, 2002). And third, overactivity of the str- blocking properties in experimental models, is limited
iatal glutamatergic receptors has been implicated as because of its side-effects (Lipton, 2004). The ideal anti-
one of the mechanisms underlying levodopa-induced glutamatergic drug should be able to block the NMDA
dyskinesias (Chase et al., 1996). receptor in situations of excessive activation but without
There are three types of glutamate ionotropic chan- altering glutamatergic transmission during normal condi-
nels: (1) N-methyl-D-aspartate (NMDA); (2) alpha- tions. It was recently noticed that agents with low affinity
amino-3-hydroxy-5-methyl-4-isoxazolepropionate for the Mg-binding site like the L-aminoadamatane deri-
(AMPA); and (3) kainate. The NMDA subtype recep- vatives amantadine and memantine only enter the NMDA
tor is composed of two subunits: NR1, whose presence channel under conditions of pathological glutamate
is mandatory, NR2A-D and in some cases NR3A or B exposure (Dingledine et al., 1999).
subunits (McBain and Mayer, 1994). The functional At present, the only antiglutamatergic agent used in
characteristics of these ionotropic receptors are regu- the management of PD patients is amantadine, an old
lated by their phosphorylation state. In animal models, compound that has proved to be particularly useful
lesions of the nigrostriatal dopaminergic system are for the treatment of levodopa-induced dykinesias.

*Correspondence to: Dr Maria Graciela Cersosimo, Pena 2225 5 C (CP 1126), Argentina. E-mail: mgracersosimo@arnet.com.
ar, Tel: 54-11-4806-2217, Fax: 54-11-4811-3076.
128 M. G. CERSOSIMO AND F. E. MICHELI
So far, despite the effort in developing new agents dine hydrocloride is used clinically as 100-mg cap-
with glutamate antagonist properties it has only been sules and recommended doses for PD treatment are
possible for a few of them, such as memantine, dextro- 100 mg b.i.d. or t.i.d. (Aoki and Sitar, 1988; Goetz,
methorphan, budipine, remacemide and riluzole, to be 1998).
tried in clinical studies but results were poor. Amantadine interacts with several different neuro-
transmitters systems; however the exact mechanism
of action is not established. It is classically described
36.2. Amantadine
that the drug has dopaminergic and anticholinergic
properties and more recently amantadines antagonist
Amantadine hydrocloride was initially introduced as
activity of NMDA glutamatergic receptors has been
an antiviral agent effective against A2 influenza
reported (Stoof et al., 1992). Amantadine exerts its
(Jackson et al., 1963). Its antiparkinsonian effect was
dopaminergic effects, acting at presynaptic and postsy-
discovered fortuitously in a 58-year-old woman with
naptic levels. Presynaptically, the drug interacts with
PD who experienced a remission of her symptoms
dopaminergic neurotransmission in two ways: first,
while taking amantadine to prevent influenza. After
enhancing the release of dopamine and other cathecho-
this single case, the authors conducted a trial with
lamines from dopaminergic terminals and second,
163 patients with PD and observed improvement in
inhibiting the reuptake process. Postsynaptically, a
66% of them. Since then it has been acknowledged
direct action of the drug on dopamine receptors has
that the beneficial effects on PD are usually short-lived
been described, resulting in changes in dopamine
(Schwab et al., 1969).
receptor affinity. Additionally, studies on the chronic
Over the years, the introduction of a number of
effect of amantadine showed an increased striatal spir-
other different agents, including dopamine agonists,
operidol binding, suggesting D2 dopamine receptor
resulted in a more limited use of amantadine in part
blockade (Von Voigtlander and Moore, 1971; Bailey
due to its modest antiparkinsonian action but also
and Stone, 1975; Gianutsos et al., 1985).
because of the short duration of the clinical benefit
Stoof and colleagues (1992) reported amantadines
(Factor and Molho, 1999). Many clinical trials have
antiglutamatergic properties as demonstrated by
investigated the efficacy of amantadine and recent
NMDA receptor blockade in the rat neostriatum
reviews on PD treatment placed amantadine as a
at therapeutic doses. It has been demonstrated that
second-line therapy for PD.
amantadine acts as a non-competitive NMDA antagonist
The interest in this agent has recently reappeared
producing a concentration-, time- and voltage-dependent
since Shannon et al. (1987) reported that amantadine
blockade of open NMDA channels (Kornhuber et al.,
improved motor fluctuations and Stoof et al. (1992)
1991; Sobolevski et al., 1998). Recently it has been
demonstrated amantadines activity at glutamate
recognized that amantadine belongs to the type of
receptors, suggesting that its antiparkinsonian effects
blocker manifesting the so-called trapping block of
might be related to NMDA receptor blockade.
NMDA channels. These drugs enter an open NMDA
Factor et al. (1998) reported that amantadine with-
channel and bind to its blocking site located deep in
drawal in patients treated for longer than 1 year may
the pore. The blocking molecules remain in the pore
result in a substantial increase of motor disability or
for a relatively long time, being trapped behind the
delirium. In addition, several controversial and unre-
closed activation gate. Agonist reapplication opens the
solved issues regarding the mechanism of action of
gate and allows the blocker to leave the channel. The
amantadine as well as the duration of the beneficial
molecular mechanisms of the trapping of blockers are
response are still open questions.
at present unclear (Banplied et al., 1997; Sobolevski
et al., 1998).
36.2.1. Basic pharmacology and mechanism Interestingly, despite the many different pharmaco-
of action logical properties attributed to this drug, it is not clear
how they contribute to its clinical effect.
Amantadine hydrocloride is L-amino adamantamine, Furthermore, the classic idea that the antipar-
the salt of a symmetric 10-carbon primary amine. It kinsonian effect of amantadine is secondary to its dopa-
is a stable, white, crystalline compound that is freely minergic or anticholinergic activities is not supported
soluble in water (Schwab et al., 1969). In humans, it by more recent studies. In fact, therapeutic doses of
is well absorbed after oral administration. Blood levels amantadine in vitro are not associated with increased
peak 14 hours after an oral dose of 2.5 mg/kg. The extracellular brain dopamine levels. Similarly, the pre-
drug has a plasma half-life of 1028.5 hours and is sumed anticholinergic effect is also unlikely as amanta-
excreted largely unmetabolized in the urine. Amanta- dine only binds to acetylcholine receptors at very high
ANTIGLUTAMATERGIC DRUGS IN THE TREATMENT OF PARKINSONS DISEASE 129
concentrations (Brown and Redfern, 1976; Verhagen two drugs: both drugs in combination resulted in a
Metman et al., 1998a). reduction of 40% of total disability.
Regarding the antidyskinetic effect of amantadine, Walker et al. (1972) also showed that amantadine is
compelling data support the hypothesis that it is related superior to placebo when administered in patients
to the anti-NMDA properties of the drug. Furthermore, receiving anticholinergics. In a double-blind, cross-
some authors propose NMDA antagonism of amantadine over trial in 42 PD patients they found amantadine
as the mechanism for its antiparkinsonian effect (Stoof was 74% superior to placebo.
et al., 1992). As adjunct therapy of levodopa, amantadine is
somewhat efficacious in the symptomatic treatment
36.2.2. Symptomatic treatment of parkinsonism of PD. In a double-blind, randomized cross-over study,
Savery (1977) added amantadine to 42 PD patients
Amantadine has a mild antiparkinsonian effect and treated with levodopa/carbidopa. The clinical eva-
appears to be more effective in the control of bradykine- luation after a period of 9 weeks resulted in a signi-
sia and rigidity and less effective in tremor. Schwab ficant improvement in symptoms. Similarly, Fehling
et al. (1969) conducted the first clinical experience to (1973) in a double-blind, randomized cross-over study
assess the efficacy of amantadine in 163 patients with compared the effect of amantadine or placebo in 21
PD. This was an open-label non-controlled study; patients receiving levodopa/carbidopa and found
patients received a maximum daily dose of 200 mg amantadine to be significantly more effective than
amantadine in addition to their usual antiparkinsonian placebo in improving PD symptoms. Nonetheless,
medication. The authors found improvement of akinesia, these findings could not be confirmed by other authors
rigidity and tremor in 66% of patients and noted that in who found no differences in the evaluation of patients
58% the benefit was sustained for a period of 38 months. treated with levodopa/carbidopa with and without
Since that first report, several trials were performed amantadine (Millac et al., 1970; Callagham et al.,
to investigate amantadines effectiveness in the treat- 1974).
ment of PD as monotherapy or associated with other It is generally described that amantadine produces
antiparkinsonian drugs. a transient clinical benefit with a loss of efficacy after
A double-blind, placebo-controlled cross-over study 612 months, probably due to a tachyphylaxis phe-
was performed on 30 PD patients, all but 3 who were nomenon (Schwab et al., 1969). Nonetheless, some
on anticholinergic or antihistaminic drugs; patients authors have questioned the validity of this concept.
received amantadine as monotherapy. Clinical assess- Factor et al. (1998) in a critical review of 22 studies
ments included evaluations of tremor, rigidity, all phy- of amantadine found that many of them had included
sical signs, daily activities, repetitive motions and an patients with postencephalitic parkinsonism, multiple
overall average. Amantadine resulted in a statistically system atrophy or progressive supranuclear palsy.
significant 12% overall improvement over placebo. The length of amantadine therapy ranged from 4 days
Ten patients continued treatment for 1012 months. to 12 months and in 13 studies a decline of efficacy
Improvements in tremor and rigidity remained rela- was not mentioned, suggesting there are not enough
tively constant although there was some apparent loss reliable data to support this notion (Marsden, 1988;
of efficacy in timed tests (Butzer et al., 1975). Factor and Molho, 1999). Zeldowicz and Huberman
Fahn and Isgreen (1975) conducted a randomized (1973) performed a clinical trial on 77 PD patients to
double-blind, cross-over, placebo-controlled trial evaluate whether there is a decline in efficacy. A total
in 23 Parkinsons disease patients. During the first of 19 patients had good to excellent response to aman-
cross-over period the patients were randomized to pla- tadine monotherapy and maintained that improvement
cebo or amantadine 200 mg/day. The authors reported for an average of 21 months, with the longest duration
a favorable response in 16 patients (70%). The most being 30 months. In addition, a marked deterioration
common adverse reactions were insomnia, anorexia, took place when amantadine was switched in a random
dizziness, nervousness, irritability, depression and fashion to placebo.
sleepiness.
Parkes and colleagues (1974) compared the efficacy 36.2.3. Treatment of motor complications
of amantadine (200 mg/day) and anticholinergics
(benzhexol 8 mg/day) or both in 17 PD patients in a Amantadine was found to be effective in the treatment
randomized, double-blind cross-over trial. The clinical of levodopa-induced dyskinesias in patients with PD.
improvement observed with amantadine or benzhexol Preclinical studies suggest that the development of
as monotherapy was a reduction of 15% in functional levodopa-induced motor complications is associated
disability without significant differences between the with an increase in phosphorylation state of NMDA
130 M. G. CERSOSIMO AND F. E. MICHELI
receptors in striatal medium spiny neurons; as a res- assessments. Results showed that the reduction in dyski-
ult, synaptic efficacy is enhanced and corticostriatal nesias severity was 56% compared with 60% 1 year
glutamatergic input is amplified, affecting striatal before. The authors conclude that the beneficial effect
GABAergic output and probably leading to motor res- of amantadine on levodopa-induced dyskinesias is
ponse complications. Therefore, it is conceivable that maintained for at least 1 year after treatment initiation
amantadine exerts its antidyskinetic effect by normal- (Metman et al., 1999).
izing striatal NMDA receptor hyperfunction (Chase In the two previous studies amantadine was admini-
et al., 1996; Dunah et al., 2000). Verhagen Metman strated orally and onset of benefit was observed from
and colleagues (1998a) conducted the first controlled days to weeks after treatment initiation. Del Dotto and
clinical trial to evaluate the effects of amantadine on colleagues (2001) investigated the possible occurrence
levodopa-associated dyskinesias and motor fluctua- of an acute effect of amantadine after an intravenous
tions in 18 advanced PD patients (average age 60 infusion. They conducted a randomized, double-blind,
years, Hoehn and Yahr stage IIV) in a double-blind, placebo-controlled study including 9 PD patients with
cross-over, placebo-controlled study during a 6-week peak-dose levodopa-induced choreiform dyskinesias.
period. All patients received amantadine or placebo Their mean age was 59.7 years, disease duration was
for 3 weeks. Amantadine doses ranged from 100 to 8.4 years, Hoehn and Yahr stage III in the off condi-
400 mg/day depending on age, renal function and tol- tion and the daily dose of levodopa immediately prior
erance. At the end of each study period the patients to study was 860 mg. All patients were evaluated on 2
received an intravenous levodopa infusion for 7 hours different days for 5 hours while taking their usual anti-
at an individually determined optimal rate, defined as parkinsonian medications. The 9 patients received their
the lowest rate producing the maximal antiparkinso- first morning levodopa dose, followed by a 2-hour in-
nian effect. The evaluations were performed during travenous amantadine (200 mg) or placebo infusion.
the last 2 hours of the levodopa infusion and chorei- Parkinsonian symptoms and dyskinesias were assessed
form dyskinesias and parkinsonian symptoms were every 15 minutes during the infusion and 3 hours post-
scored every 10 minutes. Dyskinesias were also video- infusion using the UPDRS scale part III, tapping test
taped and subsequently scored by a second blinded and the AIMS. Results showed that the average of dys-
neurologist. Parkinsonian symptoms and dyskinesias kinesia scores during amantadine infusion was 50%
were assessed using the Unified Parkinsons Disease lower when compared to placebo (P < 0.01). These
Rating Scale (UPDRS) part III, items 20, 22, 23, 26 results indicate that the antidyskinetic effect of amanta-
and 31 and a modified Abnormal Involuntary Move- dine occurs acutely and does not require days or weeks
ment Scale (AIMS). Motor fluctuations were assessed to develop (Del Dotto et al., 2001).
with the UPDRS part IV, items 32 (duration of dyski- The antidyskinetic effect of amantadine was also con-
nesias), 33 (severity of dyskinesias) and 39 (proportion firmed by other authors. Snow et al. (2000) performed a
of the day in the off state), as well as patient diaries double-blind, placebo-controlled, cross-over study to
on the last 2 or 3 days of each study arm. The sta- assess the effect of amantadine (200 mg/day oral) versus
tistical analysis of measures showed a significant placebo on levodopa-induced dyskinesias in 24 patients
(P < 0.001) reduction in the severity of dyskinesias with PD showing a significant (P < 0.004) reduction
which was 60% lower with amantadine as compared of 24% in the total dyskinesia score after amantadine
to placebo. In this study amantadine substantially ame- administration.
liorated levodopa-induced peak-dose dyskinesias with-
out worsening parkinsonian symptoms. Dysphasic 36.2.4. Prevention of disease progression
dyskinesias could not be evaluated as they did not occur
during optimal levodopa infusion rate. Only 4 patients Several recent studies suggest that amantadine has neu-
withdrew from the study because of adverse reactions roprotective properties in addition to its symptomatic
(confusion 1, hallucinations 1, palpitations 1 and nausea effects. It has been suggested that the enhanced excita-
1) (Verhagen Metman et al., 1998a). One year later tory amino acid neurotransmission may play a role in
Metman et al. (1999) reported the results of a 1-year the pathogenesis of several chronic neurodegenerative
follow-up of the previously reported study. Of the initial diseases, including PD (Kornhuber et al., 1994; Danysz
18 patients, 17 participated in this study with a non- et al., 1997). Studies in monkeys indicate that excitatory
randomized, double-blind, placebo-controlled design. amino acids such as glutamate are involved in the path-
Ten days prior to the follow-up assessment, amantadine ophysiological cascade of 1-methyl-4-phenyl-1236-
was replaced by capsules containing either amantadine tetrahydropyridine (MPTP)-induced neuronal cell death.
or placebo. On the test day, the patients received an This observation supports the hypothesis that glutamate
intravenous infusion of levodopa followed by motor antagonists such as amantadine may be able to delay
ANTIGLUTAMATERGIC DRUGS IN THE TREATMENT OF PARKINSONS DISEASE 131
the progression of the disease (Lange et al., 1997). In Amantadine toxicity may be associated with over-
addition, multiple in vitro studies have recently shown dose or renal insufficiency as 90% of an oral dose
the neuroprotective properties of amantadine and its is excreted unchanged in the urine (Macchio et al.,
congener compound memantine at therapeutically used 1993). These toxic effects may be exacerbated by the
doses (Kornhuber et al., 1994). concomitant use of anticholinergic agents.
Uitti and colleagues (1996) studied survival in Withdrawal effects of amantadine are not frequent
patients with PD and other parkinsonian syndromes but may be serious. Factor and colleagues (1998)
employing standard survival curves and a Cox regres- reported 3 PD patients who, after gradually tapering
sion model to identify independent predictive variables amantadine, experienced acute delirium, paranoia and
for survival. Patients treated with and without aman- agitation in addition to worsening of motor symptoms.
tadine were similar in terms of age, gender, type of In all of them the syndrome rapidly resolved after
parkinsonism, Hoehn and Yahr staging and cognitive amantadine restitution. Some characteristics were
status. The authors found that the use of amantadine common among the 3 patients. They were elderly
is an independent predictor of improved survival. and had hallucinations in the past, dementia, advanced
Glutamate receptor antagonist drugs appear to be PD and a long duration of amantadine therapy, repre-
promising agents for the neuroprotective therapy of senting possible risk factors for this complication
PD (Kornhuber and Weller, 1996). (Factor et al., 1998).
Several compounds with antiparkinsonian effects,
such as amantadine, memantine and budipine, have
36.3. Other antiglutamatergic drugs
been shown to be non-competitive NMDA receptor
antagonists, are candidates for clinical trials (Lange 36.3.1. Memantine
et al., 1997) and appear to be promising agents for
the neuroprotective therapy of PD (Kornhuber and Memantine (1-amino-3,5-dimethyladamantane) is an
Weller, 1996). L-aminoadamantane derivative first synthesized in
1968. It has been proposed that memantine may be
36.2.5. Side-effects useful for the treatment of PD symptoms (Fischer
et al., 1977; Schneider et al., 1984). Also, some case
Amantadine is usually well tolerated. The more fre- reports have suggested the utility of memantine in
quent adverse reactions associated with the use of levodopa-induced dyskinesia therapy (Hanagasi et al.,
amantadine include: insomnia, anxiety, dizziness, 2000) However, evidence to conclude about its effi-
impaired coordination, nervousness, nausea and vomit- cacy in any indication for PD is so far insufficient.
ing. These side-effects are usually mild, although they Clinical trials have shown that memantine is effective
can be severe in elderly patients. In fewer than 5% of in the treatment of Alzheimers disease (Reisberg
patients, irritability, headaches, depression, ataxia, et al., 2003).
confusion, hallucinations, nightmares, somnolence, Memantine is an uncompetitive and low-affinity
agitation, diarrhea, constipation, livedo reticularis and NMDA receptor antagonist with voltage-dependent
xerostomia are reported. Dry mouth and blurred vision binding (Bormann, 1989). Unlike amantadine, meman-
are considered peripheral anticholinergic side-effects. tine has no dopaminergic or anticholinergic actions.
Livedo reticularis is more often in women and is fre- Memantine has a three-ring structure with a bridge-
quently associated with persistent ankle edema (Lof- head amine that binds to the Mg binding site of the
fler et al., 1998). Unusual adverse effects include NMDA receptor (Bormann, 1989; Lipton, 1993).
hyperkinesias, urinary retention, rash and decreased Memantine is completely absorbed from gastroin-
libido whereas toxic manifestations of amantadine testinal tract with peak blood levels occurring 2030
include acute psychosis, coma, cardiovascular minutes after an oral dose of 5 mg/kg. The mean
toxicity and death. half-life of memantine is 60100 hours and approxi-
More rare adverse reactions have been reported as mately 80% of the drug circulates unchanged. The
isolated cases such as vocal myoclonus, heart failure usual recommended dose for patients with PD is 10
or peripheral neuropathy (Vale and Maclean, 1977; mg t.i.d. (Jarvis and Figgit, 2003; Lundbeck, 2002).
Pfeiffer, 1996). Shulman and colleagues (1999) Only a few qualified studies assessing the efficacy
reported a case of sensory motor peripheral neuropathy of memantine in PD are found in the literature. Merello
secondary to chronic administration of amantadine in a et al. (1999) performed a randomized, placebo-controlled,
48-year-old woman where the discontinuation of the cross-over study in 12 PD patients with motor fluctuations
drug resulted in the resolution of trophic skin ulcers, to asses the effect of memantine on the cardinal symptoms
paresthesias and distal weakness. of PD and levodopa-induced dyskinesias.
132 M. G. CERSOSIMO AND F. E. MICHELI
A significant improvement of motor symptoms in the ents reporting subjective improvement were included
off and on states evaluated by means of the UPDRS in a second phase of the study, with a double-blind,
motor scale was observed. No significant effect of placebo-controlled, cross-over design consisting of
memantine on levodopa-induced dyskinesias was 2-week arms separated by 1-week washout. Six patients
reported (Merello et al., 1999). were included in the second part of the study. Although
Memantine is usually well tolerated; reported side- parkinsonian scores were not modified by dextrometh-
effects included diarrhea, dizziness, headache, hallu- orphan, dyskinesias improved by 25 % (P  0.05). The
cinations, agitation, insomnia and urinary incontinence severity of motor fluctuations also improved significantly
(Jarvis and Figgitt, 2003). by 66% (P  0.05), according to the UPDRS part IV-item
There are no data available regarding the utility of 39. Adverse events included decreased levodopa effi-
memantine in the prevention of PD progression. cacy, increased dystonia, increase in pre-existing impo-
tence, nausea, perspiration and drowsiness (Verhagen
36.3.2. Dextromethorphan Metman et al., 1998b).
At present data are insufficient to conclude on effi-
Dextromethorphan is a widely used and safe antitus- cacy and safety of dextromethorphan in any indication
sive agent with low affinity and uncompetitive antago- of PD.
nist properties of the NMDA receptors. Based on this
knowledge, dextromethorphan was tried in PD and a 36.3.3. Budipine
variety of conditions where glutamate overactivity is
supposed to play a pathogenic role (Wong et al., Budipine (1-t-butyl-4,4-diphenylpiperidine) has a com-
1988; Church et al., 1989). plex pharmacological profile interacting with different
Dextromethorphan is well absorbed in the gastroin- neurotransmitter systems. Besides its NMDA receptor
testinal tract with peak blood levels occurring 14 hours antagonist properties it also influences GABAergic,
after an oral dose of 2.5 mg/kg and a median half-life of noradrinergic, serotoninergic and cholinergic transmis-
2 hours. There is genetic polymorphism for the oxida- sion (Jackisch et al., 1993; Kolckgether et al., 1996;
tive O-demethylation being both extensive and poor Eltze, 1999).
metabolizers. Half-life in poor metabolizers can reach Up to 2000 budipine was still available in some
up to 45 hours. Recommended dosage varies between European countries but cardiac side-effects, including
100 and 200 mg/day (Woodworth et al., 1987; Schadel arrhythmias due to an acquired long QT syndrome,
et al., 1995). led to important restrictions, further limiting its use
Only two open-label and non-controlled studies in clinical practice (Scholz et al., 2003).
assessing the efficacy of dextromethorphan for the Przuntek et al. (2002) conducted a multicenter,
treatment of motor symptoms of PD in non-fluctuating placebo-controlled, double-blind study in 84 PD patients
patients have been carried out. Montastruc et al. (1994) to assess the efficacy of 20 mg budipine three times a
performed a study to investigate the effect of adding day in addition to a stable optimum dopaminergic regi-
dextromethorphan 90 mg/day to the therapy of 13 PD men. Duration of the study was 4 months and patients were
patients. Evaluations with the UPDRS motor scale done evaluated with the Columbia University Rating Scale
at baseline and after 1 month of treatment did not show (CURS). Budipine significantly (P  0.01) decreased
any change in the scores (Montastruc et al., 1994). the CURS subscores for rigidity, tremor and akinesia com-
Conversely, Bonuccelli et al. (1992) found signifi- pared with placebo. Adverse events reported in the budi-
cant improvement on UPDRS motor scale scores of pine group were dizziness, dry mouth, loss of appetite,
6 de novo PD patients and another 6, in which dex- nervousness and visual dysfunction (Przuntek et al., 2002).
tromethorphan was added to their previous treatment. The usefulness of budipine in the treatment of
In this study doses of 180 mg/day were associated patients with motor fluctuations was investigated by
with significant improvement in tremor, rigidity and Spieker et al. (1999) in an open-label study performed
finger-tapping. in 7 PD patients medicated with budipine at doses of
Verhagen Metman et al. (1998b) conducted a study 40 mg/day in addition to their dopaminergic therapy.
with dextromethorphan in patients with levodopa- The duration of the study was 8 weeks and assess-
induced dyskinesia and motor fluctuations. Eighteen ments were UPDRS motor section in the on state
patients were included to initial open-label dose esca- and a diary over 7 days recording hours of on/off
lation screening. The dose of dextromethorphan was states, dyskinesias or dystonia. Results showed that
increased up to 180 mg/day or until side-effects occurred. time off decreased in 5 of the 7 patients; also motor
All patients in addition received quinidine 100 mg b.i.d. scores during the on period improved. Peak-dose
to inhibit O-demethylation of dextromethorphan. Pati- dyskinesias did not occur in any of the patients.
ANTIGLUTAMATERGIC DRUGS IN THE TREATMENT OF PARKINSONS DISEASE 133
At present there is agreement that the risk-to- scores in the off state (P 0.04). Finally, the group
benefit ratio for the use of budipine is unfavorable of remacemide 600 mg/day only showed significant
because of the significant increased risk of cardiac (P 0.04) improvement in the UPDRS part II scores
arrhythmias. This is the reason why the use of budipine in the on state.
in the treatment of PD is not recommended. The most common dose-related adverse events
associated with remacemide were nausea and dizziness
36.3.4. Remacemide (Shoulson et al., 2001). More rarely, headache, abnor-
mal vision, vomiting and hypokinesia can ocur (Clarke
Remacemide hydrochloride is a low-affinity, non- et al., 2001).
competitive NMDA channel antagonist which has Remacemide appears to be a safe and well-tolerated
shown antiparkinsonian efficacy in rodent and primate drug; however, larger trials are required to establish
models. The active metabolite of remacemide AR-R its efficacy in the treatment of levodopa-induced
12495 also has a moderate affinity for the NMDA dyskinesias and parkinsonian symptoms.
receptor and both interact with voltage-dependent neu-
ronal sodium channels (Greenamyre et al., 1994; 36.3.5. Riluzole
Schachter and Tarsy, 2000).
A multicenter randomized, placebo-controlled, The antiglutamatergic agent riluzole (2-amino-6-trifluor-
double-blind, parallel-group, dose-ranging study of omrthoxy-benzothiazole) is currently the only drug
remacemide was performed in 200 early PD patients approved for the treatment of amyotrophic lateral sclero-
who were not receiving levodopa or dopamine agonists. sis and its effect can only extend the survival of patients
Subjects were randomized to remacemide 150, 300 or for a few months (Bensimon et al., 1994). Riluzole has
600 mg daily, or matching placebo for 5 weeks. The multiple mechanisms of action: (1) inactivation of vol-
study failed to demonstrate any symptomatic effect of tage-dependent sodium channels (Benoit and Escande,
remacemide as monotherapy in patients with early PD 1991; Hubert et al., 1994); (2) non-competitive blockade
(Parkinson Study Group, 2000). of postsynaptic excitatory amino acid receptors (Bena-
Shoulson et al. (2001) assessed the safety, tolerabil- vides et al., 1985; Debono et al., 1993); and (3) presynap-
ity and efficacy of remacemide adjunct treatment in tic inhibition of glutamate release (Cheramy et al., 1986,
279 PD patients with motor fluctuations receiving 1992). Preclinical data suggest that riluzole has a neuro-
levodopa. The authors conducted a randomized, dou- protective effect in animal models of PD (Boireau
ble-blind, placebo-controlled, parallel-dose-ranging et al., 1994; Bezard et al., 1998).
study during a 7-week treatment period. Remacemide Jankovic and Hunter (2002) assessed neuroprotec-
doses were 300 mg b.i.d. and 600 mg q.i.d. Results tive properties of riluzole in a double-blind placebo-
showed that remacemide was safe and well tolerated controlled study in 20 patients with early PD. Subjects
as adjunct to dopaminergic therapy in patients with were randomized to 50 mg b.i.d. of riluzole or matching
PD and motor fluctuations. However, UPDRS motor placebo. All patients were evaluated at baseline, 1, 3
scores and percentage of on time did not show any and 6 months and following a 6-week washout period.
significant improvement in the remacemide group After the washout, all subjects were offered to enroll
compared with placebo (Shoulson et al., 2001). in a 1-year extension study. Assessments included
The Parkinson Study Group (2001) evaluated the UPDRS scale, Hoehn and Yahr stage and Schwab
effect of remacemide hydrochloride in the treatment and England scale. Results did not show any sympto-
of levodopa-induced dyskinesias. A total of 39 PD matic effect of riluzole on the UPDRS score and no
subjects with disabling dyskinesias were included in changes in the Hoehn and Yahr stage. Among the 17
a multicenter, randomized double-blind, placebo- patients who decided to continue in the extension phase
controlled, parallel-group study during a period of 2 of the study, the deterioration in the UPDRS score was
weeks. Daily doses of remacemide were 150, 300 or more pronounced in the placebo group but this differ-
600 mg or matching placebo. The study failed to ence was not significant. The study failed to show
demonstrated any significant changes in dyskinesia evidence of symptomatic or neuroprotective effects of
measures, but a statistically significant improvement riluzole (Jankovic and Hunter, 2002).
in the off state UPDRS motor score (P 0.01) The effect of riluzole in PD patients with dyskine-
and in the UPDRS part II (P 0.04) was observed sias was investigated by Braz et al. (2004) in a dou-
in patients receiving remacemide 150 mg/day. In sub- ble-blind, placebo-controlled pilot study. A total of
jects receiving remacemide 300 mg/day a significant 16 patients were randomly treated with riluzole 50
(P 0.004) improvement in the percentage of on mg b.i.d. or matching placebo during 7 consecutive
state daily hours was found and in the UPDRS part II days. The patients were evaluated at baseline and at
134 M. G. CERSOSIMO AND F. E. MICHELI
the end of the treatment period. Assessments included
Butzer JF, Silver DE, Sahs AL (1975). Amantadine in Par-
UPDRS part III and IV, Hoehn and Yahr stage and the kinsons disease. A double blind, placebo controlled,
Larsen scale for dyskinesia. On the day of the evaluations crossover study with long term follow up. Neurology
the patients were asked to assist after a washout period 25 (7): 603606.
of at least 8 hours without taking levodopa or any other Callagham N, McIlroy M, OConnor M (1974). Treatment of
antiparkinsonian medication. For the evaluation all Parkinsons disease with levodopa and amantadine used as
subjects were treated with subcutaneous apomorphine a single drugs and in combined therapy. Ir J Med Sci 143:
1 mg every 15 minutes until the on condition was 6778.
achieved. No significant differences were found between Chase TN, Engber TM, Mouradian MM (1996). Contribution
the two groups. Riluzole was not able to reduce apomor- of dopaminergic and glutamatergic mechanisms to the
pathogenesis of motor response complications in Parkin-
phine-induced dykinesia (Braz et al., 2004).
sons disease. Adv Neurol 69: 497501.
Chase TN, Oh JD, Konitsiotis S (2000). Antiparkinsonian and
References antidiskynetic activity of drugs targeting central glutama-
tergic mechanisms. J Neurol 247 (Suppl 2): II/36II/42.
Aoki FY, Sitar DS (1988). Clinical pharmacokinetics of aman- Cheramy A, Romo R, Godeheu G et al. (1986). In vivo pre-
tadine hydrochloride. Clin Pharmacokinet 14: 3551. synaptic control of dopamine release in the cat caudate
Bailey EV, Stone TW (1975). The mechanism of action of nucleus. Facilitatory or inhibition influence of L-gluta-
amantadine in parkinsonism: a review. Arch Int Pharma- mate. Neuroscience 19: 10811090.
codyn Ther 216: 246262. Cheramy A, Barbeito L, Godeheu G et al. (1992). Riluzole
Banplied TA, Boeckman FA, Aizenman E et al. (1997). Trap- inhibits the release of glutamate in the caudate nucleus
ping channel block of NMDA receptors activated of the cat in vivo. Neurosci Lett 147: 209212.
responses by amantadine and memantine. J Neurophysiol Church J, Jones MG, Davies SN et al. (1989). Antitussive
77 (1): 309323. agents as N-methylaspartate antagonists: further studies.
Benavides J, Camelin JC, Mitrani N et al. (1985). 2-Amino- Can J Physiol Pharmacol 67: 561567.
6-trifluoromethoxy benzothiazole, a possible antagonist of Clarke CE, Cooper JA, Holdich TA (2001). TREMOR Study
excitatory amino acid neurotransmission. Biochemical Group. A randomized, double blind, placebo controlled,
properties. Neuropharmacology 24: 10851092. ascending dose tolerability and safety study of remacemide
Benoit E, Escande D (1991). Riluzole specifically blocks as adjuvant therapy in Parkinsons disease with response
inactivated Na channels in myelinated nerve fibre. fluctuations. Clin Neuropharmacol 24 (3): 133138.
Pflugers Arch 419: 603609. Danysz W, Parsons CG, Kornhuber J et al. (1997). Ami-
Bensimon G, Lacomblez L, Meininger V (1994). The ALS noadamantanes as NMDA receptors antagonists and
Riluzole Study Group. A controlled trial of riluzole in antiparkinsonian agents preclinical studies. Neurosci
amyotrophic lateral sclerosis. N Engl J Med 330: 585601. Biobehav Rev 21 (4): 455468.
Bezard E, Stutzmann JM, Imbert C et al. (1998). Riluzole Debono MW, Le Guern J, Canton T et al. (1993). Inhibition
delayed appearance of parkinsonian motor abnormalities by riluzole of electrophysiological responses mediated by
in a chronic MPTP monkey model. Eur J Pharmacol rat kainite and NMDA receptors expressed in Xenopus
356: 101104. oocytes. Eur J Pharmacol 235: 283289.
Boireau A, Miquet JM, Dubedat P et al. (1994). Riluzole and Del Dotto P, Pavese N, Gambaccini G et al. (2001). Intrave-
experimental parkinsonism: partial antagonism of MPP nous amantadine improves levodopa-induced dyskinesias:
induced increase in striatal extracellular dopamine in rats an acute double blind placebo controlled study. Mov Dis-
in vivo. Neuroreport 5: 21572160. ord 16 (3): 515520.
Bonfoco E, Krainc D, Ankarcrona M et al. (1995). Apoptosis Dingledine R, Borges K, Bowie D et al. (1999). The gluta-
and necrosis: two distinct events induced respectively by mate receptor ion channels. Pharmacol Rev 51: 851.
mild and intense insults with NMDA or nitric oxide/super- Dunah AW, Wang Y, Yasuda RP et al. (2000). Alteration in
oxide in cortical cell cultures. Proc Natl Acad Sci USA 92: subunit expression, composition, and phosphorylation of
71627166. striatal N-methyl-D-aspartate glutamate receptors in a rat
Bonuccelli U, Del Dotto P, Piccini P et al. (1992). Dex- 6-hydroxidopamine model of Parkinsons disease. Mol
tromethorphan and parkinsonism. Lancet 340: 53. Pharmacol 57: 342352.
Bormann J (1989). Memantine is a potent blocker of Eltze M (1999). Multiple mechanisms of action: the pharma-
N-methyl-D-aspartate (NMDA) receptor channels. Eur J cological profile of budipine. J Neural Transm Suppl 56:
Pharmacol 166 (3): 591592. 83105.
Braz CA, Borges V, Ferraz HB (2004). Effect of riluzole on Factor SA, Molho ES (1999). Tansient benefit of amantadine
dyskinesia and duration of the on state in Parkinsons dis- in Parkinsons disease: the facts about the myth. Mov Dis-
ease patients: a double blind, placebo controlled pilot ord 14 (3): 515517.
study. Clin Neuropharmacol 27 (1): 2529. Factor SA, Molho ES, Brown DL (1998). Acute delirium
Brown F, Redfern PH (1976). Studies on mechanism of after withdrawal of amantadine in Parkinsons disease.
action of amantadine. Br J Pharmacol 58: 561567. Neurology 50: 14561458.
ANTIGLUTAMATERGIC DRUGS IN THE TREATMENT OF PARKINSONS DISEASE 135

Fahn S, Isgreen WP (1975). Long term evaluation of aman- Kornhuber J, Weller M, Schoppmeyer K et al. (1994). Aman-
tadine and levodopa combination in parkinsonism by tadine and memantine are NMDA receptors antagonists
double blind crossover analyses. Neurology 25 (8): with neuroprotective properties. J Neural Transm Suppl
695700. 43: 91104.
Fehling C (1973). The effect of adding amantadine to Lange KW, Riederer P (1994). Glutamatergic drugs in
optimum L-dopa dosage in Parkinsons syndrome. Acta Parkinsons disease. Life Sci 55 (2526): 20672075.
Neurol Scand 49: 245251. Lange KW, Kornhuber J, Riederer P (1997). Dopamine/glu-
Fischer PA, Jacobi P, Schneider E, Schonberger B tamate interactions in Parkinson s disease. Neurosci Bio-
(1977). Effects of intravenous administration of meman- behav Rev 21 (4): 393400.
tine in parkinsonian patients. Arzneimittelforschung 27: Lipton SA (1993). Prospects for clinically tolerated NMDA
10221023. antagonists: open channel blockers and alternative redox
Gianutsos G, Chute S, Dunn JP (1985). Pharmacological states of nitric oxide. Trends Neurosci 16: 527532.
changes in dopaminergic systems induced by long term Lipton SA (2004). Failures and successes of NMDA receptor
administration of amantadine. Eur J Pharmacol 110: antagonists: molecular basis for the use of open-channel
357361. blockers like memantine in the treatment of acute and
Goetz C (1998). New lessons from old drugs. Amantadine chronic neurologic insults. NeuroRx 1: 101110.
and Parkinsons disease. Neurology 50: 12111212. Lipton SA, Rosenberg RA (1994). Mechanisms of disease:
Greenamyre JT, Eller RV, Zhang Z et al. (1994). Antiparkin- excitatory amino acids as a final common pathway in neu-
sonian effect of remacemide hydrochloride, a glutamate rologic disorders. N Eng J Med 330: 613622.
antagonist in rodent and primate models of Parkinsons Loffler H, Habermann B, Effendy I (1998). Amantadine
disease. Ann Neurol 35: 655661. induced livedo reticularis. Hautarzt 49 (3): 224227.
Hanagasi HA, Kaptanoglu G, Sahin HA et al. (2000). Lundbeck H (2002). Summary of product characteristics.
The use of the NMDA antagonist memantine in drug resis- Ebixa (memantine hydrochloride). Lunbeck H A/S, Valby,
tant dyskinesias resulting from L-dopa. Mov Disord Denmark, 113.
15 (5): 10161017. McBain CL, Mayer ML (1994). N-methyl-aspartic-acid
Hubert JP, Delumeau JC, Glowinski J et al. (1994). Antagon- receptor structure and function. Physiol Rev 74: 723760.
ism by riluzole of entry of calcium evoked by NMDA and Macchio GJ, Ito V, Sahgal V (1993). Amantadine-induced
veratridine in rat cultured granule cells: evidence for a coma. Arch Phys Med Rehabil 74 (10): 11191120.
dual mechanism of action. Br J Pharmacol 113: 261267. Marsden CD (1988). The drug therapy of early Parkinsons
Jackisch R, Huang HY, Reimann W et al. (1993). Effects of disease. In: MB Stern, HH Hurtig (Eds.), The Compre-
the antiparkinsonian drug budipine on neurotransmitter hensive Management of Parkinsons disease. PMA Pub-
release in central nervous system tissue in vitro. J Pharma- lishing Corp, New York, NY, pp. 7988.
col Exp Ther 264: 889898. Merello M, Nouzeilles MI, Cammarota A et al. (1999). Effect
Jackson GG, Muldoon RL, Akers LW (1963). Serological of memantine (NMDA antagonist) on Parkinsons disease:
evidence for prevention of influenza infection in volun- a double blind crossover randomized study. Clin Neuro-
teers by and anti-influenza drug adamantanamine hydro- pharmacol 22 (5): 273276.
chloride. Antimicrob Agents Chemother 3: 703707. Metman LV, Del Dotto P, LePoole K et al. (1999). Amanta-
Jankovic J, Hunter C (2002). A double blind, placebo con- dine for levodopa induced dyskinesias: a 1-year follow up
trolled and longitudinal study of riluzole in early Parkin- study. Arch Neurol 56 (11): 13831386.
sons disease. Parkinsonism Relat Disord 8 (4): 271276. Millac P, Hasan I, Espir ML et al. (1970). Treatment of Par-
Jarvis B, Figgitt DP (2003). Memantine. Drugs Aging 20 (6): kinsonism with L-dopa and amantadine. Lancet 2: 720.
465476. Miller C (1998). Glutamate receptor activation: a four step
Kaur S, Starr MS (1997). Differential effects of intrastriatal program. Neuroscience 280 (5): 15471548.
and intranigral injections of glutamate antagonists on Montastruc JL, Fabre N, Rascol O et al. (1994). N-methyl-D-
motor behaviour in the reserpine treated rat. Neuroscience aspartate antagonist (NMDA) and Parkinsons disease:
76: 345354. a pilot study with dextromethorphan. Mov Disord 9: 242243.
Kemp JA, McKernan RM (2002). NMDA receptor pathways Nash JE, Brotchie JM (2002). Characterisation of striatal
as drug targets. Nat Neurosci 5: 10391042. NMDA receptors involved in the generation of parkinso-
Kolckgether T, Wullner U, Steinbach JP et al. (1996). Effects nian symptoms: intrastriatal microinjection studies in the
of the antiparkinsonian drug central neurotransmitter sys- 6-OHDA-lesioned rat. Mov Disord 17 (3): 455466.
tems. Eur J Pharmacol 301: 6773. Olney JW (1969). Glutamate induced retinal degeneration in
Kornhuber J, Weller M (1996). New therapeutic possibilities the neonatal mice: electron microscopy if the acutely evol-
with low affinity MNDA receptors antagonists. Nerve- ving lesion. J Neuropathol Exp Neurol 28: 455474.
narzt 67 (1): 7782. Olney JW, Ho OL (1970). Brain damage in infant mice fol-
Kornhuber J, Bormann J, Hubers M et al. (1991). Effects of lowing oral intake of glutamate, aspartate, or cysteine.
1-amino-adamantanes at the MK-801 binding site of the Nature 227: 609611.
NMDA-receptor-gated ion channel: a human postmortem Parkes JD, Baxter RC, Marsden CD et al. (1974). Compara-
brain study. Eur J Pharmacol 206: 297300. tive trial of benzhexol, amantadine, and levodopa in
136 M. G. CERSOSIMO AND F. E. MICHELI

the treatment of Parkinsons disease. J Neurol Neurosurg Snow BJ, Macdonald L, Mcauley D et al. (2000). The effect
Psychiatry 37: 422426. of amantadine in levodopa induced dyskinesias in Parkin-
Parkinson Study Group (2000). A multicenter controlled trial sons disease: a double blind, placebo controlled study.
of remacemide hydrochloride as monotherapy for PD. Clin Neuropharmacol 23 (2): 8285.
Neurology 54 (8): 15831588. Sobolevski AI, Koshelev SG, Khodorov BI (1998). Interac-
Parkinson Study Group (2001). Evaluation of dyskinesias in a tion of memantine and amantadine with agonist inbound
pilot, randomized, placebo controlled trial of remacemide in NMDA-receptor channels in acutely isolated rat hippo-
advanced Parkinsons disease. Arch Neurol 58: 16601668. campal neurons. J Physiol 512 (1): 4760.
Pfeiffer RF (1996). Amantadine-induced vocal myoclonus. Spieker S, Loschmann PA, Klockgether T (1999). The
Mov Disord 11 (1): 104106. NMDA antagonist budipine can alleviate levodopa
Przuntek H, Bittkau S, Bliesath H et al. (2002). Budipine induced motor fluctuations. Mov Disord 14 (3): 517519.
provides additional benefit in patients with Parkinsons Stoof JC, Booij J, Drukarch B (1992). Amantadine as N-
disease receiving a stable optimum dopaminergic drug methyl-D-aspartic acid receptor antagonist: new possibili-
regimen. Arch Neurol 59: 803806. ties for therapeutic applications. Clin Neurol Neurosurg
Reisberg B, Doody R, Stoffler A et al. (2003). Memantine 94: S4S6.
Study Group. Memantine in moderate to severe Alzhei- Uitti RJ, Rajput AH, Ahlskog JE et al. (1996). Amantadine
mers disease. N Engl J Med 348 (14): 13331341. treatment is an independent predictor of improved survival
Savery F (1977). Amantadine and a fixed combination of in Parkinsons disease. Neurology 46 (6): 15511552.
levodopa and carbidopa in the treatment of Parkinsons Vale JA, Maclean KS (1977). Amantadine-induced heart
disease. Dis Nerv Syst 38: 605608. failure. Lancet 1 (8010): 548.
Schachter SC, Tarsy D (2000). Remacemide: current status Verhagen Metman L, Del Dotto P, Van den Munckhof
and clinical applications. Expert Opin Investig Drugs P et al. (1998a). Amantadine as treatment for dyskinesias
9 (4): 871873. and motor fluctuations in Parkinsons disease. Neurology
Schadel M, Wu D, Otton SV et al. (1995). Pharmacokinetics 50: 13231326.
of dextromethorphan and metabolites in humans: influ- Verhagen Metman L, Blanchet PJ, Van den Munckhof
ence of CYP2D6 phenotype and quinidine inhibition. P et al. (1998b). A trial of dextromethorphan in parkinso-
J Clin Psychopharmacol 15: 263269. nian patients with motor response complications. Mov
Scholz EP, Kiesecker C, Lueck S et al. (2003). Drug binding Disord 13 (3): 414417.
to aromatic residues in the HERG channel pore cavity as Von Voigtlander PF, Moore KE (1971). Dopamine release
possible explanation for acquired Long QT syndrome by from the brain in vivo by amantadine. Science 174:
antiparkinsonian drug budipine. Naunyn Schmiedebergs 408410.
Arch Pharmacol 368: 404414. Walker JE, Albers JW, Tourtellotte WW et al. (1972). A
Schmidt WJ, Bubser M (1989). Anticataleptic effects of the qualitative and quantitative evaluation of amantadine in
N-methyl-D-aspartate antagonist MK-801 in rats. Pharma- the treatment of Parkinsons disease. J Chronic Dis 25:
col Biochem Behav 32: 621623. 149182.
Schneider E, Fischer PA, Clemens R et al. (1984). Effects of Wong BY, Coulter DA, Choi DW et al. (1988). Dextrorphan
oral memantine administration on Parkinson symptoms. and dextromethorpham common antitussives, are antiepi-
Results of a placebo controlled multicenter study. Dtsch leptic and antagonize N-methyl-D-asparatate in brain
Med Wochenschr 109: 987990. slices. Neurosci Lett 85: 261266.
Schwab RS, England AC, Poskanzer DC et al. (1969). Aman- Woodworth JR, Dennis SRK, Moore L et al. (1987). The
tadine in the treatment of Parkinsons disease. JAMA 208: polymorphic metabolism of dextromethorphan. J Clin
11681170. Pharmacol 27: 139143.
Shannon KM, Goetz CG, Carroll VS et al. (1987). Amanta- Zeevalk GD, Nicklas WJ (1992). Evidence that the
dine and motor fluctuations in chronic Parkinsons dis- loss of the voltage dependent Mg block of the N-
ease. Clin Neuropharmacol 10: 522526. methyl-D-aspartate receptor underlies activation during
Shoulson I, Penney J, McDermott M et al. (2001). Parkinson inhibition of neuronal metabolism. J Neurochem 59:
Study Group. A randomized, controlled trial of remace- 12111220.
mide for motor fluctuations in Parkinsons disease. Neu- Zeldowicz LR, Huberman J (1973). Long term therapy of
rology 56: 455462. Parkinsons disease with amantadine alone and combined
Shulman LM, Minagar A, Sharma K et al. (1999). Amanta- with levodopa. Can Med Assoc J 109: 588593.
dine-induced peripheral neuropathy. Neurology 53 (8):
18621865.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 37

Investigational drugs

CARLO COLOSIMO* AND GIOVANNI FABBRINI

Dipartimento di Scienze Neurologiche, Universita La Sapienza, Rome, Italy

37.1. Introduction with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine


(MPTP) induced less dyskinesia than LD (Bedard
The introduction of the dopamine precursor levodopa et al., 1992; Pearce et al., 1998), several clinical stu-
(LD) as the mainstay of treatment of Parkinsons dis- dies have been conducted, all showing that dopamine
ease (PD) has provided excellent symptomatic benefit agonists can alleviate parkinsonian symptoms in pre-
for the majority of patients. However, the effects of viously untreated patients with a much reduced inci-
dopaminergic therapy are limited by the fact that LD dence of fluctuations and dyskinesia (Rascol et al.,
has not been shown to slow the progression of the dis- 2000; Parkinson Study Group, 2004b).
ease and that PD patients still face major shortcomings Unfortunately, this strategy has its limitations since
in the chronic phase of the disease. only approximately 30% of parkinsonian patients show
Fluctuations and levodopa-induced dyskinesia (LID) a good and durable response to dopamine agonists as
are the major complications in the current therapeutic monotherapy. Chronic use of dopamine agonists is also
approach to the treatment of PD, deeply affecting patients associated with frequent psychiatric side-effects,
quality of life. More than 50% of all patients treated with including visual hallucinations, delusions and paranoid
LD for 5 years or more develop motor response fluctua- psychosis. Furthermore, dopamine agonists more easily
tions, which are usually associated with the appearance elicit dyskinesia when administered together with LD
of LID (Nutt and Holford, 1996; Colosimo and De or following priming with LD. Therefore, in the fore-
Michele, 1999; Brotchie, 2000). The exact pathophysiol- seeable future, dopamine replacement in the form of
ogy of fluctuations and LID is not fully understood, but LD is likely to remain the mainstay of therapeutic
they are probably related to striatal dopamine receptor approaches for PD. Attempts to reduce dyskinesia by
changes following dopaminergic denervation and chronic means of drug holidays, controlled-release LD prepara-
exposure to LD (Crossman, 1990). These receptor altera- tions, long-acting dopamine agonists (cabergoline), or
tions include changes of sensitivity, relative balance other adjunct therapies such as monoamine oxidase-B
between different dopamine receptor subtypes and differ- (MAO-B) or catechol-O-methyl-transferase inhibitors,
ent translational and neuromodulatory-system responses have generally met with limited success in the clinic
(Crossman, 1990; Colosimo et al., 1996; Brotchie, 2000; (Colosimo and De Michele, 1999). Continuous waking-
Bezard et al., 2001). day subcutaneous apomorphine infusion is effective in
Duration of the disease and early initiation of LD the long term, though not easily applicable to the major-
therapy are key factors for the development of motor ity of patients (Colosimo et al., 1994). As a result, several
complications, though the poor pharmacokinetics of new treatment strategies for PD are currently being
LD (very short half-life, inconsistent absorption) also investigated, particularly those targeting non-dopaminer-
plays an important role. As a result, the dopamine ago- gic pathways.
nists, compounds with a better pharmacokinetic profile Furthermore, in PD there is still no satisfactory
than LD, were developed. Based on experimental data approach to the treatment of cognitive disturbances
showing that de novo administration of dopamine ago- and dementia (Brown and Marsden, 1990), autonomic
nists to non-human primates rendered parkinsonian dysfunction (Senard et al., 2001), balance, walking

*Correspondence to: Carlo Colosimo, Dipartimento di Scienze Neurologiche, Universita La Sapienza, Viale dellUniversita
30, I-00185 Rome, Italy, E-mail: carlo.colosimo@uniroma1.it, Tel: 39-06-4991-4511, Fax: 39-06-4991-4700.
138 C. COLOSIMO AND G. FABBRINI
difficulties and the risk of falling (Bloem et al., 2004), gies will derive directly from studies directed to an
speech disorders (Pinto et al., 2004), psychiatric and understanding of the pathogenesis and mechanisms of
behavioral symptoms (Wint et al., 2004) and sleep cell death. Current information suggests that neurode-
problems (Brotini and Gigli, 2004). Based on all the generation in PD is associated with a cascade of events
previous observations, therapeutic research in the that includes oxidant stress, mitochondrial abnormal-
future is expected to be moving in several different ities, failure in the ubiquitin-proteasome system to clear
directions. Among these are: (1) development of so- unwanted proteins, excitotoxicity, inflammation and
called neuroprotective drugs, capable of blocking or possible other still not identified mechanisms (Dawson
at least slowing down the degenerative process respon- and Dawson, 2003). Considerable evidence suggests that
sible for neuron death, or even of restorative strategies, cell death in PD, regardless of cause, occurs by way of
which would allow to normal brain function to be signal-mediated apoptosis (Hirsch et al., 1999). Devel-
regained; (2) further improvement in the replacement opment of new drugs for neuroprotection is very fast.
of dopaminergic loss; (3) antidyskinetic drugs; and In a recent survey of potential neuroprotective agents
(4) symptomatic drugs acting on neurotransmitters for clinical trials in PD (Ravina et al., 2003), several
other than dopamine, or which may target the brain compounds have been identified as potential effective
in other areas rather than only in the striatum. neuroprotective agents, but only a few are real candi-
dates for phase II or III studies. The development status
37.2. Neuroprotective therapies of these compounds is summarized in Table 37.1.

Interventions that can slow or halt the progression of 37.2.1. Monoamine oxidase inhibitors
PD remain a crucial unmet need.
Several promising approaches are under development 37.2.1.1. Rasagiline
for the potential of neuroprotection in PD. However, the Rasagiline is a propargylamine and is a potent, irreversi-
confounding fact of the possible symptomatic effects of ble, selective inhibitor of MAO-B with no amphetamine-
the drugs tested, the placebo effect, the choice of optimal like metabolites and with the capability of increasing DA
endpoint and the need for validated surrogate markers release. Beside its activity on MAO enzymes, the neu-
are all significant issues when studying a putative neuro- roprotective properties of rasagiline may be linked to
protective intervention in PD, which have not been other factors, such as the capability of inhibiting apop-
completely addressed until now. Neuroprotection strate- tosis at three levels: (1) intranuclear translocation of

Table 37.1
Development status of neuroprotective/neurorestorative agents

Compound Company/institution Class of compound Phase of development

CEP-1347 Cephalon/H Lundbeck Mixed-lineage kinase inhibitor Withdrawn


Kyowa Hakko Kogyo
Leteprinim Spectrum Pharmaceuticals Nerve growth factor agonist Phase II
V-10367 Vertex Pharmaceuticals Neuroimmunophilin ligand
Schering
Liatermine Amgen Growth factor agonist Withdrawn
GPI-1485 Guildford Pharmaceuticals Neuroimmunophilin-ligand Phase II
PAN-408 Panacea Pharmaceuticals a-Synuclein oligomerization Preclinical
inhibitors
PYM-50028 Phytopharm plc/Yamanouchi Dopamine modulator Phase II
Pharmaceutical
TCH-346 Novartis GAPDH inhibitor Withdrawn
MITO-4509 Mitokor Estrogen analog Phase I
Sonic hedgehog Curis/Wyeth Pharmaceuticals Hedgehog agonist Preclinical
protein agonist
ONO-2506 ONO Pharmaceutical Astrocyte modulator Phase II
E-2007 Eisai AMPA antagonist Phase II

Adapted with permission from The Thomson Corporation and Johnston and Brotchie (2004). # 2004 The Thomson Corporation.
GAPDH, glyceraldehyde-3-phosphate dehydrogenase; AMPA, a-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid.
INVESTIGATIONAL DRUGS 139
the glycolytic enzyme glyceraldehyde-3-phosphate 1999). A pilot study using creatine and minocycline is
dehydrogenase; (2) induction of bcl-2; and (3) activa- under way.
tion of mitochondrial permeability transition. Clinical
studies on rasagiline have also confirmed the potential 37.2.3. Other mechanisms
usefulness of this drug in the symptomatic treatment of
37.2.3.1. Estrogens
PD. In early patients (TEMPO study) rasagiline was
superior to placebo in improving Unified Parkinsons Epidemiological data suggest a reduced incidence of
Disease Rating Scale (UPDRS) motor and activities of PD in women (Currie et al., 2004) and there are also
daily living scores. More patients in the placebo group several animal models in which estrogens appear as pro-
(16.7%) than those in rasagiline treatment (11.2%) mising neuroprotective agents. The mechanism may
needed LD within 12 months of the beginning of the involve neurotrophic effects as well as antioxidant
study, although this difference was not significant. This effects (Dluzen and Horstink, 2003).
study also included an initial 6-month placebo phase
(delayed treatment) which showed that patients treated 37.2.3.2. GM1 ganglioside
for 6 months with placebo and then allowed to receive GM1 ganglioside is a component of neuronal mem-
the active drug did not catch up with those patients branes, may facilitate the neurotrophic actions of
who were in active treatment since the beginning of the brain-derived nerve growth factor (BDNF) and glial-
study. These data, although preliminary, suggest a dis- derived neurotrophic factor (GDNF), may inhibit
ease-modifying effect rather than symptomatic benefit apoptosis and may protect against excitotoxicity. Some
only (Parkinson Study Group, 2002, 2004a). preliminary data in PD have shown that the compound
is well tolerated and has short-term symptomatic ben-
37.2.1.2. Zydis selegiline efit (Schneider, 1998). However concerns still exist
about its immunogenicity and the relationship with
The usefulness of conventional selegiline is confounded
GuillainBarre syndrome.
by low bioavailability, extensive first-pass metabolism
and production of amphetamine metabolites. Zydis sele-
37.2.3.3. Neuroimmunophilin
giline (a rapidly disintegrating tablet) dissolves in the
mouth on contact with saliva and undergoes pregastric Neuroimmunophilin ligands (NILs) are drugs derived
absorption, providing high plasma levels of selegiline, from the immunosuppressant FK506 (tacrolimus) that
almost completely avoiding first-pass metabolism and have been shown to have variable efficacy in reversing
markedly reducing the production of amphetamine meta- neuronal degeneration and preventing cell death (Gold
bolites (Seager, 1998). Few studies have shown the and Nutt, 2002). In animal models mimicking PD they
potential usefulness of the drug in the treatment of induce resprouting and are neurotrophic. Some evi-
advanced patients with motor fluctuations, increasing dence suggests that NILs may act through regulation
the time spent in the on phase. Side-effects more com- of steroid hormone receptors. Other evidence suggests
monly reported were dizziness, dyskinesia, hallucina- that NILs may protect neurons by upregulating the
tions, headache and dyspepsia, usually in the first antioxidant glutathione and stimulating nerve regrowth
6 weeks of treatment (Waters et al., 2004). The possible by inducing the production of neurotrophic factors.
role of zydis selegiline as a neuroprotective agent has not Initial clinical trials have had mixed success. In one,
been tested. patients with moderately severe PD showed no overall
improvement in fine motor skills following 6 months
of treatment with the neuroimmunophilin GPI 1485,
37.2.2. Modulators of mitochondrial function although there was a decreased loss of dopaminergic
nerve terminals or eventually an increase in dopami-
Coenzyme Q1 is a health supplement which is able to nergic terminals within 6 months of the higher dose
increase the activity of mitochondrial complex I activity of GPI 1485 drug treatment (Poulter et al., 2004).
and may act as an antioxidant. Preliminary evidence
suggests that doses of 1200 mg/day, which are well toler-
ated, may slow functional decline as measured by 37.2.3.4. Inhibitors of microglial inflammation
UPDRS scores (Shults et al., 2002). (antiapoptotic kinase inhibitors)
Creatine is also a nutritional supplement which is The c-Jun NH2-terminal kinase (JNK) signaling path-
converted to phosphocreatine, a metabolite functioning way is frequently induced by cellular stress and corre-
as an energy buffer able to transfer a phosphoryl group lated with neuronal death. JNK signaling is therefore a
to adenosine diphosphate. Creatine has been shown to promising target in PD for developing pharmacologi-
be protective in MPTP rodent models (Matthews et al., cal intervention. CEP-1347 blocks the activation of
140 C. COLOSIMO AND G. FABBRINI
the c-Jun/JNK apoptotic pathway in neurons exposed to Another approach is currently under investigation
various stressors and attenuates neurodegeneration in through the intraduodenal infusion delivery of LD.
animal models of PD, eventually associated with micro- Advanced parkinsonian patients may benefit from this
glial activation. CEP-1347 reduced cytokine production form of treatment in order to achieve more stable
in primary cultures of human and murine microglia and plasma LD levels (Nyholm et al., 2005).
in monocyte/macrophage-derived cell lines, stimulated
with various endotoxins or the plaque-forming peptide
37.3.2. Increase in the synaptic availability
Abeta140. Moreover, CEP-1347 inhibited brain tumor
of dopamine
necrosis factor (TNF) production induced by intracereb-
roventricular injection of lipopolysaccharide in mice.
Dopamine reuptake blockers, by enhancing and stabi-
As expected from a mixed linear inhibitor (MLK),
lizing intrasynaptic transmitter levels, could help palli-
CEP-1347 acted upstream of p38 and c-Jun activation
ate motor dysfunction in PD, in both early and
in microglia by dampening the activity of both pathways
advanced disease. In experimental animals with severe
(Lund et al., 2005). These data infer that MLKs may be
neurotoxin-induced dopaminergic neuron loss mimick-
important, yet unrecognized, modulators of microglial
ing conditions in advanced PD, LD treatment after an
inflammation and demonstrate a novel anti-inflamma-
effective pharmacologic dopamine transporter (DAT)
tory potential of CEP-1347 (Johnston and Brotchie,
blockade produces far higher elevations in extracellu-
2004). The safety and tolerability of CEP-1347 have
lar DA than normally occur (Pearce et al., 2002). Thus,
recently been studied in 30 patients with PD. Overall
DAT inhibitors should act clinically to potentiate the
the drug was well tolerated and had no acute effect
antiparkinsonian action of LD. Moreover, because a
on parkinsonian symptoms or LD pharmacokinetics
reduction in DA reuptake prolongs its striatal half-life,
(Parkinson Study Group, 2004c).
motor fluctuations as well as dyskinesia and other
adverse consequences of the intermittent stimulation
37.2.3.5. Minocycline
of striatal DA receptors might diminish in the long
Minocycline is a semisynthetic, second-generation tet- term (Nutt and Holford, 1996). A randomized, dou-
racycline derivative which crosses the bloodbrain ble-blind, placebo-controlled study compared the acute
barrier and may inhibit microglial-related inflamma- effects of the monoamine uptake inhibitor NS 2330
tory events and also the apoptotic cascade. Several stu- with those of placebo in 9 fluctuating parkinsonian
dies have shown that minocycline is neuroprotective in patients (Bara-Jimenez et al., 2004). Individuals ran-
animal models of central nervous system trauma and domly assigned to NS 2330 treatment were given an
neurodegenerative diseases; in particular, this com- initial 1-week placebo run-in, followed by a treatment
pound has greatly enhanced survival in nigrostriatal phase consisting of eight doses of 1.5 mg each, admi-
dopaminergic neurons in the MPTP model of PD (Du nistered three times weekly. The cumulative total dose
et al., 2001). Minocycline has, therefore, been incorpo- of 12 mg was selected to achieve plasma drug concen-
rated into an ongoing clinical investigation in trations in therapeutic range, based on preliminary
untreated PD patients (Ravina et al., 2003). pharmacokinetic data obtained by the drug manufac-
turer. Remaining patients received placebo throughout
37.3. Improvement of dopaminergic drugs the entire study. At the dose administered, no change
in parkinsonian scores was found when NS 2330 was
Several strategies have been proposed and developed
given alone or with LD. Moreover, NS 2330 coadmi-
in order to improve the pharmacokinetics and pharma- nistration did not appear to reduce the severity of dys-
codynamics of dopaminergic agents used in PD.
kinesia or the duration of the antiparkinsonian
The development status of all these compounds is
response to LD. Since, under the conditions of this
summarized in Table 37.2.
study, results failed to support the usefulness of dopa-
mine reuptake inhibition in the treatment of advanced
37.3.1. Improvement of levodopa bioavailability
PD, this compound was withdrawn from clinical trials.
Absorption of LD is very sensitive and depends on the
state of the stomach and gastrointestinal system. 37.3.3. Dopamine agonists
Therefore efforts are under way to improve LD
absorption. Recent attempts have shown that methyl- There is still work to be done to understand better the
ester LD (melevodopa) is effective as standard LD effects of selective D1 dopamine agonists in PD
and may have the additional advantage of a faster (Rascol et al., 1999, 2001a). It is realistic to hope for
absorption (Johnston and Brotchie, 2004). further innovations through the development of new
INVESTIGATIONAL DRUGS 141
Table 37.2
Development status of symptomatic antiparkinsonian agents

Phase of
Compound Company/Institution Class of compound development Other potential actions

Dopaminergic
Melevodopa Chiesi Farmaceutici Methyl-l-dopa/ Launched
carbidopa
SR-57667 Sanofi-Synthelabo Irreversible MAO-B Phase IIb Neuroprotective
inhibitor?
Safinamide Newron MAO-B inhibitor Phase III Neuroprotective
Pharmaceuticals Na/Ca channel
blocker
Rasagiline Teva Pharmaceutical Irreversible MAO-B Launched Neuroprotective
Industries/H inhibitor
Lundbeck/Eisai
NS-2330 NeuroSearch/ Monoamine Withdrawn Cognitive enhancement
Boehringer Ingelheim reuptake inhibitor Antidepressant
SPD-473 Shire Pharmaceutical Monoamine reuptake Phase II Cognitive enhancement
Group inhibitor Antidepressant
Sumanirole Pfizer D2-agonist Withdrawn
Rotigotine Schwarz Pharma/Otsuka D2-agonist Launched
Pharmaceutical
SLV-308 Solvay D2 partial agonist/ Phase II Antidepressant
5-HT1A agonist
Talipexole Boehringer Ingelheim D2-agonist/a2- Launched
adrenoceptor agonist
DU-127090 Solvay/H Lundbeck/Wyeth D2 partial agonist/ Phase III
Pharmaceuticals 5-HT1A agonist
BIA-3202 BIAL Group COMT inhibitor Phase I Neuroprotective
Non-dopaminergic
Istradefylline Kyowa Hakko Kogyo A2A antagonist Phase III Neuroprotective
KF-17837 Kyowa Hakko Kogyo A2A antagonist Preclinical Neuroprotective
VR-2006 Vernalis Group A2A antagonist Phase I Neuroprotective
Donepezil Eisai/Pfizer/Wyeth-Ayerst AChE inhibitor Launched Cognitive
International enhancement

Adapted with permission from The Thomson Corporation and Johnston and Brotchie (2004). # 2004 The Thomson Corporation.
MAO-B, monoamine oxidase-B; COMT, catechol-O-methyltransferase; AChE, acetylcholinesterase.

dopamine agonists, fully or partially selective or non- in a silicone-based transdermal system. The rationale
selective (Bezard et al., 2003a). D2-selective agonists behind this transdermal delivery system is based on the
such as sumanirole were developed based on the con- option of having a non-oral DA agonist which may be
cept that the D3 component action of the previous useful in terms of compliance in patients already assum-
generation of dopamine agonists, such as pramipexole, ing many drugs for the oral route and the capability of
may be detrimental to the antiparkinsonian effect stable blood levels of a dopamine agonist. Rotigotine
(Stephenson et al., 2005). patch reaches the steady state in 24 hours and allows
The concept of constant dopaminergic stimulation stable drug release in the 24-hour period (Metman et al.,
(Chase, 1998) is now leading to the study of new ways 2001). Early PD patients (n 242) were treated in a phase
of administering dopaminomimetics, such as the trans- III double-blind, placebo-controlled study against four dif-
dermal route, for the possibility of giving particularly ferent doses of rotigotine (4.5, 9.0, 13.5 or 18 mg). The
stable plasma levels (Parkinson Study Group, 2003). doses of 13.5 and 18.0 mg were superior to placebo at
Rotigotine is a non-ergot dopamine agonist, acting week 11. Side-effects were also more frequent in the
selectively on D2-receptors, which has been formulated active-treatment groups and were qualitatively similar to
142 C. COLOSIMO AND G. FABBRINI
those reported in other studies of dopamine agonists in to counteract LID was raised by the previously shown
early PD patients (nausea, application-site reaction, dizzi- efficacy of such compounds in the treatment of other
ness, somnolence, insomnia, vomiting and fatigue) (Par- types of dyskinesia.
kinson Study Group, 2003). Local skin reactions were
not uncommon (39%), leading to early withdrawal in a 37.4.1. Glutamatergic drugs
few cases. Preliminary positive data are also available
for the transdermal delivery of lisuride (Woitalla et al., Amantadine is an antiviral compound, which has been
2004). widely used as a first-line antiparkinsonian drug since
1969, but has only recently been found to act as a non-
37.3.3.1. Slv-308 competitive N-methyl-D-aspartate (NMDA) glutama-
tergic antagonist (Chase et al., 2000). Findings from
SLV-308 is a partial D2D3 agonist and an agonist of 5-
basic science suggesting that NMDA receptor block-
HT1A receptors, a profile which suggests a good anti-
ade may improve the dyskinetic complications of LD
parkinsonian action with reduced capability of indu-
therapy have thus prompted the use of this compound
cing dyskinesia. Phase II trials in the treatment of PD
as a specific antidyskinetic drug (Metman et al.,
are under way (Wolf, 2003).
1998a). Following pilot studies showing that amanta-
dine has a favorable effect on LID in a significant per-
37.3.3.2. Safinamide centage of patients with PD, further work has
This is a novel experimental drug combining several confirmed the beneficial effects of amantadine on
pharmacological properties that are potentially useful motor response complications (Pourcher et al., 1989;
in the treatment of PD. This drug has the capability of Metman et al., 1999; Luginger et al., 2000; Snow
blocking voltage-dependent sodium and N-type cal- et al., 2000; Del Dotto et al., 2001), although some
cium channel, therefore inhibiting glutamate release. authors have shown that benefit may be short-lived
Safinamide is also a highly selective and reversible (Thomas et al., 2004). The hope of better-tolerated
inhibitor of MAO-B, decreasing dopamine break- glutamate antagonists is driving the development of the
down and the production of toxic free radicals non-competitive a-amino-3-hydroxy-5-methylisoazole-
(Fariello et al., 1998). The peculiarity of this dual 4-propionate (AMPA) antagonists, for example
mechanism of action offers the possibility that safina- talampanel. Since AMPA antagonists may also have
mide might be used in PD, but also in the treatment of neuroprotective actions, some of the companies now
epilepsy. A recent double-blind trial has shown that appear to be developing AMPA antagonists for this
safinamide increased in respect to placebo (37.5 use, rather than as antidyskinetic drugs (Johnston and
versus 21.4%) the percentage of early PD patients Brotchie, 2004).
improving their motor score by >30% and also The results of other trials in which familiar drugs
improved the motor scores in a subgroup of patients have been used in LID are less convincing. For
under stable treatment with dopamine agonists instance, the efficacy of a combination of LD and
(Stocchi et al., 2004). anticholinergic drugs in parkinsonian patients to
reduce both parkinsonian symptoms and biphasic
37.4. Antidyskinetic drugs dyskinesia was only demonstrated in a single trial
(Pourcher et al., 1989). The addition of 5-methoxy
The pathophysiology underlying treatment-related 5-N, N-dimethyl-tryptamine, a non-selective serotonin
dyskinesia is not well known and cannot easily be agonist, slightly reduced dyskinesia but also reduced
fitted into current models of basal ganglia dysfunction the antiparkinsonian benefit of LD (Gomez-Mancilla
in PD. It may involve altered activity in the projection and Bedard, 1993). More recently, alleviation of par-
pathways from the striatum to the globus pallidus, kinsonian symptoms without the development of dys-
resulting in reduced activity in the output regions of kinesia has been reported in both MPTP-lesioned
the basal ganglia, i.e. the internal segment of the glo- monkeys and other experimental animals by using ade-
bus pallidus and the substantia nigra pars reticulata nosine A2A receptor antagonists and NR2B-selective
(Papa et al., 1999). Several experimental and clinical NMDA glutamatergic antagonists (Kanda et al.,
studies in parkinsonism have shown that motor fluc- 1998, 2000; Grondin et al., 1999; Steece-Collier
tuations and LID can be modulated by drugs acting et al., 2000; Bibbiani et al., 2002; Lundblad et al.,
on neurotransmitters other than dopamine, including 2003). The potential symptomatic use in PD of drugs
glutamate, g-aminobutyric acid, norepinephrine, acet- acting on serotonergic and adenosine receptors will
ylcholine, serotonin, adenosine and cholecystokinin. be discussed in more detail in the next part of this
In numerous cases, the prospect of using specific drugs chapter.
INVESTIGATIONAL DRUGS 143
A summary of the various drugs which have been of searches, using the search terms Parkinsons dis-
successfully tested in LID in controlled trials on ease, levodopa and dyskinesia on Medline and
humans is shown in Table 37.3 (Colosimo and Craus, references from relevant articles; numerous articles
2003). Data for this synopsis were identified by means were also identified through searches in the authors

Table 37.3
Symptomatic drugs for LID as proven by controlled trials

Compound Mechanism of action Reference and study design Dose*

Ritanserin Selective serotonergic 5-HT2 Meco et al. (1988): Single-blind, Mean dosage of
antagonist placebo-controlled, cross-over study 21.4 mg (range
1030 mg), 3 a day
Buspirone Serotonergic 5-HT1A-agonist Bonifati et al. (1994): Double-blind, 10 mg, 2 a day
placebo-controlled, cross-over study
Fluoxetine Selective serotonin reuptake Durif et al. (1995): Videotape 20 mg, 2 a day
inhibitor randomized evaluation
Propranolol b-Adrenergic blocker Carpentier et al. (1996): Videotape Up to 60 mg a day
randomized evaluation
Clozapine Dopaminergic D4/D1- Durif et al. (1997): Videotape 50 mg/day
antagonist, serotonergic 5-HT2 randomized evaluation
antagonist, anticholinergic? Durif et al. (2004): Double-blind, Up to 50 mg a day
parallel-group, placebo-controlled,
multicenter trial
Amantadine Non-competitive NMDA Metman et al. (1998a): Double-blind, 100 mg, 34 a day
glutamatergic antagonist placebo-controlled, cross-over study
Metman et al. (1999): Double-blind, 100 mg, 34 a day
placebo-controlled, cross-over study
Luginger et al. (2000): Double-blind, 100 mg, 3 a day
placebo-controlled, cross-over trial
Snow et al. (2000): Double-blind, 100 mg, 2 a day
placebo-controlled, cross-over trial
Del Dotto et al. (2001): Acute double- 200 mg in a 2-hour
blind, placebo-controlled study intravenous infusion
Dextromethorphan NMDA glutamatergic antagonist Metman et al. (1998b): Double-blind, Range, 60120 mg/day
placebo-controlled, cross-over study
Metman et al. (1998c): Double-blind, Up to 180 mg a day
placebo-controlled, cross-over study
Olanzapine Dopaminergic D1/D2/D4- Manson et al. (2000a): Randomized, Up to 7. 5 mg/day
antagonist placebo-controlled, double-blind,
cross-over trial
Idazoxan a2-Adrenergic antagonist Rascol et al. (2001b): Randomized, 10, 20, 40 mg, in
double-blind, placebo-controlled single dose
study
Nabilone Cannabinoid receptor agonist Sieradzan et al. (2001): Randomized, 0.03 mg/kg in two
double-blind, placebo-controlled, doses
cross-over trial
Sarizotan Serotonergic 5-HT1A full agonist, Olanow et al. (2004): Prospective, 6- Up to 10 mg/day
dopaminergic D2/D3/D4 weak month multicenter open-label dose-
antagonist? rising study and videotape evaluation
Naloxone Opioid antagonist Fox et al. (2004): Randomized, double- 0.4 mg/kg per min in
blind, placebo-controlled, cross-over, intravenous infusion
acute-challenge study

*Unless indicated, the dose was given orally; when available, the number of doses per day was given.
Adapted with permission from Colosimo and Craus (2003).
NMDA, N-methyl-d-aspartate.
144 C. COLOSIMO AND G. FABBRINI
extensive files. Abstracts and reports from meetings reduces the dyskinetic effect produced by LD, whereas
were also included. Several other drugs have been the antiparkinsonian effect is not altered regardless of
reported to improve LID in case series or uncontrolled the dosage. Yohimbine has a variety of pharmacologi-
trials, e.g. methysergide (Gomez-Mancilla and Bedard, cal properties: it is mainly an a-adrenergic receptor
1993), physostigmine (Tarsy et al., 1974; Gomez-Man- antagonist with low selectivity between a1 and a2 sub-
cilla and Bedard, 1993), estrogens (Gomez-Mancilla types, but also acts as an antagonist at dopamine D2, 5-
and Bedard, 1992), progesterone (Gomez-Mancilla and HT1A and peripheral 5-HT2B receptors and as an ago-
Bedard, 1992), riluzole (Merims et al., 1999), mirtazapine nist at 5-HT1D receptors. On the other hand the combi-
(Meco et al., 2003), quetiapine (Oh et al., 2002), magne- nation of LD with clonidine, an a2-adrenoreceptor
sium sulfate (Chassain et al., 2002), alpha-amino-3- agonist, also reduced LID, whereas at higher doses
hydroxy-5-methyl-4-isoxazole (a propionic acid receptor clonidine blocked both the antiparkinsonian and the
antagonist) (Silverdale et al., 2002), 3,4-methylenedioxy- dyskinetic effects of LD (Nishikawa et al., 1984;
methamphetamine (ecstasy) (Iravani et al., 2003) and Gomez-Mancilla and Bedard, 1993; Hill and Brotchie,
cannabis (Venderova et al., 2004). These pilot studies 1999). More recent are the data concerning fipamezole
should encourage, when appropriate, larger double-blind (JP-1730), a 2-indane imidazole a2-adrenergic receptor
controlled trials. antagonist which was also able to reduce LID in the
MPTP-lesioned marmoset model of PD (Savola
37.4.2. Noradrenergic drugs et al., 2003): this compound is currently in phase III
clinical trials (Peltonen et al., 2002). Besides, in a
A key abnormality most likely underlying LID is the single clinical trial it has been suggested that adminis-
overactivity of the direct striatal output pathway con- tration of low doses of propranolol, a non-selective -
necting the striatum with the output regions of the adrenergic blocker, may improve LID in patients with
basal ganglia. Adrenergic a2-receptors are present in advanced PD (Carpentier et al., 1996).
the basal ganglia and several experimental studies On the basis of these data, idazoxan, a new selec-
have shown that they occur in high density in the rat tive a2-adrenoreceptor antagonist, was developed and
and mouse striatum, in a position likely to influence tested in animals with experimental parkinsonism and
the direct or indirect striatal motor outputs (Scheinin subsequently in patients with LID. Two reports, one
et al., 1994; Holmberg et al., 1999; Papa et al., 1999; experimental and the other clinical, have confirmed
Bezard et al., 2001; Zhang and Ordway, 2003). that idazoxan has an interesting pharmacological pro-
As it has also been suggested that activation of a2- file, improving LID without a return to parkinsonian
adrenergic receptors can facilitate movements pro- symptoms (Fox et al., 2001; Rascol et al., 2001b). Pre-
duced by activation of the direct pathway, it is possible vious experimental data in animals have suggested
that enhanced a2-adrenergic receptor stimulation may that, although idazoxan as a monotherapy displays no
contribute to the pathophysiology of LID (Hill and antiparkinsonian effect, when given in combination
Brotchie, 1999). Other studies have indicated that with LD it not only reduces the dyskinetic side-effects
dopaminergic transmission in the striatum can also be of LD, but may even extend the antiparkinsonian
influenced by -adrenergic activity; the density of - action of this compound (Henry et al., 1999).
adrenergic receptors is one of the highest in the stria- The purpose of the experimental study by Fox and
tum and the local release of dopamine is increased coworkers (2001) was to compare the effect of ida-
by isoproterenol when applied in vivo into the caudate zoxan on dyskinesia produced by both LD and apomor-
nucleus or in vitro on rat striatal slices, an effect that is phine in the MPTP experimental model of PD. The
prevented by propranolol (Reisine et al., 1982). marmosets were treated with MPTP to induce a parkin-
In addition, motor behavior studies in animals on sonian syndrome. In the first part of the study, only LD
dopaminenorepinephrine interactions have shown that (8 mg/kg) or apomorphine (0.0750.3 mg/kg) was
the noradrenergic systems in PD are impaired and this administered in order to establish the severity of the
probably contributes to both motor and affective symp- dyskinesia each drug produced. In the second part of
toms observed in this condition (Burn, 2002). the study, doses of apomorphine and LD were individu-
These observations have encouraged studies on ally administered along with either idazoxan (2.5 mg/kg)
drugs acting on the noradrenergic system. Ever since or vehicle. A neurologist with clinical experience in
the 1980s, the coadministration of yohimbine and LD movement disorders who was blinded to the animals
to MPTP-treated primates has been known to reduce treatment then analyzed the videotapes.
LID without influencing the antiparkinsonian efficacy Part one of the study demonstrated that both apo-
of this compound (Chopin et al., 1986). Yohimbine morphine and LD increased mobility and improved
INVESTIGATIONAL DRUGS 145
bradykinesia and posture scores, when compared with AUC in the 10- and 20-mg idazoxan treatment groups
vehicle-treated marmosets. Administration with either (20 and 40%) as compared to the placebo group. No
apomorphine or LD resulted in a dose-dependent AUC trend was found in the 40-mg idazoxan treatment
increase in dyskinesia, with dyskinesia present for group. The severity score distribution indicated a
the entire period of maximal antiparkinsonian action. statistically significant treatment effect in favor of
When idazoxan was coadministered with LD, the idazoxan (20 mg) when compared with placebo
median peak-dose dyskinesia score was significantly (P 0.01). No significant treatment effects were
lower than that observed with LD alone (4 versus 16; found in the biphasic and dyskinesia distribution
P < 0.05), whereas peak posture, bradykinesia or analysis. The antiparkinsonian effects of LD were
mobility scores were not significantly different. There similar in both the idazoxan and the placebo groups,
were no significant differences between LD/idazoxan whereas adverse events were more frequent with
versus LD alone in the duration of dyskinesia, nor idazoxan than with placebo. The results of this study
were there any significant differences between the seem to indicate that idazoxan, by blocking a2-receptors,
coadministration of idazoxan and apomorphine and may reduce levels of dyskinesia in PD patients
apomorphine alone in the median peak-dose dyskine- without affecting the antiparkinsonian efficacy of LD
sia score. Idazoxan had no effect on the antiparkinso- treatment.
nian action of apomorphine or in the duration of Although the data obtained in these two studies are
dyskinesia induced by this compound. By contrast, consistent with each other, they are in contrast with the
the action of LD, following the coadministration of only other available clinical trial, which recently
idazoxan and LD, lasted longer than when LD was reported that idazoxan was not beneficial for LID in
administered alone (250  20 minutes on time versus 8 PD patients (Manson et al., 2000b). Moreover,
130  10 minutes on time; P < 0.05). The authors although not published, a subsequent multi-institu-
concluded that the dyskinesia resulting from LD tional trial investigating idazoxan as a treatment for
involves adrenoreceptor activation, whereas dyskinesia LID was negative. The reasons for the negative results
resulting from apomorphine does not. Indeed, the were complex and at least partly due to the capacity of
action of a2-adrenergic antagonists may involve the trial centers to comply with the protocol and to the
blockade of the action of norepinephrine synthesized safety profile of the drug (early drop-outs), maybe
from LD; the hypothesis is that, since dopamine ago- due to inadequate titration (O. Rascol, personal
nists are not metabolized to norepinephrine, idazoxan communication).
does not reduce dyskinesia produced by such agents. These studies with idazoxan are of theoretical and
This should be taken into consideration when trials clinical interest, but they do have several drawbacks:
with new antidyskinetic agents based on acute the results of the clinical trial, in particular, are based
challenges with apomorphine are planned. on a relatively small number of PD patients and only
In a clinical study, Rascol and coworkers (2001b) following an acute oral challenge of LD. Moreover,
assessed the effects of idazoxan on LID in patients since idazoxan may induce several adverse events,
with advanced PD. A total of 18 patients were enrolled such as hypertension, tachycardia, flushing and head-
in this single-oral-dose randomized double-blind pla- ache, it is difficult to ascertain what are the usefulness
cebo-controlled study. The study tested three different and benefit-to-risk ratio of idazoxan in the long-term
idazoxan doses (10, 20 and 40 mg) and placebo. management of dyskinetic parkinsonian patients.
Assessment consisted of a single oral dose of either In summary, it seems that, though the mechanisms
idazoxan or placebo, followed an hour later by the underlying the manifestations and the priming process
patients usual first morning dose of LD plus an addi- for dyskinesia have yet to be fully elucidated, non-
tional 50 mg. A trained physician used the UPDRS to dopaminergic compounds may provide an effective
monitor the treatment effect. Patients were videotaped way of limiting the expression of involuntary move-
for the assessment of dyskinesia and were identified as ments in PD.
either peak-dose or biphasic. Data were analyzed by
means of three different measurements: (1) calculation 37.5. Symptomatic non-dopaminergic drugs
of the area under the curve (AUC) of the changes from
baseline in the dyskinesia and UPRDS scores; (2) cal- Dopamine replacement therapy effectively treats the
culation of the score distribution; and (3) an analysis early motor symptoms of PD. However, its association
of the peak-dose and biphasic dyskinesia scores. with the development of motor complications limits
The AUC analysis revealed a statistically insignifi- its usefulness in late stages of the disease (Nutt and
cant dose-related reduction for the mean dyskinesia Holford, 1996; Colosimo and De Michele, 1999) and
146 C. COLOSIMO AND G. FABBRINI
a non-dopaminergic approach to therapy might thus prolong its half-life. If 5-HT1A agonists produce the
provide an effective way of preventing the development same pharmacologic effects in PD patients, then a
of these complications in PD. reduction in peak-dose dyskinesias and wearing-off
fluctuations might be expected. Recent observations
37.5.1. Drugs acting at adenosine receptors in parkinsonian animals support this possibility
(Bibbiani et al., 2001).
Adenosine A2A receptors are localized to the indirect To evaluate this hypothesis, the effects of a selective
striatal output function and control motor behavior. 5-HT1A agonist, sarizotan, given orally at 2 and 5 mg
They are active in predictive experimental models of twice daily to 18 relatively advanced parkinsonian
PD and appear to be promising as the first major patients, were recently compared with baseline placebo
non-dopaminergic therapy for PD (Johnston and function during a 3-week, double-blind, placebo-
Brotchie, 2004). The initial results from a controlled controlled, proof-of-concept study (Bara-Jimenez
clinical trial of an adenosine A2A antagonist, theophyl- et al., 2005). Sarizotan alone or with intravenous
line, conducted on 10 patients with PD, have been LD had no effect on parkinsonian severity. But at
contrasting (longer beneficial response on akinesia safe and tolerable doses, sarizotan coadministration
without significant changes in the other clinical reduced LID and prolonged its antiparkinsonian
parameters of the disease) (Kulisevsky et al., 2002). response (P < 0.05). The findings suggest that 5-
Istradefylline (KW-6002) is a novel adenosine A2A HT1A receptor stimulation in LD-treated parkinsonian
receptor antagonist designed to treat patients with patients can modulate striatal dopaminergic function
motor fluctuations and dyskinesias (Jenner, 2005). and that 5-HT1A agonists may be useful as LD adju-
Istradefylline is currently in phase III clinical trials vants in the treatment of advanced PD. In another
for efficacy in patients with PD; results from phase II open-label trial on 64 patients with advanced PD,
clinical trials demonstrated that it provides a signifi- sarizotan treatment induced a significant reduction
cant reduction in off time and increased on time in dyskinesia and particularly in troublesome dyski-
with non-troublesome dyskinesia in LD-treated nesia (Olanow et al., 2004). These benefits were
patients with established motor complications and is obtained without change in total off time or change
safe and well tolerated. In a 12-week, double-blind, in UPDRS or activities of daily living scores. Unfor-
randomized, placebo-controlled, exploratory study tunately, this compound was recently withdrawn from
(Hauser et al., 2003), patients with motor fluctuations clinical trials.
and peak-dose dyskinesias were randomly assigned to
placebo (n 29), istradefylline (maximum of 20 mg/ 37.5.3. Drugs acting at synaptic vesicular proteins
day: n 26), or istradefylline (maximum 40 mg/day:
n 28). As assessed by patient home diary, off time Levetiracetam (LEV; (S)-alpha-ethyl-2-oxo-1-pyrro-
was reduced and severity of dyskinesia was unchanged, lidine acetamide) is an antiepileptic drug that is
whereas the on time with dyskinesia increased. approved as add-on therapy in the treatment of par-
In addition, there is increasing preclinical evidence that tial-onset seizures (Kumar and Smith, 2004). LEV
A2A receptor antagonists may also have neuroprotective has been shown to reduce LID in preclinical studies
properties (Johnston and Brotchie, 2004). Thus, their of animal models of PD (Bezard et al., 2003c).
eventual use as symptomatic therapy may lead to the Furthermore, a few case reports and small open-label
identification of disease-modifying properties. studies indicate that LEV may reduce various abnor-
mal involuntary movements, including myoclonus,
37.5.2. Serotonergic drugs paroxysmal kinesiogenic choreoathetosis and
Meiges syndrome. The tolerability and preliminary
With progressive degeneration of dopaminergic neu- efficacy of LEV in reducing LID in PD patients were
rons in PD, dopamine formation from exogenous LD evaluated in a prospective open-label pilot study
increasingly takes place in striatal serotonergic nerve (Zesiewicz et al., 2005). Nine PD patients who were
terminals (Bezard et al., 2003b). It is thus not surpris- experiencing peak-dose dyskinesias for at least 25%
ing that pharmacologic agents affecting serotonergic of the awake day and were at least moderately dis-
nerve impulse activity, by interacting with 5-HT1A abled were treated with LEV in doses up to 3000
autoreceptors, can regulate serotonin release under mg for up to 60 days. The primary outcome measure
normal conditions and dopamine release in LD-treated was the percentage of the awake day that patients
parkinsonian animals. In the latter circumstances, spent on without dyskinesia or with non-troublesome
drugs that stimulate these autoreceptors tend to attenu- dyskinesia (good on time). The mean dose of LEV at
ate peak striatal concentrations of dopamine and endpoint was 625  277 mg/day. LEV significantly
INVESTIGATIONAL DRUGS 147
improved the percentage of the awake day on without
Bezard E, Brotchie JM, Gross CE (2001). Pathophysiology
dyskinesia or with non-troublesome dyskinesia at of levodopa-induced dyskinesia: potential for new thera-
endpoint compared to baseline (43  12% versus 61 pies. Nat Rev Neurosci 2: 577588.
 17%; P 0.02). Percentage on time with trouble- Bezard E, Ferry S, Mach U et al. (2003a). Attenuation of
some dyskinesia decreased from 23  10% at base- levodopa-induced dyskinesia by normalizing dopamine
line to 11  6% at endpoint, although not D3 receptor function. Nat Med 9: 762767.
significantly. There was no significant increase in Bezard E, Gross CE, Brotchie JM (2003b). Presymptomatic
off time from baseline to endpoint. There was a compensation in Parkinsons disease is not dopamine-
56% drop-out rate, in most of the cases due to somno- mediated. Trends Neurosci 26: 215221.
lence. These preliminary data suggest that LEV is a Bezard E, McGuire SG, Crossman AR et al. (2003c). Novel
antiepileptic drug levetiracetam decreases dyskinesia
promising drug in PD patients who experience severe
elicited by L-dopa and ropinirole in the MPTP-lesioned
peak-dose dyskinesia; as a result, LEV is currently in
marmoset. Mov Disord 18: 13011305.
phase III clinical trials. Bibbiani F, Oh JD, Chase TN (2001). Serotonin 5-HT1A ago-
nist improves motor complications in rodent and primate
37.6. Conclusions parkinsonian models. Neurology 57: 18291834.
Bibbiani F, Oh JD, Petzer JP et al. (2002). A2A receptor
Future developments in drug therapy of PD appear to antagonist prevents the development of dopamine agonist-
be exciting and it is likely that PD will remain one induced motor complications in primate and rodent models
of the most active areas of drug development in neu- of Parkinsons disease. Mov Disord 17 (Suppl 5): S78S79.
rology for many years to come. In the near future it Bloem BR, Hausdorff JM, Visser JE et al. (2004). Falls and
will probably be feasible to demonstrate in vivo if freezing of gait in Parkinsons disease: a review of two
interconnected, episodic phenomena. Mov Disord 19:
drugs act as neuroprotective or even restorative. It is
871884.
also very likely that there will be improvements in
Bonifati V, Fabrizio E, Cipriani R et al. (1994). Buspirone in
the way physicians may be able to replace dopaminer- levodopa-induced dyskinesias. Clin Neuropharmacol 17:
gic loss and more and more attention is expected on 7382.
new ways of treating non-dopaminergic symptoms. Brotchie JM (2000). The neural mechanisms underlying
Efforts are expected too in a better characterization levodopa-induced dyskinesia in Parkinsons disease. Ann
and treatment of cognitive disturbances which fre- Neurol 47 (4 Suppl 1): S105S114.
quently accompany PD. This chapter is by definition Brotini S, Gigli GL (2004). Epidemiology and clinical fea-
a section which must be intended in evolution. It is tures of sleep disorders in extrapyramidal disease. Sleep
possible that by the time of publication some studies Med 5: 169179.
will have appeared showing the efficacy or the failure Brown RG, Marsden CD (1990). Cognitive function in
Parkinsons disease: from description to theory. Trends
of a certain treatment. We have tried to give an overview
Neurosci 13: 2129.
of the closest prospective in the new pharmacological
Burn DJ (2002). Depression in Parkinsons disease. Eur
strategies in PD. J Neurol 9 (Suppl 3): 4454.
Carpentier AF, Bonnet AM, Vidailhet M et al. (1996).
Acknowledgments Improvement of levodopa-induced dyskinesia by propra-
nolol in Parkinsons disease. Neurology 46: 15481551.
The authors thank W. Poewe and O. Rascol for their Chase TN (1998). The significance of continuous dopami-
data on ongoing trials in Parkinsons disease. nergic stimulation in the treatment of Parkinsons disease.
M. Bologna and C. Aurilia helped with the manuscript Drugs 55 (Suppl 1): 19.
preparation. Chase TN, Oh JD, Konitsiotis S (2000). Antiparkinsonian
and antidyskinetic activity of drugs targeting central
glutamatergic mechanisms. J Neurol 247 (Suppl 2):
References
II36II42.
Chassain C, Eschalier A, Durif F (2002). Magnesium sul-
Bara-Jimenez W, Dimitrova T, Sherzai A et al. (2004). Effect
phate potentiates the antiparkinsonian levodopa effect
of monoamine reuptake inhibitor NS2330 in advanced
and has antidyskinetic effect in MPTP-lesioned monkeys.
Parkinsons disease. Mov Disord 19: 11831186.
Mov Disord 17 (Suppl 5): S22S23.
Bara-Jimenez W, Bibbiani F, Morris MJ et al. (2005). Effects
Chopin P, Pellow S, File SE (1986). The effects of yohimbine
of serotonin 5-HT1A agonist in advanced Parkinsons dis-
on exploratory and locomotor behaviour are attributable to
ease. Mov Disord 20 (8): 932936.
its effects at noradrenaline and not at benzodiazepine recep-
Bedard PJ, Mancilla BG, Blanchet P et al. (1992). Levodopa-
tors. Neuropharmacology 25: 5357.
induced dyskinesia: facts and fancy. What does the MPTP
Colosimo C, Craus A (2003). Noradrenergic drugs for levodopa-
monkey model tell us? Can J Neurol Sci 19: 134137.
induced dyskinesia. Clin Neuropharmacol 26: 299305.
148 C. COLOSIMO AND G. FABBRINI

Colosimo C, De Michele M (1999). Motor fluctuations in Gomez-Mancilla B, Bedard PJ (1993). Effect of nondopami-
Parkinsons disease: pathophysiology and treatment. Eur nergic drugs on L-dopa-induced dyskinesias in MPTP-
J Neurol 6: 121. treated monkeys. Clin Neuropharmacol 16: 418427.
Colosimo C, Merello M, Albanese A (1994). Clinical useful- Grondin R, Bedard PJ, Hadj Tahar A et al. (1999). Anti-
ness of apomorphine in movement disorders. Clin Neuro- parkinsonian effect of a new selective adenosine A2A
pharmacol 17: 243259. receptor antagonist in MPTP-treated monkeys. Neurology
Colosimo C, Merello M, Hughes AJ et al. (1996). Motor 52: 16731677.
response to acute dopaminergic challenge with apomor- Hauser RA, Hubble JP, Truong DD (2003). Istradefylline
phine and levodopa in Parkinsons disease: implications US-001 Study Group. Randomized trial of the adenosine
for the pathogenesis of the on-off phenomenon. J Neurol A (2A) receptor antagonist istradefylline in advanced
Neurosurg Psychiatry 60: 634637. PD. Neurology 61: 297303.
Crossman AR (1990). An hypothesis on the pathophysiological Henry B, Fox SH, Peggs D et al. (1999). The alpha2-adrener-
mechanisms that underlie levodopa- or dopamine agonist- gic receptor antagonist idazoxan reduces dyskinesia and
induced dyskinesia in Parkinsons disease: implications for enhances anti-parkinsonian actions of L-dopa in the
future strategies in treatment. Mov Disord 5: 100108. MPTP-lesioned primate model of Parkinsons disease.
Currie LJ, Harrison MB, Trugman JM et al. (2004). Postmeno- Mov Disord 14: 744753.
pausal estrogens use affects the risk of Parkinson disease. Hill MP, Brotchie JM (1999). The adrenergic receptor ago-
Arch Neurol 61: 886888. nist, clonidine, potentiates the anti-parkinsonian action
Dawson TM, Dawson VL (2003). Molecular pathways of neuro- of the selective kappa-opioid receptor agonist, enadoline,
degeneration in Parkinsons disease. Science 302: 819822. in the monoamine-depleted rat. Br J Pharmacol 128:
Del Dotto P, Pavese N, Gambaccini G et al. (2001). Intrave- 15771585.
nous amantadine improves levadopa-induced dyskinesias: Hirsch EC, Hunot S, Faucheux B et al. (1999). Dopaminergic
an acute double-blind placebo-controlled study. Mov neurons degenerate by apoptosis in Parkinsons disease.
Disord 16: 515520. Mov Disord 14: 383385.
Dluzen D, Horstink M (2003). Estrogens as neuroprotectant
Holmberg M, Scheinin M, Kurose H et al. (1999). Adrenergic
of nigrostriatal dopaminergic system: laboratory and clin-
alpha2C-receptors reside in rat striatal GABAergic projec-
ical studies. Endocrine 21: 6775.
tion neurons: comparison of radioligand binding and
Du Y, Ma Z, Lin S (2001). Minocycline prevents nigrostria-
immunohistochemistry. Neuroscience 93: 13231333.
tal dopaminergic neurodegeneration in the MPTP model
Iravani MM, Jackson MJ, Kuoppamaki M et al. (2003).
of Parkinsons disease. Proc Natl Acad Sci USA 98:
3,4-Methylenedioxymethamphetamine (ecstasy) inhibits
1466914674.
dyskinesia expression and normalizes motor activity in
Durif F, Vidailhet M, Bonnet AM et al. (1995). Levodopa-
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated
induced dyskinesias are improved by fluoxetine. Neurol-
primates. J Neurosci 23: 91079115.
ogy 45: 18551858.
Durif F, Vidailhet M, Assal F et al. (1997). Low-dose cloza- Jenner P (2005). Istradefylline, a novel adenosine A2A recep-
pine improves dyskinesias in Parkinsons disease. tor antagonist, for the treatment of Parkinsons disease.
Neurology 48: 658662. Expert Opin Investig Drugs 14: 729738.
Durif F, Debilly B, Galitzky M et al. (2004). Clozapine Johnston TH, Brotchie JM (2004). Drugs in development for
improves dyskinesias in Parkinson disease: a double-blind, Parkinsons disease. Curr Opin Investig Drugs 5 (7):
placebo-controlled study. Neurology 62: 381388. 720726.
Fariello RG, McArthur RA, Bonsignori A et al. (1998). Pre- Kanda T, Jackson MJ, Smith LA et al. (1998). Adenosine
clinical evaluation of PNU-151774E as a novel anticon- A2A antagonist: a novel antiparkinsonian agent that does
vulsivant. J Pharmacol Exp Ther 285: 397403. not provoke dyskinesia in parkinsonian monkeys.
Fox S, Silverdale M, Kellett M et al. (2004). Non-subtype- Ann Neurol 43: 507513.
selective opioid receptor antagonism in treatment of levo- Kanda T, Jackson MJ, Smith LA et al. (2000). Combined use
dopa-induced motor complications in Parkinsons disease. of the adenosine A(2A) antagonist KW-6002 with
Mov Disord 19: 554560. L-DOPA or with selective D1 or D2 dopamine agonists
Fox SH, Henry B, Hill MP et al. (2001). Neural mechanisms increases antiparkinsonian activity but not dyskinesia in
underlying peak-dose dyskinesia induced by levodopa and MPTP-treated monkeys. Exp Neurol 162: 321327.
apomorphine are distinct: evidence from the effects of the Kulisevsky J, Barbanoj M, Gironell A et al. (2002). A dou-
alpha(2) adrenoceptor antagonist idazoxan. Mov Disord ble-blind crossover, placebo-controlled study of the ade-
16: 642650. nosine A2A antagonist theophylline in Parkinsons
Gold BG, Nutt JG (2002). Neuroimmunophilin ligands in the disease. Clin Neuropharmacol 25: 2531.
treatment of Parkinsons disease. Curr Opin Pharmacol 2: Kumar SP, Smith PE (2004). Levetiracetam as add-on ther-
8286. apy in generalised epilepsies. Seizure 13: 475477.
Gomez-Mancilla B, Bedard PJ (1992). Effect of estrogen and Luginger E, Wenning GK, Bosch S et al. (2000). Beneficial
progesterone on L-dopa induced dyskinesias in MPTP- effects of amantadine on L-dopa-induced dyskinesias in
treated monkeys. Neurosci Lett 135: 129132. Parkinsons disease. Mov Disord 15: 873878.
INVESTIGATIONAL DRUGS 149

Lund S, Porzgen P, Mortensen AL et al. (2005). Inhibition of Papa SM, Desimone R, Fiorani M et al. (1999). Internal
microglial inflammation by the MLK inhibitor CEP-1347. globus pallidus discharge is nearly suppressed during
J Neurochem 92: 14391451. levodopa-induced dyskinesias. Ann Neurol 46: 732738.
Lundblad M, Vaudano E, Cenci MA (2003). Cellular and Parkinson Study Group (2002). A controlled trial of rasagi-
behavioural effects of the adenosine A2a receptor line in early Parkinson disease: the TEMPO Study.
antagonist KW-6002 in a rat model of L-DOPA-induced Arch Neurol 59: 19391943.
dyskinesia. J Neurochem 84: 13981410. Parkinson Study Group (2003). A controlled trial of rotigo-
Manson AJ, Schrag A, Lees AJ (2000a). Low-dose olanza- tine monotherapy in early Parkinsons disease.
pine for levodopa induced dyskinesias. Neurology 55: Arch Neurol 60: 17211728.
795799. Parkinson Study Group (2004a). A controlled, randomized,
Manson AJ, Iakovidou E, Lees AJ (2000b). Idazoxan is inef- delayed-start study of rasagiline in early Parkinson
fective for levodopa-induced dyskinesia in Parkinsons disease. Arch Neurol 61: 561566.
disease. Mov Disord 15: 336337. Parkinson Study Group (2004b). Pramipexole vs levodopa as
Matthews RT, Ferrante RJ, Klivenyi P et al. (1999). Creatine initial treatment for Parkinson disease: a 4-year rando-
and cyclocreatine attenuate MPTP neurotoxicity. Exp mized controlled trial. Arch Neurol 61: 10441053.
Neurol 157: 142149. Parkinson Study Group (2004c). The safety and tolerability
Meco G, Marini S, Lestingi L et al. (1988). Controlled of a mixed lineage kinase inhibitor (CEP-1347) in PD.
single-blind cross-over study of ritanserin and placebo in Neurology 62: 330332.
L-dopa induced dyskinesias in Parkinsons disease. Pearce RK, Banerji T, Jenner P et al. (1998). De novo
Curr Ther Res 43: 262270. administration of ropinirole and bromocriptine induces
Meco G, Fabrizio E, Di Rezze S et al. (2003). Mirtazapine in less dyskinesia than LD in the MPTP-treated marmoset.
L-dopa-induced dyskinesias. Clin Neuropharmacol 26: Mov Disord 13: 234241.
179181. Pearce RK, Smith LA, Jackson MJ et al. (2002). The mono-
Merims D, Ziv I, Djaldetti R et al. (1999). Riluzole for amine reuptake blocker brasofensine reverses akinesia
levodopa-induced dyskinesias in advanced Parkinsons without dyskinesia in MPTP-treated and levodopa-primed
disease. Lancet 353: 17641765. common marmosets. Mov Disord 17: 877886.
Metman LV, Del Dotto P, van de Munckhof P et al. (1998a). Peltonen JM, Huupponen R, Ahokoski O et al. (2002). Tolerabil-
Amantadine as treatment for dyskinesias and motor fluctua- ity and safety of buccal JP-1730. A phase I study in healthy
tions in Parkinsons disease. Neurology 50: 13231326. male volunteers. Mov Disord 17 (Suppl 5): S100S101.
Metman LV, Blanchet PJ, van den Munckhof P et al. (1998b). Pinto S, Ozsancak C, Tripoliti E et al. (2004). Treatments for
A trial of dextromethorphan in parkinsonian patients with dysarthria in Parkinsons disease. Lancet Neurol 3: 547556.
motor response complications. Mov Disord 13: 414417. Poulter MO, Payne KB, Steiner JP (2004). Neuroimmunophi-
Metman LV, Del Dotto P, Natt R et al. (1998c). Dextro- lins: a novel drug therapy for the reversal of neurodegen-
methorphan improves levodopa-induced dyskinesias in Par- erative disease? Neuroscience 128: 16.
kinsons disease. Neurology 51: 203206. Pourcher E, Bonnet AM, Kefalos J et al. (1989). Effects of ety-
Metman LV, Del Dotto P, LePoole K et al. (1999). Amanta- benzatropine and diazepam on levodopa-induced diphasic
dine for levodopa-induced dyskinesias: a 1-year follow-up dyskinesias in Parkinsons disease. Mov Disord 4: 195201.
study. Arch Neurol 56: 13831386. Rascol O, Blin O, Thalamas C et al. (1999). ABT-431, a D1
Metman LV, Gillespie M, Farmer C et al. (2001). Continuous receptor agonist prodrug, has efficacy in Parkinsons
transdermal dopaminergic stimulation in advanced Parkin- disease. Ann Neurol 45 (6): 736741.
sons disease. Clin Neuropharmacol 24: 163169. Rascol O, Brooks DJ, Korczyn AD et al. (2000). A five-year
Nishikawa T, Tanaka M, Tsuda A et al. (1984). Clonidine study of the incidence of dyskinesia in patients with early
therapy for tardive dyskinesia and related syndromes. Clin Parkinsons disease who were treated with ropinirole or
Neuropharmacol 7: 239245. levodopa. 056 Study Group. N Engl J Med 342: 14841491.
Nutt JG, Holford NH (1996). The response to levodopa in Rascol O, Nutt JG, Blin O et al. (2001a). Induction by dopa-
Parkinsons disease: imposing pharmacological law and mine D1 receptor agonist ABT-431 of dyskinesia similar
order. Ann Neurol 39: 561573. to levodopa in patients with Parkinson disease. Arch
Nyholm D, Nilsson Remahl AI, Dizdar N et al. (2005). Duo- Neurol 58: 249254.
denal levodopa infusion monotherapy vs oral polyphar- Rascol O, Arnulf I, Peyro-Saint Paul H et al. (2001b). Ida-
macy in advanced Parkinson disease. Neurology 64: zoxan, an alpha-2 antagonist, and L-DOPA-induced dyski-
216223. nesias in patients with Parkinsons disease. Mov Disord
Oh JD, Bibbiani F, Chase TN (2002). Quetiapine attenuates 16: 708713.
levodopa-induced motor complications in rodent and pri- Ravina BM, Fagan SC, Hart RG et al. (2003). Neuroprotec-
mate parkinsonian models. Exp Neurol 177: 557564. tive agents for clinical trials in Parkinsons disease: a sys-
Olanow CW, Damier P, Goetz CG et al. (2004). Multicenter, tematic assessment. Neurology 60: 12341240.
open-label, trial of sarizotan in Parkinson disease patients Reisine TD, Chesselet MF, Lubetzki C et al. (1982). A role
with levodopa-induced dyskinesias (the SPLENDID for striatal beta-adrenergic receptors in the regulation of
Study). Clin Neuropharmacol 27: 5862. dopamine release. Brain Res 241: 123130.
150 C. COLOSIMO AND G. FABBRINI

Savola JM, Hill M, Engstrom M et al. (2003). Fipamezole Stocchi F, Arnold G, Onofrj M et al. (2004). Improvement of
(JP-1730) is a potent alpha2 adrenergic receptor antagonist motor function in early Parkinson disease by safinamide.
that reduces levodopa-induced dyskinesia in the MPTP- Neurology 63: 746748.
lesioned primate model of Parkinsons disease. Mov Tarsy D, Leopold N, Sax DS (1974). Physostigmine in chor-
Disord 18: 872883. eiform movement disorders. Neurology 24: 2833.
Scheinin M, Lomasney JW, Hayden-Hixson DM et al. Thomas A, Iacono D, Luciano AL et al. (2004). Duration of
(1994). Distribution of alpha2-adrenergic receptor subtype amantadine benefit on dyskinesia of severe Parkinsons
gene expression in rat brain. Brain Res Mol Brain Res 21: disease. J Neurol Neurosurg Psychiatry 75: 141143.
133149. Venderova K, Ruzicka E, Vorisek V et al. (2004). Survey on
Schneider JS (1998). GM1 ganglioside in the treatment of cannabis use in Parkinsons disease: subjective improve-
Parkinsons disease. Ann NY Acad Sci 845: 363373. ment of motor symptoms. Mov Disord 19: 11021106.
Seager H (1998). Drug-delivery products and the Zydis fast- Waters CH, Sethi KD, Hauser RA et al. (2004). Zydis selegi-
dissolving dosage form. J Pharm Pharmacol 50: 375382. line reduces off time in Parkinsons disease with motor
Senard JM, Brefel-Courbon C, Rascol O et al. (2001). Orthostatic fluctuations: a 3-month, randomized, placebo-controlled
hypotension in patients with Parkinsons disease: pathophy- study. Mov Disord 19: 426432.
siology and management. Drugs Aging 18: 495505. Wint DP, Okun MS, Fernandez HH (2004). Psychosis in Par-
Shults CW, Oakes D, Kieburtz K et al. (2002). Parkinson kinsons disease. J Geriatr Psychiatry Neurol 17: 127136.
study group. Effects of coenzyme Q10 in early Parkinson Woitalla D, Muller T, Benz S et al. (2004). Transdermal
disease: evidence of slowing of the functional decline. lisuride delivery in the treatment of Parkinsons disease.
Arch Neurol 59: 15411550. J Neural Transm Suppl 68: 8995.
Sieradzan KA, Fox SH, Hill M et al. (2001). Cannabinoids Wolf WA (2003). SLV-308. Solvay. Curr Opin Investig
reduce levodopa-induced dyskinesia in Parkinsons dis- Drugs 4: 878882.
ease: a pilot study. Neurology 57: 21082111. Zesiewicz TA, Sullivan KL, Maldonado JL et al. (2005).
Silverdale MA, Nicholson SL, Crossman AR et al. (2002). Open-label pilot study of levetiracetam (Keppra) for the
Anti-dyskinetic actions of AMPA receptor antagonists in treatment of levodopa-induced dyskinesias in Parkinsons
the MPTP-lesioned marmoset model of Parkinsons dis- disease. Mov Disord 20 (9): 12051209.
ease. Mov Disord 17 (Suppl 5): S41. Zhang W, Ordway GA (2003). The alpha2C-adrenoceptor
Snow BJ, Macdonald L, Mcauley D et al. (2000). The effect modulates GABA release in mouse striatum. Brain Res
of amantadine on levodopa-induced dyskinesias in Parkin- Mol Brain Res 112: 2432.
sons disease: a double-blind, placebo-controlled study.
Clin Neuropharmacol 23: 8285.
Steece-Collier K, Chambers LK, Jaw-Tsai SS et al. (2000).
Further Reading
Antiparkinsonian actions of CP-101,606, an antagonist of Holloway RG, Shoulson I, Fahn S et al. (2004). Parkinson
NR2B subunit-containing N-methyl-d-aspartate receptors. Study Group. Pramipexole vs levodopa as initial treatment
Exp Neurol 163: 239243. for Parkinson disease: a 4-year randomized controlled
Stephenson DT, Meglasson MD, Connell MA et al. (2005). trial. Arch Neurol 61: 10441053.
The effects of a selective D2 receptor agonist on beha- Parkes JD, Debono AG, Marsden CD (1976). Bromocriptine
vioral and pathological outcome in 1-methyl-4-phenyl- in parkinsonism: long-term treatment dose response, and
1,2,3,6-tetrahydropyridine-treated squirrel monkeys. comparison with levodopa. J Neurol Neurosurg Psychiatry
J Pharmacol Exp Ther 314 (3): 12571266. 39: 11011108.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 38

The importance of patient groups and collaboration

MARY BAKER1* AND JILL RASMUSSEN2

1
European Parkinsons Disease Association (EPDA), Sevenoaks, Kent, UK
2
Merstham Clinic, Redhill, Surrey, UK

38.1. Introduction short of actual need as well. For the general well-being,
it is essential that the economic, social and policy issues
Historically, patient groups (PGs) were formed to offer that arise are settled in such ways that deliver an opti-
mutual support, not only for patients with serious mal balance between patients needs and available
or chronic debilitating disorders, but also for their rela- resources. PAGs hold the keys to this balance, but if
tives and friends, who in many cases undertook primary these are to be properly applied, PAGs must broaden
responsibility for day-to-day care. As these groups their perspectives to consider a patients general wel-
expanded, they focused first on access to professional fare, not just that related to a specific disease that,
and institutional care as needed and to improving insofar in many cases, will be complicated by comorbid or
as possible patients quality of life and a dignified and consequential disorders.
peaceful end. The next logical step soon followed when
PGs became legal charities to raise funds to increase pub- 38.2. The burden of diseases
lic awareness of particular diseases, to lobby policy-
makers to direct public attention and resources to obtain The World Health Organization (WHO) has developed
better access to treatment and to promote the search for methods to assess generally the burden of disease on
more effective remedies. In essence, they became patient societies (Murray and Lopez, 1996). The diseases
advocacy groups (PAGs) providing a public voice for underlying this burden vary significantly from region
patients and lobbying for their particular interests. to region, with the burden in developing countries aris-
There is little doubt that PAGs have made major contri- ing mainly from perinatal disorders, malnutrition, com-
butions to patients well-being and quality of life. In some municable disease and violence, whereas in developed
cases, PAGs have developed resources of their own that areas the burden arises mainly from non-communicable
have made material contributions to the discovery of more diseases (Murray and Lopez, 1996).
effective treatments and sometimes to cures. However, The WHO summarizes the burden of disease in terms
the majority of such development has depended and con- of disability-adjusted life-years (DALYs) which, in
tinues to depend upon commercial interests, academic turn, is the sum of years of life lost (YLL) and years of
institutions or publicly funded resources. life with disability (YLD). YLL represents the reduction
Not only because both charitable and public finan- in life expectancy whereas YLD represents the number
cial resources are limited, but also because the research of years lived with disability adjusted for the incidence
infrastructure required for both fundamental and applied of disease and the severity of associated disability, a
research into complex disorders, often with poorly factor ranging from none (0) to death (1) (Olesen and
understood etiology, is equally limited, there is compe- Leonardi, 2003).
tition for the available resources. This occurs against a Considering the European region (roughly, all
background where worldwide expenditure on health European countries west of Russia plus a few others),
care falls well short of demand and, in many cases, Olesen and Leonardi (2003) using the WHO data have

*Correspondence to: Dr. Mary Baker, c/o Lizzie Graham, European Parkinsons Disease Association (EPDA), 4 Golding Road,
Sevenoaks, Kent TN13 3NJ, UK. E-mail: lizzie@epda.demon.co.uk, Tel/Fax: 44-(0)-1732-457-683, Cell: 44-(0)-7787-554-
856, Website: http://www.epda.eu.com
152 M. BAKER AND J. RASMUSSEN
calculated that brain diseases are responsible for 23% of the PAGs whose principal goals of health care
of the years of healthy life lost and 50% of YLDs. In this advocacy are to raise awareness of specific issues
context, brain disease included neurological, neurosur- and make them national priorities (Carroll, 2004).
gical and psychiatric disorders together with half of the Instead of competing with one another for scarce
burden of injuries and congenital problems. Considering resources, PAGs must recognize that many specific dis-
the key summary measure of lost health, DALY, 35% orders have important perhaps critical features in
was related to brain diseases (Olesen and Leonardi, common and that synergy, especially at the level of
2003). Based on these data, these authors suggested that fundamental science, is very likely. Rather than com-
a third of the curriculum at medical school should deal peting on narrowly defined disease entities, the PAGs
with the brain and that a like proportion of life-science must coalesce in broadly based consortia that have a
funding should go to basic and clinical neuroscience much greater opportunity to influence public policy
and that overall a third of health care expenditure should and to define the objectives of basic research that is
be allocated to the prevention, diagnosis and treatment of certainly necessary to achieve breakthrough therapies
brain disease. for many, if not most, brain diseases. Application to
Given that brain disease underlies a significant pro- specific disorders could then follow.
portion of health care costs in Europe, a proportion
probably similar in other developed areas of the world, 38.4. Need for collaboration and partnerships
it will be incumbent upon PAGs to prioritize the actual
needs rather than the demands of patients so that PAGs must recognize that their roles and responsibil-
resources that may be available are applied most effec- ities, if first to their members, are not just for this par-
tively. In this respect, all PAGs, not just those represent- ticular group of patients, but also for society for
ing neuroscience, hold the key to resolving insofar as intelligent and equitable policy, for politics, friends,
possible the resource/demand dilemma. families, supporters, employers, scientists, health care
professionals, and no less for those who produce and
38.3. Responsible advocacy supply pharmaceuticals and devices. We are all fellow
travelers on a challenging journey. And we will all be
In representing patients, PAGs have a duty to listen to patients.
their patients wants, to filter out trivial wants and to If PAGs are to fulfill their wider responsibilities to
translate the remainder into prioritized needs consider- public welfare, they must achieve a difficult balance
ing in that process what is most probably achievable in between representing the needs of their patient consti-
the short, medium and longer term. For such an assess- tuency and the needs of the wider community of
ment, scientific and medical expertise will be required patients and society generally.
and for this most PAGs will require collaborators. There can be no doubt that the primary objective for
Having established a set of priorities that is scientifi- any PAG is to listen to and recognize the needs of its
cally, clinically and financially credible in terms of a members. There is otherwise no point in its existence.
specific disease, a PAG has an equal duty to consider However, the needs of patients and their best interests
whether the resource demands of its priorities are not are unlikely to be most effectively served by a narrow
disproportionate in light of the burden of disease. focus that excludes the interests of other patients or
Whereas PAGs naturally have a self-centered focus those of the larger society. Although its member
on specific diseases, this must necessarily be tempered patients may applaud such an exclusive focus simply
by a wider social responsibility to share equitably the because their own disorder looms largest in the frame-
limited resource pool. PAGs must share with govern- work of scientific or medical issues that urgently
ments, policy-makers as well as with medical and require resolution, such resolution is most likely to be
scientific communities the responsibility for achieving achieved not only through collaboration with the whole
an optimum balance between resources and the needs range of individuals and organizations with specific
of all patients, not just the few. expertise, but also with other PAGs and groups that
Even though brain disorders underlie more than a share similar representative and advocacy objectives.
third of DALYs, they remain rather a poor relation
when it comes to research. If health care is to have 38.5. Need for research partnerships and
any hope of efficient delivery, public policy and coalitions
research must target big ticket issues such as brain
disease. Although a policy change of this sort would Because PAGs in general have neither the expertise nor
constitute something of a paradigm shift, especially the financial resources to conduct primary research them-
in Europe, it requires no less a shift in the perspectives selves, when representing the interests of patients, PAGs
THE IMPORTANCE OF PATIENT GROUPS AND COLLABORATION 153
must seek and facilitate collaboration. Although PAGs 38.6. Partnerships for policy change
have recognized this, collaborations are usually limi-
ted to individuals or entities that are able and willing to Beyond partnerships of PAGs to promote progress on
focus narrowly on the particular disease entity the PAG issues of common interest, even wider alliances with
represents. other professional organizations undoubtedly improve
For example, in a workshop for PAGs sponsored both credibility and leverage when advancing the
by the American Society for Experimental Neuro social and economic justifications for directing public
Therapeutics, a speaker suggesting ways of engaging policy and resources. Whether or not within an alli-
academic partners (Benderly, 2004) recommended ance, PAGs must be effective at enabling, at bringing
that PAGs should try to attract promising young people of disparate interests together to spawn original
academic researchers who were just starting their thinking and to induce change.
careers. A small grant might be used to direct their When attempting to influence both public percep-
attention toward areas specific to the PG. Having tion and by extension public policy, the size of the pro-
thus captured an academic, the PAG could encou- blem obviously matters. Groups representing a large
rage continued loyalty by contributing to the indivi- patient population with significant demands on state
duals professional career by supporting grant or other resources will have greater success in getting
applications to other funding bodies, by offering their message across. They will be more able to high-
opportunities to publish and by other similar means. light the burden of illness substantiated by economic
However, such methods are inherently competitive, evidence, including both direct and indirect personal,
pitting PAGs against one another in the attempt to financial and societal costs.
harness the next generation of scientific and clinical Having got the attention of the public and policy-
expertise. makers, the PAGs must mediate between conflicting
Rather than competing, albeit with the best of interests and perspectives. For example, they need to
motives, greater progress is likely to arise from colla- engage clinicians and health economists (who are not
boration between PAGs. In many chronic, disabling natural bedfellows) with patients who have a sharp
conditions and certainly in neurology and psychiatry focus on their distress, but very little idea of the cost.
the disparate disease states as defined in the current Such engagement presents real potential for conflict,
nosology have more in common than not and separat- as the various interests are not easily reconciled.
ing one from another can be very problematic in vivo. Even apart from economics, patients and doctors
On this basis, common pathological mechanisms and have different perspectives and priorities. PAGs have
perhaps of etiology seem likely and the disease- an important mediating role in listening to patients
focused PAGs should join forces to support the and helping them to express their needs clearly and
search for those underlying mechanisms that could to encourage partnership in their care. Doctors typi-
allow earlier identification of risk or that may be cally focus on the specific disease, the site of pathol-
interrupted or modified before irreversible damage ogy and upon symptomatology. They measure illness,
has finally overwhelmed the compensating resources its progression and response to treatment, for example,
of the brain. according to neurological parameters. Patients, by con-
For example, a number of neurodegenerative disor- trast, focus on the effect of the illness and its treatment
ders associated with dementia (Alzheimers disease, on their daily lives (mood, pain, sleep, bodily function,
dementia of the Lewy body type, frontotemporal disability). Patients look at things in terms of their
dementia, prion disease) have a different predominant self-respect (work, independence) and relationships
pathology, but share a common mechanism of produc- with others. From that highly personalized perspective,
tion of the hallmark pathology, namely protein mis- specialized clinical assessments are largely meaning-
folding (Hammarstrom et al., 2001; Hardy, 2003). less and the primary debilitating effects often arise
What distinguishes their specific development is not from the underlying disease, but from such comor-
unclear, but is probably important in the search for bid conditions as depression or anxiety, from employ-
therapies or prevention. Coalitions of all PAGs repre- ment or financial concerns, or from sensitivity to
senting the disparate diseases should be formed to public stigma.
support research in such cases. It would also be rea- To insure that patients have access to management
sonable to expect that an alliance of PAGs represent- of all of their problems, not simply access to specia-
ing disparate diseases with some common features lized care, important as that is, PAGs need on the
could encourage thinking outside the box that often one hand to engender evidence-based policies, treat-
seems to produce breakthrough concepts. ment guidelines and management practices based on
154 M. BAKER AND J. RASMUSSEN
mass statistics whilst on the other hand representing nuclear family coupled with increasing full-time em-
the spectrum of disease and the real problems that par- ployment for both sexes results in an increasing
ticular cohorts experience. These problems often cross responsibility of the state to provide care. This is
specialist boundaries. inherently expensive and exacerbated by the demo-
graphic reality of an increasing proportion of older
38.7. Advocacy is not enough people in the population.
It is perhaps natural and certainly understandable
Although advocacy is a necessary, important and chal- that diseases that lead to premature death are frighten-
lenging function, it is not in itself sufficient. It is ing for most people. Understanding the personal
equally important that PAGs also take responsibility impact of an illness that gradually erodes the senses,
to inform and educate their members. They must pro- intellectual function or the ability to perform simple
vide their members not only with encouragement and motor tasks is much harder for healthy people to ima-
support, but also with realistic expectations. Patients gine. It therefore devolves to the PAGs to use every
have a right to see a doctor who understands their dis- possible tool at their disposal to educate the healthy
ease, but also a team that can recognize associated not only to recognize their personal risk, but more
problems and address them. Patients have a right to importantly, to gain some concept of the impact of
expect continuity of care, that is, to see the same doc- living with a disability that will only become more
tor and team at successive visits. But patients also severe. It must be obvious that the risk of a single dis-
have a duty to educate themselves about their disease, order such as Parkinsons disease is small compared
to recognize and understand its possible complications to the risk of one of the range of equally or more
and to have reasonable expectations concerning their debilitating degenerative neurological conditions.
care and to be prepared to participate fully and effec- Again, fragmenting the message among many specific
tively in their own treatment. It is a prime responsibil- disorders, each equally devastating, can hardly be as
ity of the PAGs to provide and deliver this sort of effective as a coordinated message. The PAGs have a
patient education. duty to their members, if not to society generally, to
As part of this educational responsibility, PAGs collaborate in the effort to educate the public, the
need to make it clear to their members that there is politicians and the policy-makers.
fierce competition for health care resources and that
patients with a particular disorder have no intrinsic 38.8. Research and development issues
right to a disproportionate share of the limited
resources. Distribution of available resources is inher- In the same way, the PAGs have a duty to collaborate
ently political. As such, politicians and policy-makers when representing their members in the many areas of
are tempted to respond to the views of the much larger common interest such as the ethical dimensions of
population of the generally healthy who tend to con- stem cell or animal research. The effort to relieve the
sider most serious those disorders that they believe suffering of patients should not be compromised by
present the greatest immediate risk to them personally. the fact that some necessary aspects of scientific and
PAGs representing patients with disorders that predo- clinical research may offend the sensibilities of those
minantly affect the elderly, including most of the with dogmatic views, however fervently held.
degenerative neurological diseases, are disadvantaged Collaboration of PAGs is also necessary if a uni-
in the competition for resources simply because the form standard of care is to be accessible to members
general public consider these to be remote risks. regardless of their disease, their station in life or where
Neurological disorders are often considered a threat they live. There are, for example, significant differ-
only to the old and victims are largely hidden from ences in reimbursement and in the time lags for access
society, whereas psychiatric disorders are associated to new treatments.
with public stigma and likewise hidden or denied Regulatory requirements and the associated time-
wherever possible. PAGs representing these patients costs of drug development coupled with a high risk of
must strive to illustrate the burden of these disorders failure and short patent terms constrain the develop-
and to raise their profile in public perception. In terms ment of new drugs. In particular, development for rela-
of DALYs and consequent direct and indirect costs tively rare conditions is unlikely. In addition, the cost
to society, brain diseases are very important. In the of new drugs is necessarily high to recoup development
past, spouses or other members of the immediate costs not only for the one registered drug, but also for
family have absorbed much of this personal as well the three or four that failed in development. Short
as financial cost. However, increasing dispersion of patent terms mean that most cost recovery has to be
the extended family as well as dissolution of the achieved before the patent expires and manufacturers
THE IMPORTANCE OF PATIENT GROUPS AND COLLABORATION 155
of generics who contribute nothing to research or the competition for an equitable distribution of limited
development flood the market. If the PAGs wish to research, development and care-centered resources.
encourage new therapies, they need to collaborate to Although it may well be desirable that disease-
lobby for regulatory changes that will encourage rather centered PAGs should retain a measure of individual
than stifle research and development. identity and autonomy, a much greater measure of
association and collaboration among the PAGs dealing
38.9. What next for patient advocacy? with brain-related diseases will be necessary if they
are to serve most effectively the needs of patients and
Finally, although there is no doubt that PAGs have to achieve a share of resources commensurate with the
made major contributions to the well-being of their impact of brain diseases on individuals, society and
patients and that continuing such contributions will be the health care systems.
required in future, changing times now require that the
PAGs should seriously consider altering their operating References
methods in areas of mutual interest.
In the encouragement and/or sponsorship of research Benderly BL (2004). Advocacy groups are crucial players in
into the underlying mechanisms of neurological disor- developing new neurotherapeutics. NeuroRx 1: 500502.
ders, a broader scope of enquiry that could promote lat- Carroll PR (2004). The impact of patient advocacy: the
eral thinking should prove more productive than one University of California-San Francisco experience. J Urol
based upon a narrow focus on a single disease entity. 172 (5 Pt 2): S58S61.
Close collaboration of several PAGs would be most Hammarstrom P, Schneider F, Kelly JW (2001). Trans-
appropriate and perhaps necessary to engender such suppression of misfolding in an amyloid disease. Science
293: 24592462.
research.
Hardy J (2003). Impact of genetic analysis on Parkinsons
In particular, there is compelling evidence from the disease research. Mov Disord 18 (Suppl 6): S96S98.
WHO for the importance of brain disorders, mainly Murray CJ, Lopez AD (1996). Evidence-based health policy
neurological and psychiatric, among the diseases con- lessons from the Global Burden of Disease Study. Science
tributing most significantly to the global burden of dis- 274: 740743.
ease. Fragmenting the voice of all affected patients Olesen J, Leonardi M (2003). The burden of brain diseases in
along disease-specific lines is counterproductive in Europe. Eur J Neurol 10: 471477.
Section 6

Complications of therapy
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 39

Motor and non-motor fluctuations

SUSAN H. FOX* AND ANTHONY E. LANG

Division of Neurology, University of Toronto, Toronto, ON, Canada

39.1. Introduction gait seen in response to a dose of levodopa (Table 39.1).


A PD patient exhibiting parkinsonian symptoms is
The cardinal symptoms of Parkinsons disease (PD), described as being off and an improvement in symp-
bradykinesia, rigidity and tremor, occur in large part toms after dopaminergic medication is termed on.
secondary to progressive degeneration of the dopami- The doseresponse to levodopa is often divided into
nergic neurons within the substantia nigra pars com- three timeframes: (1) the initial improvement or begin-
pacta of the midbrain. Treatment of PD is therefore ning of dose, when the patient first begins to notice an
based on dopamine replacement, either in the form of improvement in symptoms and switches on; (2) the
the dopamine precursor, levodopa (co-administered maximal time of improvement in parkinsonian symp-
with a peripheral dopa-decarboxylase inhibitor) or via toms, called peak dose; and (3) the beginning of loss
directly acting dopamine receptor agonists. Early treat- of benefit and re-emergence of parkinsonian symptoms
ment of PD with these agents results in effective and or end of dose. Motor fluctuations can affect all or part
sustained control of motor symptoms which may last of these levodopa dose-related timeframes. Thus a care-
for several years. With the progression of the disease ful history of the patients problems in relation to the
and the requirement for higher doses of levodopa, the timing of a dose of levodopa is critical to management
vast majority of patients start to experience a reduced and treatment of motor fluctuations. Measurement of
and variable benefit from medication. At this stage, motor fluctuations in response to medication is incorpo-
patients often experience fluctuations in response to rated into the Unified Parkinsons Disease Rating Scale
medication in both the motor symptoms (motor fluctua- (UPDRS).
tions), but also in non-motor symptoms (non-motor
fluctuations), such as psychiatric/behavioral problems, 39.2.1.1. Predictable wearing-off
autonomic dysfunction and sensory/pain symptoms. In Motor fluctuations tend to begin with a predictable
addition, PD patients may also experience spontaneous shortening in the duration of response to levodopa with
fluctuations in symptoms, unrelated to levodopa dos- gradual emergence of parkinsonian symptoms, called
ing. The presence of fluctuations in motor and non- wearing-off. This is usually apparent when the dura-
motor symptoms is almost invariable in PD patients tion of levodopa benefit is 4 hours or less (Muenter
and as such the absence of fluctuations may suggest that and Tyce, 1971; Shoulson et al., 1975; Fahn, 1982).
a patient does not have idiopathic PD (Quinn, 1998). The earliest manifestation of this is usually morning
akinesia, since the time between doses is generally
39.2. Levodopa-induced motor fluctuations longest overnight before the morning dose.
39.2.1. Definitions
39.2.1.2. Unpredictable, sudden offs
The commonest fluctuations to emerge in advancing PD Patients can also experience wearing-off that is fast,
are the variable and often unpredictable benefits on the random and unrelated to the timing of the last dose
motor symptoms of bradykinesia, rigidity, tremor and of levodopa, called unpredictable offs. Such patients

*Correspondence to: Susan H. Fox, Toronto Western Hospital, Movement Disorders Clinic, Division of Neurology, University
of Toronto, 399 Bathurst Street MCL7412, Toronto, ON M5T 2S8, Canada. E-mail: sfox@uhnresearch.ca, Tel: 1-416-603-
5383, Fax: 1-416-603-5004.
160 S. H. FOX AND A. E. LANG
Table 39.1 lower-volume and increasingly inaudible; this is often
associated with high doses of levodopa and is an on-
Levodopa-induced motor fluctuations in Parkinsons disease
period phenomenon. Tachyphemia is often associated
Predictable wearing-off with a similar phenomenon in walking: an increasing
Unpredictable, sudden offs speed of walking even to the point of running, with
Dose failure, delayed or partial on response smaller steps, which may lead to falls (Giladi et al.,
Beginning-of-dose worsening 1997).
End-of-dose rebound
On- and off-period freezing (motor blocks) 39.2.1.6. Dyskinesia
Tachyphemia and running gait
Dyskinesia At the same time as the above fluctuations start to
Peak dose/square-wave dyskinesia occur, patients often experience involuntary move-
Wearing-off/off-period dystonia ments or dyskinesia. Dyskinesia can occur at variable
Diphasic dyskinesia times in response to levodopa. Thus dyskinesia most
Onoff fluctuations/yo-yoing commonly occurs at the peak dose of levodopa action
(peak-dose dyskinesia) or throughout the duration of
the on-period (square-wave dyskinesia) and can be a
can suffer off-periods that come on over a few seconds
mixture of chorea, ballism and dystonia and to a lesser
with resultant severe akinesia, called a sudden off
extent myoclonus (Nutt, 1990). Choreiform move-
(Fahn, 1974).
ments in the limbs are most common but dystonic pos-
turing in the limbs and craniocervical dystonia and
39.2.1.3. Dose failure, beginning-of-dose worsening,
chorea are also common (Luquin et al., 1992). Off-per-
end-of-dose rebound
iod or wearing-off-period dyskinesia is predominantly
Patients may also experience delay in benefit dystonic and tends to affect the limbs, particularly
(delayed on) or absence of benefit from a dose of the legs and feet (Poewe et al., 1988; Bravi et al.,
levodopa, called dose failure (Melamed and Bitton, 1993). Patients often experience a more fixed posture
1984). Some patients can experience a transient wor- (e.g. ankle intorsion with toe flexion or extension),
sening of symptoms at the beginning of dose, often especially early morning, often associated with pain
as an increase in tremor (beginning-of-dose worsen- (early-morning dystonia) (Melamed, 1979). Less com-
ing) (Merello and Lees, 1992). In some cases, the off mon is dyskinesia occurring at the beginning and end
state may be worse than in the untreated state or after of dose, when the levels of levodopa are rising and
longer periods of drug withdrawal (super off) and falling, respectively. This is known as diphasic dyski-
these patients may experience an exacerbation or nesia, or beginning-of-dose and end-of-dose dyskine-
rebound in their symptoms at the end of dose (end- sia. This has also been referred to as dystonia
of-dose rebound) (Nutt et al., 1988). improvementdystonia or DID pattern (Muenter
et al., 1977). Diphasic dyskinesia tends to affect the
39.2.1.4. On- and off-period freezing (motor blocks) legs predominantly (Marsden et al., 1982) and can
One particular problem that can occur is a transient involve stereotypical rapid alternating leg movements,
difficulty initiating a movement a sudden transient as well as unusual ballistic kicking or dystonia
freezing (also called motor blocks) that may occur when (Luquin et al., 1992).
starting to walk (start hesitation), while turning (turning
hesitation) or going through a narrow doorway (Giladi 39.2.1.7. Onoff fluctuations
et al., 1992; Fahn, 1995). Once the block is overcome
With further disease progression and levodopa treat-
then the activity can be completed smoothly. In addi-
ment, patients can experience predictable or unpre-
tion, freezing can occur during other activities such as
dictable switching from being on and mobile with
speech and writing (Fahn, 1995). Motor blocks can also
dyskinesia to being off and immobile, known as
occur due to sudden stress or anxiety (startle hesitation).
onoff fluctuations (Duvoisin, 1974; Fahn, 1974).
Such freezing episodes most commonly occur when the
The term onoff implies a transition from one state
patient is in the off state but may also occur in the on
to the other akin to switching on and off a light.
state (on-period freezing).
Patients with advanced PD can develop a yo-yoing
effect where they rapidly, unpredictably and repeat-
39.2.1.5. Tachykinetic problems edly switch from being on with dyskinesia to off
Some patients with PD may experience increased and then on again (Fahn 1974, 1982; Marsden and
speed of speaking (tachyphemia) that also becomes Parkes, 1976).
MOTOR AND NON-MOTOR FLUCTUATIONS 161
39.3. Spontaneous motor fluctuations system and possibly other dopamine pathways, e.g. the
nigropallidal pathway, together with peripheral pharma-
Motor fluctuations can also occur spontaneously as cokinetic factors. Disease progression with loss of
part of the disease and not necessarily related to nigrostriatal dopaminergic terminals results in altered
long-term levodopa therapy. There is often a diurnal central pharmacokinetics of levodopa (presynaptic
variation in symptoms with improvement in the morn- changes); peripheral pharmacokinetics of levodopa and
ing and deterioration through the day (Struck et al., the duration and dose of levodopa therapy result in
1990; Nutt and Holford, 1996). Marked diurnal varia- changes to postsynaptic dopamine receptor signaling
tion is seen in young-onset PD patients with mutations (central pharmacodynamic or postsynaptic changes).
in the parkin gene (Khan et al., 2003). Another sponta- These factors result in abnormal intrasynaptic dopamine
neous motor fluctuation, first observed in the prelevo- concentration with loss of normal constant stimulation
dopa era, is paradoxical kinesia. This was particularly of postsynaptic dopamine receptors.
apparent in postencephalitic parkinsonism. Paradoxical
kinesia is a sudden improvement in motor activity in 39.5.1. Central pharmacokinetics of levodopa
response to a mental stress, e.g. a fire alarm or as a
reflex response to being thrown a ball (Quinn, 1998). In early PD, the clinical response to a single dose of
Although some fluctuations mentioned earlier can be levodopa is stable and lasts for several hours, despite
exacerbated by levodopa, they can also be a feature the half-life of levodopa being only 6090 minutes
of the underlying disease; these include motor blocks (Muenter and Tyce, 1971). Thus if a patient misses a
and tachykinetic symptoms. dose of levodopa, there is no recurrence of symptoms.
This is called the long-duration response (LDR) to
39.4. Epidemiology of motor fluctuations levodopa (Nutt and Holford, 1996; Obeso et al.,
2000a). The duration of the LDR, i.e. the time to com-
The incidence of motor fluctuations in PD has been plete loss of drug effect after stopping levodopa, is
consistently shown to increase with both duration of thought to last 12 weeks (Olanow et al., 1995; Hauser
levodopa therapy and duration of disease. The earliest et al., 2000), but recent studies have suggested that this
reports estimated that 10% of patients per year devel- may extend for up to 4 weeks (Hauser and Holford,
oped motor fluctuations after the initiation of levodopa 2002; Fahn et al., 2004) or even longer. The exact ori-
therapy (Marsden and Parkes, 1977). The frequency of gin of this LDR is unknown but it may be partly due to
wearing-off/onoff motor fluctuations and dyskinesia levodopa conversion to dopamine in remaining nigros-
has been estimated from a cumulative literature review triatal terminals and storage in synaptic vesicles for
(between 1966 and 2000) to be 40% and 50%, respec- release as required, thus mimicking the normal physio-
tively, after 46 years of levodopa treatment (Ahlskog logical function of the dopamine terminals (Quattrone
and Muenter, 2001). In recent prospective randomized et al., 1995; Zappia et al., 1999). The LDR may also
clinical trials comparing initiation of levodopa with a be due to postsynaptic changes in dopamine receptor
dopamine agonist, where the development of motor pharmacology and second-messenger systems, as the
fluctuations was the primary endpoint of the study, LDR is similar in dopa-responsive dystonia where
4054% experienced dyskinesia and 3462.7% wear- there is no presynaptic loss of dopamine terminals
ing-off after 45.5 years of follow-up (Rascol et al., (Nutt and Nygaard, 2001) (see below). In PD patients
2000; Parkinson Study Group, 2004). with motor fluctuations, the duration of the LDR short-
Accurate measurement of the current prevalence of ens and the rate of decline in LDR after stopping levo-
motor fluctuations experienced by PD patients, how- dopa is greatest in more severely affected patients
ever, is marred by variability in ascertainment (Marras (Quattrone et al., 1995; Zappia et al., 1999; Nutt
and Lang, 2003) and the effect of changes in treatment et al., 2002).
practice over the past two decades. A community-based In advanced PD, the response to levodopa is domi-
study of 124 PD patients found that 20% were experien- nated by a short-duration response (SDR), which is
cing dyskinesia and 30% response fluctuations after a the loss of benefit, typically less than 4 hours following
mean disease duration of 6 (4.3) years (Schrag et al., a dose of levodopa. At this stage, patients start to
2002). experience fluctuations in their response to levodopa
with wearing-off and thus become more dependent on
39.5. Pathophysiology of motor fluctuations the timing of each levodopa dose. These wearing-off
symptoms are probably due to a progressive loss of pre-
Motor fluctuations are due to a combination of pre- and synaptic dopaminergic terminals with a loss of dopa-
postsynaptic changes in the nigrostriatal dopaminergic mine storage (Fabbrini et al., 1988; Chase et al., 1993;
162 S. H. FOX AND A. E. LANG
Verhagen Metman et al., 2000). Loss of dopaminergic 39.5.3. Central pharmacodynamics
terminals means levodopa is converted to dopamine in
other aromatic acid-decarboxylase-containing cells, Central pharmacodynamics or postsynaptic mechan-
such as serotonergic and endothelial cells (Ng et al., isms are probably the most important factor in the
1971; Melamed et al., 1980). These cells lack the ability development of motor fluctuations. Thus, wearing-off
to store dopamine and thus non-regulated release of and dose failures also occur following long-term levo-
dopamine into the synaptic space may cause fluctua- dopa therapy in animal models of parkinsonism, where
tions. In patients with motor fluctuations, compared to there is no progressive loss of nigrostriatal terminals
those without, there is decrease in the latency to onset (Papa et al., 1994; Chase, 1998a). In normal primates,
and peak response of levodopa (Nutt et al., 1992, long-term treatment with levodopa can induce dyski-
2002; Colosimo et al., 1996). In early PD there is a lin- nesia which is dose-dependent; thus nigrostriatal
ear relationship between dose of levodopa and response; degeneration is not a prerequisite for motor fluctua-
however, in advanced PD with motor fluctuations, this tions to develop (Pearce et al., 2001; Togasaki et al.,
doseresponse switches to a sigmoid curve; thus 2001). (However, fluctuations will develop quicker
patients switch on at a critical level, below which they with a greater loss of nigrostriatal terminals: see
are off and above which they are essentially fully on below.) In addition, the shortening of the LDR as mea-
(Mouradian et al., 1988). sured by the rate of antiparkinsonian response decline
Altered synaptic dopamine turnover has also been following withdrawal of medication (turning off a
suggested to underlie motor fluctuations (Torstenson constant intravenous infusion of levodopa), which
et al., 1997). [11C]Raclopride positron emission becomes increasingly faster from mild, untreated PD
tomography (PET) studies have shown that PD patients to those with severe onoff fluctuations, is
patients who subsequently develop motor fluctua- also seen using infusions of the dopamine agonists
tions, even before these occur, have three times apomorphine and lisuride, which act predominantly
greater levels of synaptic dopamine 1 hour after levo- on postsynaptic dopamine receptors, independent of
dopa administration compared to non-fluctuators and, presynaptic terminals (Bravi et al., 1994; Verhagen
after 4 hours, only the non-fluctuators were able to Metman et al., 1997; Stocchi et al., 2001a). In addi-
maintain a stable level of synaptic dopamine (de la tion, eliminating variability in synaptic dopamine
Fuente-Fernandez et al., 2001). However, in PD patients levels with a constant infusion of levodopa still results
experiencing peak-dose dyskinesia, synaptic dopamine in fluctuations as measured by changes in tapping
levels were higher 1 hour post-levodopa but there was speed as an indicator of bradykinesia (Nutt et al.,
no difference after 4 hours with non-dyskinetic patients 1997). Thus central pharmacokinetics of levodopa is
(de la Fuente-Fernandez et al., 2004). Thus, it is unclear not the only factor in the development of a fluctuating
if levodopa-induced changes in synaptic dopamine are response. This implies that the basis for the develop-
compensatory to underlying disease or a cause of motor ment of motor fluctuations is probably due to erratic
fluctuations. or pulsatile stimulation of the postsynaptic dopamine
receptor and consequent downstream processes.
39.5.2. Peripheral pharmacokinetics of levodopa However, the extent of presynaptic denervation
clearly contributes by altering the frequency of dopa-
Gastrointestinal factors may also play an important role mine receptor stimulation, which then sets up changes
in motor fluctuations due to variability of levodopa within the postsynaptic signaling pathways. Thus in
delivery to the brain. Levodopa crosses the wall of PD patients, dyskinesia occurs earlier and is more
the small intestine and the bloodbrain barrier via a severe on the most affected side (Horstink et al.,
saturable carrier-mediated transporter. Competition 1990). In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
for this transporter between dietary amino acids and (MPTP) parkinsonism, with a greater degree of dopami-
levodopa can result in delayed switching on or failure nergic denervation than idiopathic PD, dyskinesia
of a dose of levodopa to have any benefit (Nutt et al., occurs earlier, on average 6 months, after initiation of
1984). Other factors that affect levodopa absorption levodopa (Ballard et al., 1985).
include slow or erratic gastric emptying, which may The exact postsynaptic mechanisms responsible for
occur secondary to concurrently used anticholinergics inducing motor fluctuations are unclear but probably
and food (Nutt and Fellman, 1984), as well as to age consist of multiple changes to neurotransmitter signal-
and PD itself (see section 39.6.2.2). All of these fac- ing pathways within the basal ganglia (Crossman,
tors combined can result in an important contribution 1990; Obeso et al., 2000b; Bezard et al., 2001). To date,
of peripheral pharmacokinetics to the chance of erratic most investigations into postsynaptic mechanisms have
and unpredictable responses to levodopa. evaluated levodopa-induced dyskinesia specifically.
MOTOR AND NON-MOTOR FLUCTUATIONS 163
On the other hand, animal models of PD with levodopa- pathway, which is an inhibitory GABAergic pathway
induced dyskinesia also exhibit wearing-off fluctuations from the caudate putamen (striatum) to the internal
(Papa et al., 1994) and, in advanced PD, patients with segment of the globus pallidus, this may lead to
dyskinesia invariably exhibit other types of motor increased activation of this pathway, which is thought
fluctuations, thus the neural mechanisms underlying to underlie levodopa-induced dyskinesia (Bezard et al.,
dyskinesia may be similar to those underlying other 2001) (see Ch. 40).
motor fluctuations. However, postmortem studies have Non-dopaminergic neurotransmitter systems have
suggested that some differences exist between PD also been shown to be associated with the develop-
patients who experience dyskinesia only compared to ment of motor fluctuations. In animal models of PD
those with wearing-off only. For example, in patients with motor fluctuations, activation of the dopamine
with wearing-off only, putaminal dopamine was mainly D1-receptor results in enhanced phosphorylation of
metabolized intraneuronally whereas in patients with N-methyl-d-aspartate (NMDA) glutamate receptor
dyskinesia only, the dopamine was mainly extraneuro- subunits within the striatum (Oh et al., 1999; Dunah
nal (Rajput et al., 2004). In addition, increased GABAA and Standaert, 2001; Calon et al., 2002a). NMDA
receptors in the putamen have been linked to the patho- receptor antagonists will reduce motor fluctuations
physiology of wearing-off but not to dyskinesia (Calon in animal models of PD (Papa et al., 1995; Papa and
et al., 2003). To date, however, the exact distinction, Chase, 1996) and the non-selective NMDA receptor
if any, between the neural mechanisms underlying dys- antagonist amantadine is effective in reducing the
kinesia, wearing-off and other on/off fluctuations (that severity of dyskinesia in clinical practice (Verhagen-
are unrelated solely to the peripheral pharmacokinetics Metman et al., 1998). The NMDA receptor is critical
of levodopa, such as delayed-on) is unknown. to synaptic plasticity and in levodopa-induced
The phasic stimulation of postsynaptic dopamine dyskinesia it appears that stimulation of striatal dopa-
receptors by repeated doses of levodopa is clearly a minergic receptors functionally linked to NMDA
factor in the development of motor fluctuations. Con- neurotransmission may lead to a form of synaptic plas-
sistent with this is the observation in animal models ticity similar to long-term potentiation (LTP) in the
of PD that de novo treatment with chronic continuous hippocampus involved in memory and learning. Thus
levodopa results in fewer or no motor fluctuations in the striatum in levodopa-induced dyskinesia, exces-
compared to repeat dosing with intermittent levodopa sive LTP may contribute to the development of motor
(Engber et al., 1989; Blanchet et al., 1995). In PD fluctuations (Calabresi et al., 2000, Picconi et al.,
patients, de novo therapy with long-acting dopamine 2003).
agonists reduces motor fluctuations compared to Increases in the opioid peptides within the striatum
short-acting levodopa (Rascol et al., 2000; Parkinson may also be associated with motor fluctuations, as
Study Group, 2004). Thus, less pulsatile dopaminergic shown by evidence from animal models (Henry and
stimulation reduces the risk of developing motor fluc- Brotchie, 1996; Andersson et al., 1999; Henry et al.,
tuations by preventing postsynaptic changes that occur 1999; Klinteberg et al., 2002), human postmortem stu-
with intermittent stimulation. dies (Calon et al., 2002b; Henry et al., 2003) and PET
To date, no consistent changes have been demon- studies (Piccini et al., 1997). In PD patients with motor
strated in dopamine D1-, D2- or D3-receptor subtype fluctuations, non-selective opioid receptor antagonists
number or affinity in levodopa-treated PD patients to extend on-time (Fox et al., 2004) but have no effect on
account for the development of motor fluctuations established dyskinesia (Rascol et al., 1994; Manson
(Rinne et al., 1991; Turjanski et al., 1997). The old et al., 2001; Fox et al., 2004). The exact role of opioids
explanation of dopamine receptor supersensitivity, in dyskinesia is unclear, as recent studies in MPTP-
as a consequence of dopamine depletion, causing lesioned primates have shown that opioid antagonists
motor fluctuations and particularly dyskinesia, is unli- can increase dyskinesia (Samadi et al., 2003). Evidence
kely, as dyskinesia rarely appears with the first doses that other non-dopaminergic neurotransmitter systems
of levodopa, even in severely affected patients, or in are involved in motor fluctuations comes from preclinical
MPTP-lesioned primates with profound dopamine studies in animal models of PD (Brotchie et al., 2005)
terminal loss. Some recent evidence suggests that there and from clinical studies (Silverdale et al., 2003) using
may be increased signaling downstream of striatal selective a2-adrenoceptor antagonists (Bara Jimenez
dopamine D1-receptors in MPTP-lesioned primates et al., 2004); 5HT1A-receptor agonists (Olanow et al.,
with dyskinesia compared to non-dyskinetic animals, 2004) and cannabinoids (Sieradzan et al., 2001). Thus a
despite the absence of receptor number changes range of non-dopaminergic neurotransmitter systems are
(Aubert et al., 2005). As dopamine D1-receptor signal- probably involved in the pathophysiology of motor
ing regulates the activity of the so-called direct fluctuations.
164 S. H. FOX AND A. E. LANG
39.6. Treatment of levodopa-induced motor 2000). A new patch formulation of the dopamine ago-
fluctuations nist rotigotine is in clinical development (Parkinson
Study Group, 2003). To date, there are no comparisons
39.6.1. Predictable wearing-off
of efficacy between the newer dopamine agonists.
39.6.1.1. Alter dose interval or preparation of Side-effects of dopamine agonists have been discussed
levodopa earlier (see Ch. 33).
End-of-dose or wearing-off phenomenon, if mild, can
be initially treated by decreasing the time interval
39.6.1.3. Add a catechol-O -methytransferase
between dosing of levodopa to give the next dose just
(COMT) inhibitor
before the beneficial effects have worn off. Alterna-
tively or in addition, if the patient is not experiencing COMT is an enzyme involved in the breakdown of
dyskinesia and off-periods predominate, then the dose levodopa and dopamine to methylated derivatives.
of levodopa can be increased. As discussed earlier, Inhibition of this enzyme therefore increases the elim-
because of the change in the doseresponse curve to ination half-life of levodopa, thus increasing bioavail-
levodopa in advanced PD, increasing individual doses ability (Ruottinen and Rinne, 1996a). The other
of levodopa will not improve the quality of the on- pharmacological effect of repeated dosing with COMT
period, but will simply increase the duration (Marsden, inhibitors is a reduction in the usual peaks and troughs
1994). of levodopa levels, i.e. a smoothing-out of levodopa
Longer-acting levodopa preparations such as Sine- levels through the course of the day, resulting in an
met CR or Madopar CR may help improve on-time increase in mean daily plasma levodopa levels (Nutt
by 11.5 hours (Ahlskog et al., 1988; MacMahon et al., 1994).
et al., 1990). However, the bioavailability of these There are currently two COMT inhibitors available,
longer-acting preparations is 2030% less than stan- entacapone and tolcapone. However, tolcapone is una-
dard-release preparations and patients usually end up vailable in many European countries and Canada due
on higher overall daily doses of levodopa, with a risk to concerns regarding liver toxicity and, in those coun-
of exacerbating dyskinesia, particularly later in the tries where available, restrictions apply to its use.
day (Lieberman et al., 1990). In addition, these pre- Tolcapone is administered three times daily, usually
parations also typically have a longer latency to bene- in 100-mg doses, irrespective of levodopa dosing.
fit so patients may experience delayed-on problems; The shorter half-life of entacapone (1 hour) means that
especially if doses are not overlapping (i.e. the patient each 200-mg dose has to be co-administered with
wears off before the next dose). The problem with all levodopa. A combined preparation of levodopa/carbi-
the above strategies is that, because the dose of levo- dopa and entacapone (Stalevo) in a single tablet is
dopa is increased, there is a risk of exacerbating motor now available and is undergoing clinical trials (Koller
fluctuations in the future. et al., 2005). Many clinical trials have demonstrated
improved on-time with the addition of entacapone
(Ruottinen and Rinne, 1996b; Parkinson Study Group,
39.6.1.2. Add a dopamine receptor agonist 1997) or tolcapone (Kurth et al., 1997; Rajput et al.,
Adding an oral dopamine receptor agonist is a better 1997) to levodopa, on average by 12 hours. There
option as it minimizes the need to increase the dose are no differences in the responses obtained using
of levodopa. All dopamine agonists in clinical prac- levodopa/carbidopa, levodopa/benserazide prepara-
tice, such as bromocriptine, ropinirole, pergolide, pra- tions or with controlled-release preparations of levo-
mipexole and cabergoline, reduce off-time and dopa (Poewe et al., 2002). Although randomized
improve the duration of on-time when combined with comparative studies have not been performed, it is
levodopa (Hoehn and Elton, 1985; Lieberman et al., common clinical impression that tolcapone may be
1993; Olanow et al., 1994; Rascol et al., 1996; Pinter more efficacious than entacapone in controlling motor
et al., 1999). In addition, it may be possible to reduce fluctuations (Factor et al., 2001). This may relate
the dose of levodopa. Systematic review and analysis to the purely peripheral effects of entacapone on
of clinical trials comparing bromocriptine with ropinir- degradation of levodopa as compared to the additional
ole, cabergoline and pramipexole as add-on therapy central effects of tolcapone on dopamine metabolism
have shown no significant differences in efficacy (Kaakkola et al., 1994; Forsberg et al., 2003).
(Clarke et al., 2000; Clarke and Deane, 2001a, b; Early side-effects of COMT inhibitors include
Goetz, 2003). In two trials, pergolide was more effica- prolongation of existing on-period dyskinesia, usually
cious at reducing motor impairment and disability without an increase in severity, as there is no change
compared to bromocripine (Clarke and Speller, in peak levodopa level after a single dose. However,
MOTOR AND NON-MOTOR FLUCTUATIONS 165
as the level of levodopa may accumulate following not specifically due to worsening of dyskinesia (Tetrud
repeated dosing, then severity of dyskinesia may also and Koller, 2004). To date, no comparisons have been
increase (Ruottinen and Rinne, 1996a; Kurth et al., made with conventional selegiline in terms of improv-
1997; Muller et al., 2000). The dyskinesia can be ing wearing-off symptoms.
improved by reducing the dose of levodopa (Myllyla Rasagiline, a newly developed irreversible MAO-B
et al., 2001). Later side-effects include abdominal pain inhibitor, is 1015 times more potent than selegiline
and severe diarrhea that can occur after a few weeks of and is not associated with amphetamine metabolites
treatment and may lead to discontinuation in 510% of (Finberg et al., 1999). A double-blind RCT showed
patients (Rajput et al., 1997; Myllyla et al., 2001). Tol- that in PD patients with onoff fluctuations, rasagiline
capone requires liver enzyme monitoring; significant up to 2 mg/day improved motor and activities of daily
elevation of transaminases can occur in about 3% of living on-period UPDRS scores compared to placebo,
patients and 3 unmonitored patients died of acute but there was no significant decrease in off-periods
hepatic failure before this complication was recog- (Rabey et al., 2000). However, a more recent study
nized (Rajput et al., 1997; Assal et al., 1998). Entaca- found that rasagiline 0.5 and 1 mg/day reduced total
pone does not cause elevation of liver enzymes daily off-time by 1.41 and 1.85 hours, respectively
(Myllyla et al., 2001). The difference between tolca- (Parkinson Study Group, 2005) (see Ch. 34).
pone and entacapone in inducing liver enzymes does
not relate to the COMT inhibition per se but rather to 39.6.2. Dose failure, delayed or partial On
an effect on other metabolic pathways, possibly via
inhibition of mitochondrial adenosine triphosphate 39.6.2.1. Improve gastric emptying by taking
synthesis (Korlipara et al., 2004) (see Ch. 32). levodopa on an empty stomach
Impaired absorption of levodopa in the small intestine
may cause either a delayed or partial improvement of
39.6.1.4. Add a monoamine oxidase B inhibitor
parkinsonian symptoms or result in no benefit a dose
Monoamine oxidase B (MAO-B) inhibition can extend failure or no-on. Levodopa absorption may be impaired
the duration of action of levodopa by inhibiting the by delay in gastric emptying secondary to the presence of
metabolism of dopamine and increasing dopamine food (Fahn, 1977; Baruzzi et al., 1987). Initially patients
levels by up to 70% in the brain (Riederer and Youdim, are advised to take levodopa with food to prevent nausea
1986). Several early clinical studies demonstrated that but with time this requirement diminishes. Thus advising
the MAO-B inhibitor, selegiline (510 mg/day) had a a patient to take levodopa on an empty stomach will
mild effect on PD patients with wearing-off motor fluc- improve absorption and reduce dose failures (Contin
tuations (Lees et al., 1977; Golbe et al., 1988). There et al., 1998).
was no benefit in patients with marked, disabling on
off fluctuations and the main side-effects related to 39.6.2.2. Improve gastric emptying by stopping
increased dopamine levels, such as nausea and dyskine- anticholinergics and treating constipation
sia, which improved on reduction of levodopa dose.
Gastric emptying is also erratic and slow in PD due to
A new transmucosal preparation of selegiline
the underlying disease per se as well as secondary to
(Zydis selegiline) is now available. This is placed on
dopaminergic and anticholinergic medications (Evans
the tongue and is rapidly absorbed directly into the
systemic circulation, resulting in more reliable and et al., 1981; Edwards et al., 1992). In addition, consti-
pation via a cologastric reflex leads to delayed gastric
higher blood levels of selegiline (Seager, 1997; Clarke
emptying (Bojo and Cassuto, 1992). A slower speed of
et al., 2003). This bypasses first-pass hepatic metabo-
gastric emptying has been demonstrated to correlate
lism and reduces production of theoretically toxic
with the presence of motor fluctuations (Djaldetti
amphetamine-like metabolites (Clarke et al., 2003).
et al., 1996).
A single double-blind, randomized, placebo-controlled
trial (RCT) has shown that Zydis selegiline (2.5 mg/
day) significantly reduced total daily off-time in PD 39.6.2.3. Liquid/soluble levodopa preparations
patients, with predictable wearing-off, by 2.2 hours Soluble levodopa preparations are more rapidly and
compared to 0.6 hours for placebo (Waters et al., reliably absorbed compared with conventional levodopa
2004). The main side-effects were dizziness, halluci- (Stocchi et al., 1994). Preparations that exist include
nations, headache and dyskinesia, although there was levodopa/benserazide (Madopar dispersible), levodopa
no significant difference in on-time with dyskinesia methyl ester and levodopa/carbidopa (Parcopa). Alter-
between placebo and Zydis selegiline. Levodopa doses natively, patients can crush oral levodopa preparations
were reduced by 20% with Zydis selegiline, although and take them with a carbonated beverage. Patients
166 S. H. FOX AND A. E. LANG
requiring frequent smaller doses to control complicated 39.6.3. Unpredictable OnOff fluctuations
fluctuations and disabling dyskinesias can take liquid
levodopa preparations by crushing tablets in water Unpredictable onoff fluctuations appear in more
combined with ascorbic acid; however, the solutions advanced disease and are more resistant to treatment
are unstable after 24 hours (Kurth et al., 1993a). Such than predictable wearing-off. Patients are unable to
soluble preparations of levodopa are more rapidly and correlate on- and off-periods with their levodopa dos-
reliably absorbed than conventional tablets but have a ing schedule and they may suddenly switch from one
shorter duration of action 11.5 hours. to the other over a few seconds. Such fluctuations are
typically associated with concurrent dyskinesia, thus
39.6.2.4. Bypass gastric problems pharmacological treatment is difficult and often such
There have been several attempts at delivering levo- patients will require surgical intervention to allow a
dopa, in various preparations, directly into the duode- reduction in levodopa dosage.
num as a way of circumventing the stomach and so
improving absorption (Kurlan et al., 1986; Kurth 39.6.3.1. Increase oral dopaminergic agents (as for
et al., 1993b; Nyholm et al., 2005). One preparation of predictable wearing-off)
levodopa used is Duodopa, a stable suspension of levo- Increasing dopaminergic stimulation with increased
dopa and carbidopa in methylcellulose, which improves frequency and/or dosing of levodopa may reduce sud-
levodopa solubility (Nyholm et al., 2005). These studies den onoff fluctuations by smoothing out dopamine
have been performed in small numbers of patients and levels. However, these unpredictable onoff fluctua-
have not been double-blind or placebo-controlled, so tions do not appear to correlate with levodopa timing
the true efficacy is unclear. However, these studies have or levels. Patients often have associated on-period
shown improved on-time without an increase in dyski- choreiform dyskinesia and so this regime can exacer-
nesia in advanced PD patients (see later). One study bate involuntary movements. Specific therapies to
followed patients for 10 years and noted particular ben- reduce on- and off-period dyskinesias are discussed
efit in reducing dyskinesia (Syed et al., 1998). In all in Chapter 40.
cases, however, technical and mechanical issues may Increasing the dose of a dopamine agonist is a better
limit widespread use. option as this may permit a reduction in the levodopa
dose. The addition of entacapone is less helpful for
39.6.2.5. Take levodopa when still in On state unpredictable sudden offs compared to predictable
Occasionally patients with advanced PD experiencing wearing-off (Gordin et al., 2003). Whatever approach
motor fluctuations may report that the first dose of is taken, PD patients with advanced disease often
the day is less effective (Melamed and Bitton, 1984) cannot tolerate even small increases in dopaminergic
(in general, however, PD patients tend to notice that stimulation because of a disabling increase in dyskine-
their response to medication is usually better in the sia or the development of behavioral and psychiatric
morning and deteriorates over the course of the day). problems. At this stage, the response to intermittent oral
Some patients need a larger dose to kick in; or if they medication becomes brittle and unpredictable.
become very parkinsonian or super off, then a More frequent, smaller doses attempting to address
usually effective dose of levodopa does not switch the shortening response without aggravating dyskine-
them on. This can often be observed when performing sias sometimes result in a more unpredictable response
acute levodopa challenges in the practically defined pattern with a greater number of dose failures. At this
off-state after an overnight withdrawal of PD medica- stage many patients seem to have an all-or-none
tions. Studies have shown that patients taking Sinemet response to levodopa that requires a threshold level
CR have a better response to the second dose com- of dopaminergic stimulation to obtain (see section
pared to the first morning dose, suggesting there is a 39.5.1). Frequent small doses may undershoot this
delay in absorption in the off-state (Yeh et al., 1989). threshold. In such patients with very complicated,
One explanation for these observations has been that unpredictable fluctuations, simplifying the dosage
PD patients with fluctuations have an increase in gas- schedule using fewer but larger doses will often result
tric emptying in the on-state but decreased gastric in a return to a more predictable response pattern.
emptying in the off-state (Hardoff et al., 2001). The
authors suggest that taking the next dose of levodopa 39.6.3.2. Use liquid/soluble preparations
while still partly on may reduce dose failures as the The more rapidly absorbed, fast-acting soluble levo-
increased gastric emptying will improve absorption dopa preparations may be helpful for rapid reversal
via the small intestine. of off-periods (see above).
MOTOR AND NON-MOTOR FLUCTUATIONS 167
39.6.3.3. Modify dietary protein dyskinesia also results in changes within neurotransmit-
Onoff fluctuations have been suggested to be second- ter signaling pathways in the basal ganglia circuitry
ary to impaired absorption of levodopa across the gut (Chase, 1998b; Hadj Tahar et al., 2000; Bezard et al.,
and bloodbrain barrier due to competition with large 2001). In effect this results in a depriming of the
neutral amino acids found in dietary protein (Nutt system with a reduction in motor fluctuations and dys-
et al., 1984; Leenders et al., 1986; Alexander et al., kinesia. In clinical studies, continuous dopaminergic
1994). As such, either reducing protein intake or leav- stimulation using infusions of levodopa (Sage et al.,
ing large protein meals to the end of the day may 1988; Kurth et al., 1993b; Nutt et al., 2000) or lisuride
reduce onoff fluctuations (Pincus and Barry, 1987; (Stocchi et al., 2002) can also reduce established on
Karstaedt and Pincus, 1992). This effect is probably off fluctuations and dyskinesia. However, in terms of
not at the level of the gut as the neutral amino acid ease of use and tolerability, infusion of apomorphine
transporter capacity is unlikely to become saturated subcutaneously is a better option. Unfortunately, this
and there is no difference in plasma levodopa kinetics depriming is short-lived and patients who have to stop
following administration of levodopa after a low- the infusions and return to intermittent doses of oral
protein meal compared to a normal-protein meal in medication often experience a rapid return to their
advanced PD patients (Carter et al., 1989; Simon motor complications. Thus such patients may need to
et al., 2004). be considered for surgery to treat motor fluctuations
(see later).
The main side-effect of apomorphine is formation
39.6.3.4. Add apomorphine of skin nodules. These can occur in 2078% of
The injectable mixed dopamine D1/D2-receptor ago- patients (Colzi et al., 1998; Tyne et al., 2004) but are
nist, apomorphine, has been used in Europe for many only bothersome in about a third of these patients
years as a treatment for onoff motor fluctuations (Manson et al., 2002). Management includes rotating
(Stibe et al., 1988; Stocchi et al., 2001b) and has the injection site, ensuring strict aseptic technique;
recently been approved for use in the USA. Due to diluting the apomorphine 1:1 with normal saline,
the proemetic action of apomorphine, prior treatment massage or local skin ultrasound. Neuropsychiatric
for 23 days and continued treatment using the oral side-effects appear to be less common than with oral
antiemetics domperidone (20 mg t.i.d.) or trimethoben- dopamine agonists but hallucinations and confusion
zamide hydrochloride (300 mg t.i.d.) is required. Some can occur at high doses. In a small proportion of
patients can discontinue the antiemetic after a few patients with PD, addictive behaviors develop and
weeks. include excessive use of their dopaminergic medica-
A single subcutaneous injection of apomorphine tions (see later). Escalating doses of apomorphine
alleviates parkinsonian symptoms within 515 minutes and levodopa in such patients may result in major
for 6090 minutes and so can be used as a rescue behavioral and psychiatric side-effects such as hyper-
for off-period disability (Chaudhuri et al., 1988; Frankel sexuality and gambling, termed hedonistic homeo-
et al., 1990; Dewey et al., 2001). Apomorphine can be static dysregulation (Giovannoni et al., 2000).
used either as intermittent subcutaneous injections Management of this may require hospitalization and
(28 mg per injection, average daily total dose approxi- an aggressive reduction in medication doses. On the
mately100 mg/day) and/or as an infusion (20160 mg) other hand, in some cases, a reduction in neuropsy-
ranging over 1024 hours (Manson et al., 2002; Tyne chiatric problems, such as depression, occurs with apo-
et al., 2004). The antiparkinsonian actions of apomor- morphine (Manson et al., 2002).
phine and levodopa are equivalent (Cotzias et al., New methods of administering apomorphine are
1970). Long-term use of apomorphine does not result being developed in an attempt to reduce the problems
in loss of benefit (Hughes et al., 1993; Gancher et al., associated with injections. To date, sublingual (Montas-
1995). For patients with frequent off-periods, infusion truc et al., 1991), intranasal (Kapoor et al., 1990) and
of apomorphine is useful. This is more practical when rectal administration (Hughes et al., 1991) have all been
patients are requiring several injections per day. tried but with suboptimal absorption and tolerability.
Continuous infusion of apomorphine not only A recent epicutaneous transdermal (ApoMTD) route
markedly reduces off-time but also reduces on-period has been developed and a preliminary study has shown
dyskinesia. This reduction in dyskinesia is partly due improved off-periods with mild side-effects, including
to a concomitant reduction in levodopa dose (Colzi some transient skin erythema (Priano et al., 2004). In
et al., 1998; Manson et al., 2002). However, continuous all cases, however, the rate of absorption of apomor-
dopaminergic stimulation of striatal dopamine receptors phine is much slower than via an injection and as such
in animal models of established motor fluctuations and limits use as a rescue therapy for off-periods.
168 S. H. FOX AND A. E. LANG
Despite the obvious advantages, there has been a in dopaminergic medication (Kumar et al., 2000; Loher
general underusage of apomorphine (Chaudhuri and et al., 2002; Volkmann et al., 2004). Although there
Clough, 1998). In many countries, this has been have been no large RCTs comparing STN to GPi
because of a lack of a peripheral dopamine receptor DBS, there is a general belief that the benefit obtained
antagonist such as domperidone. Another common with STN DBS is on the whole greater and more sus-
reason also relates to the practicalities of using apo- tained than that obtained with GPi DBS (Deep-Brain
morphine as most patients will need support from Stimulation for Parkinsons Disease Study Group,
family and specialist PD nurses (Manson et al., 2002; 2001; Boucai et al., 2004; Peppe et al., 2004).
Tyne et al., 2004). The efficacy of bilateral subthala-
mic nucleus deep brain stimulation (STN DBS) for 39.6.4. Treatment of sudden transient
motor fluctuations has resulted in this becoming the motor blocks
preferred option, particularly since doses of dopami-
nergic medication can often be substantially reduced PD patients with off-period freezing may respond to
(Moro et al.,1999), with resultant reduced risk of, or various approaches designed to reduce the off-periods,
an improvement in, other adverse side-effects (see as outlined above. However, freezing of gait is often
later). However, in elderly patients where the risks of more resistant to increasing dopaminergic stimulation
surgery are thought too great, apomorphine infusion compared to other parkinsonian symptoms. In addi-
for treatment of motor fluctuations is a viable option. tion, freezing can occur during on-periods (on-period
To date, no direct comparison of apomorphine infusion freezing) and this can be extremely difficult to treat.
with bilateral STN DBS has been made. At times this on-period freezing worsens in response
to increasing the dose of dopaminergic drugs. Bilateral
STN DBS may have no effect on on-period freezing
39.6.3.5. Bilateral Subthalamic Nucleus Deep Brain
(Stolze et al., 2001). Reductions in dopaminergic
Stimulation
agents may help the problem but can also result in an
In patients with advanced PD, bilateral STN DBS is cur- increase in other parkinsonian symptoms. Such
rently the most effective treatment for all motor fluctua- patients may respond by developing tricks that utilize
tions (Limousin et al., 1998; Kumar et al., 1998; a variety of sensory cues, such as having to step over
Lagrange et al., 2002; see Ch. 43). In particular, predict- an object, whether the foot of another person, lines
able and unpredictable onoff motor fluctuations, drawn on the floor, a specifically designed cane
as measured by the UPDRS part IV subscale, are signif- or walker with either a laser or modified lower part
icantly reduced by about 50% and on-period dyskinesia of the cane at right angles, or other cues such as play-
is reduced by > 50 % (Kumar et al., 1998; Limousin ing music, marching in time or counting that can then
et al., 1998; Moro et al., 1999). As discussed in the next trigger a movement (Quinn, 1998; Kompoliti et al.,
section, due to the profound reduction in off-periods, 2000).
patients may also experience significant benefit in other
non-motor off-period symptoms, such as pain and auto-
nomic symptoms. Long-term follow-up to 5 years has
39.7. Non-motor fluctuations
demonstrated that patients continue to have fewer motor 39.7.1. Definitions
fluctuations (Krack et al., 2003). In most patients, there
is a reduction in dopaminergic medication of about In addition to fluctuations in the motor symptoms of
50% and in rare instances no dopaminergic drugs are parkinsonism, patients can also experience fluctuations
required after surgery (Kumar et al., 1998; Limousin in non-motor symptoms, which may be as debilitating,
et al., 1998; Moro et al., 1999; Molinuevo et al., 2000). or more so, than immobility and akinesia (Lang and
This in part accounts for the reduction in on-period dys- Lozano, 1998). These fluctuations have been classed
kinesia. However, even when challenged with the same into three groups of symptoms: neuropsychiatric, auto-
preoperative dose of levodopa, patients will have signif- nomic and sensory/pain (Riley and Lang, 1993; Hillen
icantly less levodopa-induced dyskinesia, thus suggest- and Sage, 1996) (Table 39.2). These non-motor symp-
ing that continuous STN DBS has a depriming effect toms appear to follow a similar pattern to the wearing-
on the basal ganglia circuitry in a similar fashion to con- off phenomena seen in parkinsonian symptoms and
tinuous dopaminergic stimulation (Bejjani et al., 2000; may appear during wearing-off and off-periods and
Moro et al., 2002; Varma et al., 2003). improve after taking levodopa or dopaminergic drugs.
Bilateral internal globus pallidus (GPi) DBS is also In addition, some symptoms may be associated with
effective at improving off-periods and especially in les- elevated levels of levodopa and so appear during the
sening dyskinesia, even without a concurrent reduction on-periods.
MOTOR AND NON-MOTOR FLUCTUATIONS 169
Table 39.2 awareness of the non-motor complications and fluctua-
tions experienced by PD patients.
Non-motor fluctuations

1. Neuropsychiatric 39.7.3. Neuropsychiatric fluctuations


(a) Mood 39.7.3.1. Mood fluctuations
Anxiety, depression, irritability, panic attacks, screaming
Apathy 39.7.3.1.1. Anxiety/depression
Fatigue PD patients commonly experience fluctuations in mood,
(b) Psychotic symptoms generally with worsening in the off-periods and improve-
Euphoria, agitation, dopamine dysregulation syndrome
ment in the on-periods (Brown et al., 1984; Cantello et al.,
Visual hallucinations, delusion, paranoia
1986; Nissenbaum et al., 1987; Menza et al., 1990). The
(c) Cognitive functions
2. Autonomic commonest symptoms experienced are off-period anxi-
(a) Thermoregulation: sweating, facial flushing, pallor, ety, irritability and depression (Menza et al., 1990; Riley
hyperthermia and Lang, 1993; Siemers et al., 1993; Witjas et al.,
(b) Sphincter function: urinary disturbances, bloating, 2002). PD patients can also have panic attacks (Vazquez
abdominal discomfort, constipation et al., 1993) and off-period moaning and screaming
(c) Dysphagia and drooling of saliva; dry mouth episodes have been reported (Steiger et al., 1991).
(d) Orthostatic hypotension, tachycardia
(e) Dyspnea 39.7.3.1.2. Apathy
(f) Peripheral edema
3. Sensory Apathy is a distinct neurobehavioral syndrome experi-
(a) Pain enced by PD patients that consists of reduced interest
(b) Numbness, paresthesia and motivation, behavioral changes with reduced
(c) Restless-legs syndrome; akathisia initiative and decreased spontaneous action and emo-
tional changes with a flattened affect (Pluck and
Brown, 2002). PD patients may experience fluctua-
tions in apathy with worsening in off-periods (Witjas
et al., 2002). Apathy is thought to involve limbic basal
39.7.2. Epidemiology
ganglia anterior cingulate cortical loops (Isella et al.,
2002). Patients who have undergone STN DBS and
The epidemiology of these non-motor fluctuations is not
have a reduction in their levodopa dose are reported
well defined. On the current UPDRS scale, Activities of
to develop apathy, suggesting a dopaminergic etiology
Daily Living part II subscale, only three questions speci-
(Funkiewiez et al., 2004).
fically address non-motor symptoms related to on- and
off-periods: sensory symptoms, swallowing and drooling
39.7.3.1.3. Fatigue
saliva. Attempts have been made to determine the extent
of non-motor symptoms with questionnaires (Riley and Fatigue is a common symptom in PD patients, and is
Lang, 1993; Hillen and Sage, 1996; Gunal et al., 2002; defined according to the patients own perceived state
Witjas et al., 2002). These studies have shown that of lack of energy (Krupp and Pollina, 1996; Herlofson
fluctuations in neuropsychiatric symptoms are probably and Larsen, 2002). The cause is unclear but fatigue is
the most common, with mood fluctuations, especially usually distinct from depression or sleepiness, not asso-
off-period anxiety, occurring in up to 75% of patients ciated with disease severity (Friedman and Friedman,
(Quinn, 1998; Witjas et al., 2002). PD patients with 1993; van Hilten et al., 1993; Alves et al., 2004) and
non-motor fluctuations usually experience more than is probably an independent symptom of PD (Herlofson
one symptom and often have multiple symptoms and Larsen, 2002). Single-photon emission computed
(Shulman et al., 2001). However, the exact incidence tomography (SPECT) studies have suggested that PD
and prevalence are not known. patients with fatigue have impaired frontal-lobe perfu-
To try and address this problem, the new UPDRS sion (Abe et al., 2000). In questionnaire studies, the fre-
rating scale currently under development has incorpo- quency of fatigue in PD patients is over 50% and
rated more non-motor aspects, such as apathy, sleep fluctuates through the day, and is predominantly worse
patterns and various autonomic functions (Movement in off-periods (Witjas et al., 2002; Zenzola et al., 2003).
Disorder Society Task Force, 2003). New non-motor
questionnaires are also being developed (Gulati et al., 39.7.3.1.4. Pathophysiology
2004; Brown et al., 2005; Stacy et al., 2005). These tools As outlined in the sections above, the origin of mood
require clinimetric testing but represent an increased symptomatology varies and includes frontal, basal
170 S. H. FOX AND A. E. LANG
ganglia and limbic circuits as well as brainstem bioa- et al., 2003). These patients consume much larger
mine nuclei and their projections. The pathophysiol- doses of levodopa and other dopaminergic drugs than
ogy underlying fluctuations of these mood disorders they require to maintain a good on-state, and can
is unclear. Improvements in mood have been demon- experience a range of symptoms that occur with on-
strated to be proportional to levodopa dose, suggesting periods, including hypersexuality, pathological gam-
a relationship to brain dopamine concentration (Maricle bling and shopping, aggression and compulsive eating
et al., 1995). In addition, the improvements in mood (Molina et al., 2000; Lawrence et al., 2003; Kurlan,
with infusion of levodopa precede the improvement in 2004). In addition to or separate from the dopamine
motor function, suggesting that patients do not simply dysregulation syndrome, patients may demonstrate
feel better because they are more mobile (Maricle stereotypical behavior termed punding. Here patients
et al., 1995). Mood fluctuations have also been shown will perform the same, meaningless, seemingly purpo-
to be associated with a longer disease duration and seless task over and over again (Friedman, 1994;
younger age at onset, as well as with the presence and Evans et al., 2004). These behaviors, usually related
severity of motor fluctuations (Cantello et al., 1986; to a prior trade or hobby, occur at the expense of sleep
Vazquez et al., 1993; Racette et al., 2002; Witjas and patients may spend hours with a single purposeless
et al., 2002; Richard et al., 2004), thus the underlying activity followed by severe exhaustion and fatigue.
pathophysiology may be similar to that causing fluc-
tuations in parkinsonian symptoms. However, there 39.7.3.2.2. Visual hallucinations, delusions, paranoia
is not always a consistent relationship between motor Psychotic symptoms of illusions, well-formed visual
and mood fluctuations (Richard et al., 2004). In addi- hallucinations, paranoia and delusions are often
tion it is not entirely clear whether fluctuations in reported in PD and are associated with disease severity,
mood are related to an underlying persistent, comor- age, depression and cognitive impairment (Goetz and
bid mood disorder. The limited conflicting available Stebbins, 1993; Factor et al., 2003). Visual hallucinations
evidence derives from case reports or small series will fluctuate through the day and are most commonly
(Menza et al., 1990; Racette et al., 2002; Richard reported in the evening and overnight (Sanchez-Ramos
et al., 2004). et al., 1996; Fenelon et al., 2000). In questionnaire studies
of prevalence of non-motor fluctuations, patients have
39.7.3.1.5. Management reported hallucinations specifically related to on-periods
Mood deterioration, which is a wearing-off or off- (Witjas et al., 2002) but others experience them specifi-
period phenomenon, may respond to treatment of the cally related to off-periods (Nissenbaum et al., 1987;
off-periods. There have been no clinical trials formally Witjas et al., 2002).
assessing treatment of fluctuations in anxiety or depres-
sion. Patients with persistent anxiety and depression
39.7.3.2.3. Pathophysiology
despite reduced off-periods may benefit from specific
anxiolytic or antidepressant therapy. However, the The underlying pathophysiology of psychotic symp-
influence of this treatment on off-period-related exacer- toms in PD may relate to alterations in neurotransmit-
bations of mood is unknown. ter pathways, particularly involving mesolimbic
dopamine and serotonin (Meltzer, 1992; Breier, 1995).
39.7.3.2. Fluctuations in psychotic symptoms Pathological studies have suggested that the inferotem-
poral cortex may be implicated as Lewy bodies are
39.7.3.2.1. Euphoria, agitation, dopamine dysregula- increased in the amygdala and parahippocampus of
tion syndrome the temporal lobe in PD patients with hallucinations
In the on-periods, PD patients can experience an eleva- compared to those without (Harding et al., 2002).
tion in mood that may be associated with alertness and The cause of fluctuations in psychotic symptoms, how-
euphoria (Keshavan et al., 1986; Nissenbaum et al., ever, is unclear, although it may relate to changes in
1987; Lees, 1989). Some patients report a feeling of mesolimbic dopaminergic stimulation as all dopami-
euphoria that just precedes the motor on-state (Witjas nergic agents can cause psychosis and reducing or dis-
et al., 2002). An extreme form of mood elevation asso- continuing dopaminergic medication can resolve the
ciated with on-periods may result in psychomotor agi- symptoms (Friedman and Sienkiewicz, 1991; Poewe,
tation and hyperactivity, increased excitability and 2003). However, psychotic symptoms such as halluci-
even a hypomanic or true manic state. This has been nations are not always related to the level of dopami-
described in a small number of patients, predominantly nergic stimulation, since infusion of levodopa in
male, and has been termed the dopamine dysregula- hallucinating patients did not trigger hallucinations in
tion syndrome (Giovanni et al., 2000; Lawrence one study (Goetz et al., 1998) and several reports have
MOTOR AND NON-MOTOR FLUCTUATIONS 171
found no relationship between either type or dose of phenomenon (Brown et al., 1984; Poewe et al., 1991;
dopaminergic drug and psychosis (Sanchez-Ramos Green et al., 2002). Other patients report slowing of
et al., 1996; Graham et al., 1997; Fenelon et al., thoughts predominantly in the off-periods (Matison
2000; Merims et al., 2004). Disturbances in the et al., 1982; Witjas et al., 2002). Indeed, in our experi-
sleepwake cycle are often found in patients with psy- ence, patients complain of cognitive slowing and
chotic symptoms and may account for fluctuations in difficulty concentrating more often related to the off-
these symptoms. Thus, patients with psychotic symp- periods. Some patients show reduction in selective areas
toms have more daytime somnolence and have vivid of cognitive function in off-periods such as delayed
dreams or nightmares (Moskovitz et al., 1978; Nau- recall of complex verbal material (Mohr et al., 1989)
sieda et al., 1982; Pappert et al., 1999; Fenelon et al., and verbal fluency (Gotham et al., 1988). However, this
2000), although the exact nature of this relationship has been suggested to be more related to a reduction in
is not entirely clear (Goetz et al., 2005). In addition, attention/arousal than a true cognitive effect (Brown
there is a disturbance in rapid-eye movement (REM) et al., 1984).
sleep pattern in PD patients with hallucinations
(Comella et al., 1993; Arnulf et al., 2000). Indeed, in 39.7.3.3.1. Pathophysiology
some patients hallucinations may relate to intrusions
Patients with fluctuating cognition and no dementia
of REM sleep into wakefulness comparable to hypna-
often have dysfunctions in frontal lobe function, sug-
gogic hallucinations in narcolepsy. Thus pathology
gesting impaired basal gangliaprefrontal cortical
in brainstem structures controlling sleep may also
loops (Gotham et al., 1988; Green et al., 2002). The
cause fluctuating hallucinations in PD (Manford and exact circuitry involved or underlying mechanisms,
Andermann, 1998).
however, remain unclear. Memory function in PD
has been shown to correlate with changes in dopamine
39.7.3.2.4. Management levels, rather than with a constant high or low level
On-period elevations in mood and psychotic symptoms (Huber et al., 1987, 1989), suggesting that fluctuations
may resolve with reduction in levodopa or dopamine in cognitive function may relate to levodopa dosing.
agonist dose. Some studies have suggested that dopa-
mine agonists are more likely to be associated with 39.7.4. Autonomic symptoms
psychotic symptoms than levodopa (Saint Cyr et al.,
1993), but the relationship to dopaminergic stimulation Patients with PD develop a variety of symptoms of
is not entirely clear (see above). In the case of the dysautonomia as part of the underlying disease process
dopamine dysregulation syndrome, patients benefit (Koike and Takahashi, 1997; see Ch. 14). These com-
from a reduction and rationing of levodopa and dopa- monly include sweating and thermoregulatory dys-
mine agonists (Lawrence et al., 2003; Evans et al., function, sphincter disturbances, abdominal bloating
2004). In general, psychotic symptoms usually require and discomfort, drooling, dry mouth, orthostatic hypo-
specific treatment with atypical neuroleptics such as tension, peripheral edema and a range of other rarer
quetiapine or clozapine. Other patients may have coex- symptoms. Many patients report fluctuations in these
istent depression and sleep disturbances which will symptoms predominantly occurring with off-periods
require specific treatment (Lawrence et al., 2003; (Riley and Lang, 1993; Hillen and Sage, 1996). How-
Evans et al., 2004). ever, in other studies, many autonomic symptoms were
independent of on- or off-periods (Gunal et al., 2002;
39.7.3.3. Fluctuations in cognitive function Witjas et al., 2002). Patients with fluctuations in auto-
nomic symptoms usually have a collection of symp-
Cognitive problems ranging from mild cognitive
toms that occur together consistently, usually in the
impairment to dementia are common in advanced PD
off-periods (Raudino, 2001; Swinn et al., 2003).
(Aarsland et al., 1996; see Ch. 18). Patients with
dementia with Lewy bodies often demonstrate quite
prominent fluctuations in cognition and alertness 39.7.4.1. Disturbances of thermoregulation
(McKeith et al., 1996; see Ch. 60). However, fluctua- One of the commonest types of non-motor fluctuation
tions in cognition can also occur in PD patients with- reported by patients is off-period sweats, which can be
out dementia (Delis et al., 1982; Girotti et al., 1986; sudden and drenching, sometimes referred to as
Cooper et al., 1991; Meco et al., 1991). Thus some sweating crises (Sage and Mark, 1995; Raudino,
patients report on-period slowing of thoughts or brady- 2001; Gunal et al., 2002). In one study, 64% of patients
phrenia and difficulty or delay in memory retrieval, experienced sweating in the off-period, whereas 16%
sometimes referred to as the tip of the tongue reported symptoms when on with dyskinesia (Witjas
172 S. H. FOX AND A. E. LANG
et al., 2002). In a cohort of PD patients all reporting 2002). Alternatively some patients report a dry mouth
sweating, 35% had sweating episodes in the off-period, during off-periods (Witjas et al., 2002), possibly
18% when on with dyskinesia but 39% apparently unre- related to breathing through a persistently open mouth.
lated to their motor state (Swinn et al., 2003). Patients
also often complain of facial flushing or a sensation of 39.7.4.4. Orthostatic hypotension
feeling hot occurring during the off-periods (Hillen PD patients can experience a symptomatic postural fall
and Sage, 1996; Gunal et al., 2002; Witjas et al., in blood pressure (BP) which may cause a range of
2002). During formal autonomic function testing in symptoms, including dizziness, fainting, fatigue, slee-
the off-state, PD patients have increased sweating and piness, shoulder pain or falls (Senard et al., 1997;
lower skin temperatures to a heat stimulus which Mathias and Kimber, 1998). Generally this is second-
reverses with levodopa therapy (Goetz et al., 1986a). ary to pathological involvement of the sympathetic
There have been rare reports of severe hyperthermia nervous system but also occurs de novo or is aggra-
induced by off-periods in patients with motor fluctua- vated by dopaminergic medication since all such
tions, similar to the neuroleptic malignant syndrome-like agents are capable of lowering BP (Wakabayashi
state that may occur in response to sudden withdrawal et al., 1993; Korchounov et al., 2004). However, the
of levodopa (Pfeiffer and Sucha, 1989; Keyser and presence of motor fluctuations appears to be an addi-
Rodnitzky, 1991). tional independent risk factor for fluctuations in BP
control. In one study, PD patients with wearing-off
39.7.4.2. Disturbance of sphincter function fluctuations compared to a group without motor fluc-
Off-period urinary urgency is a common problem (Riley tuations had significantly higher supine and erect
and Lang, 1993; Raudino, 2001; Gunal et al., 2002). BP during the off-phase as compared to the on-phase
Urodynamic studies in the on- and off-state have shown (Baratti and Calzetti, 1984).
that apomorphine will improve voiding by decreasing
bladder outflow resistance (Christmas et al., 1988), 39.7.4.5. Dyspnea
whereas levodopa gives inconsistent results (Fitzmaurice Dyspnea occurring as an off-period phenomenon has
et al., 1985; Uchiyama et al., 2003; Winge et al., 2004). been reported in PD patients with fluctuations (Hillen
Another sphincter problem occurring predominantly in and Sage, 1996; Raudino. 2001; Gunal et al., 2002;
the off-period is constipation (Witjas et al., 2002), rarely Witjas et al., 2002) and may be accompanied by a
to the extent of anismus. Apomorphine has been demon- bothersome, subjective sense of air hunger. Alterna-
strated to relieve paradoxical striated muscle contraction tively, some patients may demonstrate tachypnea with-
during defecation (Mathers et al., 1989). Some patients out much in the way of accompanying complaints,
report fluctuating abdominal discomfort and bloating although the care-giver may express concerns about
due to suppression of peristalsis that occurs during off- the symptoms. These symptoms may occur due to
periods (Hillen and Sage, 1996; Raudino, 2001; Gunal upper-airway obstruction (Vincken et al., 1984). How-
et al., 2002; Witjas et al., 2002). ever, a study by Weiner et al. (2002) has shown that
patients perception of dyspnea is improved by levo-
39.7.4.3. Dysphagia and drooling of saliva dopa without any objective change in pulmonary func-
Swallowing difficulties with choking on food can tion. In addition, off-period dystonia of the cervical or
occur in PD in the off-periods (Bushmann et al., laryngeal musculature can also induce stridor (Corbin
1989; Witjas et al., 2002). Some patients experience and Williams, 1987). Breathing difficulties can also
an improvement in symptoms with levodopa, although occur as an on-period problem and may be a form of
it is not consistent. Barium studies before and after respiratory dyskinesia (De Keyser and Vincken, 1985;
administration of levodopa have demonstrated that Jankovic and Nour, 1986).
about 50% of PD patients experience improved oro-
pharyngeal or esophageal function (Bushmann et al., 39.7.4.6. Pathophysiology
1989). Off-period belching secondary to esophageal Dysautonomic features in PD are related to pathologi-
motility dysfunction, relieved by apomorphine, has cal involvement of the peripheral and central sympa-
also been reported (Kempster et al., 1989). PD patients thetic and parasympathetic nervous systems that
often report drooling of saliva secondary to dysphagia, occurs with advancing disease (Edwards et al., 1992;
which can range from mild to severe with a constantly Wakabayashi et al., 1993). Fluctuations in autonomic
wet face and clothes (Bateson et al., 1973). Patients function have been shown to correlate with both dis-
often report fluctuations in salivation with worsening ease duration and severity of disease (Gunal et al.,
in the off-periods (Raudino, 2001; Gunal et al., 2002; Witjas et al., 2002). Patients with fluctuating
MOTOR AND NON-MOTOR FLUCTUATIONS 173
autonomic symptoms tend to have higher daily levo- Witjas et al., 2002). Fluctuations in pain are due to a
dopa doses compared to non-fluctuators (Gunal et al., number of factors. One important cause is the presence
2002). Fluctuations in these autonomic functions may of dystonia and rigidity associated with off-periods.
relate to fluctuating peripheral dopaminergic stimula- Pain associated with dystonia most commonly occurs
tion (Goetz et al., 1986a). Dopamine D1- and D2- accompanying early-morning dystonia, typically
receptor agonists can reduce the bladder threshold for affecting the feet and toes, and improving when the
the micturition reflex in the MPTP-lesioned primate patient switches on (Melamed, 1979; Currie et al.,
(Yoshimura et al., 1993, 1998), suggesting improved 1998). Painful dystonic spasms may also occur as an
bladder function in the on-state. Dopamine plays a role end-of-dose or off-period pattern (Ilson et al., 1984)
in the regulation of BP by inhibition of sodium trans- or in a diphasic pattern (Luquin et al., 1992).
port in renal proximal tubules and relaxation of vascu- Patients also report painful sensations without evi-
lar smooth muscles (Velasco and Luchsinger, 1998), dence of dystonia, most often associated with off-
thus accounting for fluctuating orthostatic and cardio- periods (Quinn et al., 1986). Such pain can be severe
vascular symptoms. However, some symptoms, such and have a burning, aching or stabbing quality. Often
as sweating, are not related to disease severity or dura- the symptoms are diffuse and poorly localized, but
tion or to levodopa dose. In these cases the pathophy- may be more severe on the more affected parkinsonian
siology is less clear but may relate to disease side. Non-specific oral and genital pain related to off-
involvement of more central autonomic stuctures such periods has also been reported, in all cases responding
as the hypothalamus (Langston and Forno, 1978; to treatment with levodopa (Ford et al., 1996). On-
Braak et al., 2004). period pain is also a recognized problem and may be
secondary to peak-dose dyskinesia; however, it has
39.7.4.7. Management not been well characterized (Goetz et al., 1986b;
Quinn, 1998). In some patients, this may simply relate
In general, off-period autonomic dysfunction will
to a combination of a musculoskeletal cause (e.g.
respond to treatment of the off-periods (Sage and
arthritis) with prominent choreoathetoid involuntary
Mark, 1995; Olanow and Koller, 1998). Urinary
movements.
dysfunction related to a hyperactive bladder may be
helped with anticholinergic medications such as
oxybutinin or tolteradine. Bilateral STN DBS is also 39.7.5.2. Numbness/dysesthesia
effective at reducing urinary symptoms due to detrusor PD patients with motor fluctuations can also experi-
hyperreflexia (Seif et al., 2004). Sweating episodes ence fluctuating sensory symptoms without objective
may respond to beta-blockers (Stocchi et al., 1997; sensory loss. These symptoms include numbness, cold-
Olanow and Koller, 1998). Sialorrhea may respond to ness, tightening or paresthesia that is most often asso-
botulinum toxin injections into the salivary glands ciated with off-periods (Snider et al., 1976; Koller,
(Mancini et al., 2003; Ondo et al., 2004). 1984). Patients are reported to experience symptoms
more in the legs than the arms, with the neck and face
39.7.5. Sensory being rarely affected (Snider et al., 1976).

PD patients can experience a number of fluctuating 39.7.5.3. Restless-legs syndrome/akathisia


symptoms that have been loosely termed sensory to Restlessness is a common complaint among patients
distinguish them from symptoms that may occur due with PD; however, it may be difficult to distinguish
to the motor features of parkinsonism. Such symptoms restlessness due to leg pain or discomfort as a wearing-
may include pain, numbness and paresthesias, akathisia off phenomenon from true restless-legs syndrome and
and restless-legs syndrome. These symptoms can fluctu- akathisia (Poewe and Hogl, 2004). Fluctuations in symp-
ate and are either most often experienced exclusively toms of restlessness, with a predominantly off-period
during off-periods or are accentuated during these distribution, have been reported in a number of case
times. Such symptoms are often the most distressing series; however, the term was often not clearly defined
to patients and in some cases become more disabling (Gunal et al., 2002; Witjas et al., 2002).
and bothersome than the motor fluctuations. Akathisia is a sensation of inner restlessness and
inability to sit still that has been reported to occur in
39.7.5.1. Pain 26% of PD patients, when underlying factors such as
Pain is a common symptom in PD and fluctuations in discomfort due to parkinsonism-related symptoms are
pain are reported in 2346% of PD patients (Goetz excluded (Lang and Johnson, 1987). Fluctuations in
et al., 1986b; Quinn et al., 1986; Gunal et al., 2002; akathisia are common among PD patients with motor
174 S. H. FOX AND A. E. LANG
fluctuations (Witjas et al., 2002). The relationship to Factor et al., 2000). Severe nocturnal pain may be
the motor state is not consistent: some patients report helped by apomorphine infusion (Reuter et al., 1999).
akathisia related to off-periods but also when switch- Bilateral STN DBS and pallidotomy are also an effec-
ing on or in an end-of-dose pattern (Lang and Johnson, tive therapy for fluctuating off-period sensory symp-
1987; Comella and Goetz, 1994; Witjas et al., 2002). toms (Honey et al., 1999; Krack et al., 1999; Loher
et al., 2002). Other treatment options for off-period
39.7.5.4. Pathophysiology dystonia include specifically targeted botulinum toxin
injections ((Pacchetti et al., 1995), baclofen (Lees
Fluctuating pain and other sensory symptoms in PD
et al., 1978) and lithium (Quinn and Marsden, 1986).
may be secondary to peripheral mechanisms asso-
On-period pain and sensory symptoms may respond
ciated with sustained muscle contraction in dystonia,
to a reduction in dopaminergic mediation. Sensory
rigidity or musculoskeletal problems as well as to
symptoms unrelated to dystonia and unresponsive to
primary central mechanisms, the nature of which
manipulations in dopaminergic therapy are often
remains unclear. Off-period fluctuating pain and other
intractable and difficult to treat. There are no published
sensory symptoms are often dramatically alleviated by
clinical trials to date and thus treatment options are
dopaminergic medication, suggesting that the patho-
based on case reports and include opiates (Stein and
physiology is associated with the neural mechanisms
Read, 1997) and clozapine (Factor and Freidman,
generating the underlying parkinsonian symptoms
1997; Trosch et al., 1998). It should be noted that,
and dystonic symptoms. In addition, pain, which may
although bilateral STN DBS is extremely effective at
or may not be associated with dystonia, can be a pre-
treating motor fluctuations in PD and thus reducing
senting feature of untreated PD (LeWitt et al., 1986).
many off-period-related non-motor symptoms (Funkie-
The basal ganglia are involved in pain processing,
wiez et al., 2004), the concomitant reduction in dopami-
via the ventral anterior and ventrolateral thalamic
nergic medications can often induce or unmask new
nuclei (Chudler and Dong, 1995; Tracey et al.,
non-motor symptoms that were not present prior to the
2000). The pathways involved in primary parkinsonian
surgery, such as restless-legs syndrome (Kedia et al.,
pain are unknown; however a PET study has shown
2004). The short- and long-term influence of bilateral
that increased pain perception correlates with lower
STN DBS on the broad spectrum of non-motor
numbers of striatal dopamine D2-receptors (Hagelberg
fluctuations outlined above is at present unknown.
et al., 2002), suggesting a link between the dopaminer-
gic system and pain processing. Spinal cord dopami-
References
nergic pathways have also been suggested to play a
role in fluctuating pain, since spinal anesthesia but
Aarsland D, Tandberg E, Larsen JP et al. (1996). Frequency of
not sympathetic or epidural block effectively alle- dementia in Parkinson disease. Arch Neurol 53: 538542.
viated diphasic leg pain in one PD patient (Sage Abe K, Takanashi M, Yanagihara T (2000). Fatigue in
et al., 1990). More recently it has been demonstrated patients with Parkinsons disease. Behav Neurol 12:
that, in PD, there is increased sensitivity to some pain- 103106.
ful stimuli such as heat threshold; however, this did Ahlskog JE, Muenter MD (2001). Frequency of levodopa-
not change between the on- and off-state in PD related dyskinesias and motor fluctuations as estimated
patients with motor fluctuations (Djaldetti et al., from the cumulative literature. Mov Disord 16: 448458.
2004). This suggests that some painful sensory symp- Ahlskog JE, Muenter MD, McManis PG et al. (1988). Con-
toms may be secondary to involvement of non-dopa- trolled-release Sinemet (CR-4): a double-blind crossover
minergic systems such as noradrenergic structures study in patients with fluctuating Parkinsons disease.
Mayo Clin Proc 63: 876886.
(Buzas and Max, 2004).
Alexander GM, Schwartzman RJ, Grothusen JR et al. (1994).
Effect of plasma levels of large neutral amino acids and
39.7.5.5. Management degree of parkinsonism on the blood-to-brain transport of
Sensory symptoms clearly related to off-periods, levodopa in naive and MPTP parkinsonian monkeys. Neu-
or low levels of levodopa as in diphasic dyskinesia, rology 44: 14911499.
Alves G, Wentzel-Larsen T, Larsen JP (2004). Is fatigue an
will improve with increased dopaminergic medication
independent and persistent symptom in patients with Par-
and treatment of onoff fluctuations (Sage, 2004).
kinson disease? Neurology 63: 19081911.
Early-morning off-period dystonia will respond to the Andersson M, Hilbertson A, Cenci MA (1999). Striatal fosB
use of long-acting dopamine agonists or controlled- expression is causally linked with l-DOPA-induced abnor-
release levodopa prior to bedtime (Lees, 1987). Rescue mal involuntary movements and the associated upregula-
medication for severe off-period pain includes soluble tion of striatal prodynorphin mRNA in a rat model of
levodopa preparations or apomorphine (Lees, 1993; Parkinsons disease. Neurobiol Dis 6: 461474.
MOTOR AND NON-MOTOR FLUCTUATIONS 175

Arnulf I, Bonnet AM, Damier P et al. (2000). Hallucinations, fluctuations in motor function in Parkinsons disease.
REM sleep, and Parkinsons disease: a medical hypoth- J Neurol Neurosurg Psychiatry 47: 454465.
esis. Neurology 55: 281288. Brown RG, Dittner A, Findley L et al. (2005). The Parkinson
Assal F, Spahr L, Hadengue A et al. (1998). Tolcapone and fatigue scale. Parkinsonism Relat Disord 11: 4955.
fulminant hepatitis. Lancet 352: 958. Bushmann M, Dobmeyer SM, Leeker L et al. (1989). Swal-
Aubert I, Guigoni C, Hakansson K et al. (2005). Increased lowing abnormalities and their response to treatment in
D1 dopamine receptor signaling in levodopa-induced Parkinsons disease. Neurology 39: 13091312.
dyskinesia. Ann Neurol 57: 1726. Buzas B, Max MB (2004). Pain in Parkinson disease.
Ballard PA, Tetrud JW, Langston JW (1985). Permanent Neurology 62: 21562157.
human parkinsonism due to 1-methyl-4-phenyl-1,2,3,6- Calabresi P, Giacomini P, Centonze D et al. (2000). Levodo-
tetrahydropyridine (MPTP): seven cases. Neurology 35: pa-induced dyskinesia: a pathological form of striatal
949956. synaptic plasticity? Ann Neurol 47 (4 Suppl 1): S60S68.
Bara-Jimnez W, Morris M, Dimitrova T et al. (2004). Alpha Calon F, Morissette M, Ghribi O et al. (2002a). Alteration of
2A adrenergic antagonistic effects in advanced Parkin- glutamate receptors in the striatum of dyskinetic 1-methyl-4-
sons diseases. 8th International Congress of Parkinsons phenyl-1,2,3,6-tetrahydropyridine-treated monkeys following
Disease and Movement Disorders, 14th17th June 2004, dopamine agonist treatment. Prog Neuropsychopharmacol
Rome, Italy. Biol Psychiatry 26: 127138.
Baratti M, Calzetti S (1984). Fluctuation of arterial blood Calon F, Birdi S, Rajput AH et al. (2002b). Increase of pre-
pressure during end-of-dose akinesia in Parkinsons disease. proenkephalin mRNA levels in the putamen of Parkinson
J Neurol Neurosurg Psychiatry 47: 12411243. disease patients with levodopa-induced dyskinesias.
Baruzzi A, Contin M, Riva R et al. (1987). Influence of meal J Neuropathol Exp Neurol 61: 186196.
ingestion time on pharmacokinetics of orally administered Calon F, Morissette M, Rajput AH et al. (2003). Changes of
levodopa in parkinsonian patients. Clin Neuropharmacol GABA receptors and dopamine turnover in the postmor-
10: 527537. tem brains of parkinsonians with levodopa-induced motor
Bateson MC, Gibberd FB, Wilson RS (1973). Salivary symp- complications. Mov Disord 18: 241253.
toms in Parkinson disease. Arch Neurol 29: 274275. Cantello R, Gilli M, Riccio A et al. (1986). Mood changes
Bejjani BP, Arnulf I, Demeret S et al. (2000). Levodopa- associated with end-of-dose deterioration in Parkinsons
induced dyskinesias in Parkinsons disease: is sensitization disease: a controlled study. J Neurol Neurosurg Psychiatry
reversible? Ann Neurol 47: 655658. 49: 11821190.
Bezard E, Brotchie JM, Gross CE (2001). Pathophysiology Carter JH, Nutt JG, Woodward WR et al. (1989). Amount and
of levodopa-induced dyskinesia: potential for new thera- distribution of dietary protein affects clinical response to
pies. Nat Rev Neurosci 2: 577588. levodopa in Parkinsons disease. Neurology 39: 552556.
Blanchet PJ, Calon F, Martel JC et al. (1995). Continuous Chase TN (1998a). Levodopa therapy: consequences of the
administration decreases and pulsatile administration nonphysiologic replacement of dopamine. Neurology
increases behavioral sensitivity to a novel dopamine 50 (5 Suppl 5): S17S25.
D2 agonist (U-91356A) in MPTP-exposed monkeys. Chase TN (1998b). The significance of continuous dopami-
J Pharmacol Exp Ther 272: 854859. nergic stimulation in the treatment of Parkinsons disease.
Bojo L, Cassuto J (1992). Gastric reflex relaxation by colo- Drugs 55 (Suppl 1): 19.
nic distension. J Auton Nerv Syst 38: 5764. Chase TN, Mouradian MM, Engber TM (1993). Motor
Boucai L, Cerquetti D, Merello M (2004). Functional surgery response complications and the function of striatal efferent
for Parkinsons disease treatment: a structured analysis of a systems. Neurology 43 (12 Suppl 6): S23S27.
decade of published literature. Br J Neurosurg 18: 213222. Chaudhuri KR, Clough C (1998). Subcutaneous apomor-
Braak H, Ghebremedhin E, Rub U et al. (2004). Stages in the phine in Parkinsons disease. BMJ 316: 641.
development of Parkinsons disease-related pathology. Chaudhuri KR, Critchley P, Abbott RJ et al. (1988). Subcuta-
Cell Tissue Res 318: 121134. neous apomorphine for on-off oscillations in Parkinsons
Bravi D, Mouradian MM, Roberts JW et al. (1993). End-of- disease. Lancet 2 (8622): 1260.
dose dystonia in Parkinsons disease. Neurology 43: Christmas TJ, Kempster PA, Chapple CR et al. (1988). Role
21302131. of subcutaneous apomorphine in parkinsonian voiding
Bravi D, Mouradian MM, Roberts JW et al. (1994). Wear- dysfunction. Lancet 2: 14511453.
ing-off fluctuations in Parkinsons disease: contribution Chudler EH, Dong WK (1995). The role of the basal ganglia
of postsynaptic mechanisms. Ann Neurol 36: 2731. in nociception and pain. Pain 60: 338.
Breier A (1995). Serotonin, schizophrenia and antipsychotic Clarke A, Brewer F, Johnson ES et al. (2003). A new for-
drug action. Schizophr Res 14: 187202. mulation of selegiline: improved bioavailability and
Brotchie JM, Lee J, Venderova K (2005). Levodopa-induced selectivity for MAO-B inhibition. J Neural Transm 110:
dyskinesia in Parkinsons disease. J Neural Transm 12411255.
112 (3): 359391. Clarke CE, Deane K (2001a). Ropinirole versus bromocrip-
Brown RG, Marsden CD, Quinn N et al. (1984). Alterations tine for levodopa-induced complications in Parkinsons
in cognitive performance and affect-arousal state during disease. Cochrane Database Syst Rev CD001517.
176 S. H. FOX AND A. E. LANG

Clarke CE, Deane K (2001b). Cabergoline versus bromocrip- Deep-Brain Stimulation for Parkinsons Disease Study
tine for levodopa-induced complications in Parkinsons Group (2001). Deep-brain stimulation of the subthalamic
disease. Cochrane Database Syst Rev CD001519. nucleus or the pars interna of the globus pallidus in
Clarke CE, Speller JM (2000). Pergolide versus bromocrip- Parkinsons disease. N Engl J Med 345: 956963.
tine for levodopa-induced motor complications in Parkin- Delis D, Direnfeld L, Alexander MP et al. (1982). Cognitive
sons disease. Cochrane Database Syst Rev CD000236. fluctuations associated with on-off phenomenon in Parkin-
Clarke CE, Speller JM, Clarke JA (2000). Pramipexole ver- son disease. Neurology 32: 10491052.
sus bromocriptine for levodopa-induced complications in Dewey RB Jr, Hutton RB, LeWitt PA et al. (2001). A rando-
Parkinsons disease. Cochrane Database Syst Rev mized, double-blind, placebo-controlled trial of subcuta-
CD002259.2000. neously injected apomorphine for parkinsonian off-state
Colosimo C, Merello M, Hughes AJ et al. (1996). Motor events. Arch Neurol 58: 13851392.
response to acute dopaminergic challenge with apomor- Djaldetti R, Baron J, Ziv I et al. (1996). Gastric emptying in
phine and levodopa in Parkinsons disease: implications Parkinsons disease: patients with and without response
for the pathogenesis of the on-off phenomenon. J Neurol fluctuations. Neurology 46: 10511054.
Neurosurg Psychiatry 60: 634637. Djaldetti R, Shifrin A, Rogowski Z et al. (2004). Quantitative
Colzi A, Turner K, Lees AJ (1998). Continuous subcuta- measurement of pain sensation in patients with Parkinson
neous waking day apomorphine in the long term treatment disease. Neurology 62: 21712175.
of levodopa induced interdose dyskinesias in Parkinsons Dunah AW, Standaert DG (2001). Dopamine D1 receptor-
disease. J Neurol Neurosurg Psychiatry 64: 573576. dependent trafficking of striatal NMDA glutamate receptors
Comella CL, Goetz CG (1994). Akathisia in Parkinsons to the postsynaptic membrane. J Neurosci 21: 55465558.
disease. Mov Disord 9: 545549. Duvoisin RC (1974). Variations in the on-off phenomena.
Comella CL, Tanner CM, Ristanovic RK (1993). Polysomno- Adv Neurol 5: 339340.
graphic sleep measures in Parkinsons disease patients Edwards LL, Quigley EM, Pfeiffer RF (1992). Gastrointest-
with treatment-induced hallucinations. Ann Neurol 34: inal dysfunction in Parkinsons disease: frequency and
710714. pathophysiology. Neurology 42: 726732.
Contin M, Riva R, Martinelli P et al. (1998). Effect of meal Engber TM, Susel Z, Juncos JL et al. (1989). Continuous and
timing on the kinetic-dynamic profile of levodopa/carbi- intermittent levodopa differentially affect rotation induced
dopa controlled release [corrected] in parkinsonian by D-1 and D-2 dopamine agonists. Eur J Pharmacol
patients. Eur J Clin Pharmacol 54: 303308. 168 (3): 291298.
Cooper JA, Sagar HJ, Jordan N et al. (1991). Cognitive Evans MA, Broe GA, Triggs EJ et al. (1981). Gastric empty-
impairment in early, untreated Parkinsons disease and ing rate and the systemic availability of levodopa in the
its relationship to motor disability. Brain 114: 20952122. elderly parkinsonian patient. Neurology 31: 12881294.
Corbin DO, Williams AC (1987). Stridor during dystonic Evans AH, Katzenschlager R, Paviour D et al. (2004). Pund-
phases of Parkinsons disease. J Neurol Neurosurg Psy- ing in Parkinsons disease: its relation to the dopamine
chiatry 50: 821822. dysregulation syndrome. Mov Disord 19: 397405.
Cotzias GC, Papavasiliou PS, Fehling C et al. (1970). Simila- Fabbrini G, Mouradian MM, Juncos JL et al. (1988). Motor
rities between neurologic effects of L-dipa and of apomor- fluctuations in Parkinsons disease: central pathophysiolo-
phine. N Engl J Med 282: 3133. gical mechanisms, Part I. Ann Neurol 24: 366371.
Crossman AR (1990). A hypothesis on the pathophysiologi- Factor SA, Friedman JH (1997). The emerging role of cloza-
cal mechanisms that underlie levodopa- or dopamine pine in the treatment of movement disorders. Mov Disord
agonist-induced dyskinesia in Parkinsons disease: impli- 12: 483496.
cations for future strategies in treatment. Mov Disord 5: Factor SA, Brown DL, Molho ES (2000). Subcutaneous apo-
100108. morphine injections as a treatment for intractable pain in
Currie LJ, Harrison MB, Trugman JM et al. (1998). Early Parkinsons disease. Mov Disord 15: 167169.
morning dystonia in Parkinsons disease. Neurology 51: Factor SA, Molho ES, Feustel PJ et al. (2001). Long-term
283285. comparative experience with tolcapone and entacapone
De Keyser J, Vincken W (1985). L-dopa-induced respiratory in advanced Parkinsons disease. Clin Neuropharmacol
disturbance in Parkinsons disease suppressed by tiapride. 24: 295299.
Neurology 35: 235237. Factor SA, Feustel PJ, Friedman JH et al. (2003). Longitudinal
de la Fuente-Fernandez R, Lu JQ, Sossi V et al. (2001). outcome of Parkinsons disease patients with psychosis.
Biochemical variations in the synaptic level of dopamine Neurology 60: 17561761.
precede motor fluctuations in Parkinsons disease: PET Fahn S (1974). On-off phenomenon with levodopa therapy
evidence of increased dopamine turnover. Ann Neurol in parkinsonism: clinical and pharmacologic correlations
49: 298303. and the effects of intramuscular pyridoxine. Neurology
de la Fuente-Fernandez R, Sossi V, Huang Z et al. (2004). 24: 431441.
Levodopa-induced changes in synaptic dopamine levels Fahn S (1977). Episodic failure of absorption of levodopa: a
increase with progression of Parkinsons disease: implica- factor in the control of clinical fluctuations in the treat-
tions for dyskinesias. Brain 127: 27472754. ment of parkinsonism. Neurology 27: 390.
MOTOR AND NON-MOTOR FLUCTUATIONS 177

Fahn S (1982). Fluctuations of disability in Parkinsons dis- off phases of the disease. J Neurol Neurosurg Psychiatry
ease: pathophysiological aspects. In: CD Marsden, S Fahn 49: 657660.
(Eds.), Movement Disorders. Butterworth Scientific, Golbe LI, Lieberman AN, Muenter MD et al. (1988). Depre-
London, pp. 123145. nyl in the treatment of symptom fluctuations in advanced
Fahn S (1995). The freezing phenomenon in parkinsonism. Parkinsons disease. Clin Neuropharmacol 11: 4555.
Adv Neurol 67: 5363. Goetz CG (2003). Treatment of advanced Parkinsons disease:
Fahn S, Oakes D, Shoulson I et al. (2004). Parkinson Study an evidence-based analysis. Adv Neurol 91: 213228.
Group. Levodopa and the progression of Parkinsons dis- Goetz CG, Stebbins GT (1993). Risk factors for nursing
ease. N Engl J Med 351: 24982508. home placement in advanced Parkinsons disease. Neurol-
Fenelon G, Mahieux F, Huon R et al. (2000). Hallucinations ogy 43: 22272229.
in Parkinsons disease: prevalence, phenomenology and Goetz CG, Lutge W, Tanner CM (1986a). Autonomic dys-
risk factors. Brain 123: 733745. function in Parkinsons disease. Neurology 36: 7375.
Finberg JP, Lamensdorf I, Weinstock M et al. (1999). Phar- Goetz CG, Tanner CM, Levy M et al. (1986b). Pain in Par-
macology of rasagiline (N-propargyl-1R-aminoindan). kinsons disease. Mov Disord 1: 4549.
Adv Neurol 80: 495499. Goetz CG, Pappert EJ, Blasucci LM et al. (1998). Intravenous
Fitzmaurice H, Fowler CJ, Rickards D et al. (1985). Micturi- levodopa in hallucinating Parkinsons disease patients:
tion disturbance in Parkinsons disease. Br J Urol 57: high-dose challenge does not precipitate hallucinations.
652656. Neurology 50: 515517.
Ford B, Louis ED, Greene P et al. (1996). Oral and genital Goetz CG, Wuu J, Curgian LM et al. (2005). Hallucinations
pain syndromes in Parkinsons disease. Mov Disord 11: and sleep disorders in PD: six-year prospective longitudi-
421426. nal study. Neurology 64: 8186.
Forsberg M, Lehtonen M, Heikkinen M et al. (2003). Phar- Gordin A, Kaakkola S, Teravainen H (2003). Position of
macokinetics and pharmacodynamics of entacapone and COMT inhibition in the treatment of Parksons disease.
tolcapone after acute and repeated administration: a com- In: A Gordin, S Kaakkola, H Teravainen (Eds.), Vol. 91.
parative study in the rat. J Pharmacol Exp Ther 304: Lippincott Williams and Williams, USA, pp. 237250.
498506. Gotham AM, Brown RG, Marsden CD (1988). Frontal cog-
Fox S, Silverdale M, Kellett M et al. (2004). Non-subtype- nitive function in patients with Parkinsons disease on
selective opioid receptor antagonism in treatment of and off levodopa. Brain 111: 299321.
levodopa-induced motor complications in Parkinsons Graham JM, Grunewald RA, Sagar HJ (1997). Hallucinosis
disease. Mov Disord 19: 554560. in idiopathic Parkinsons disease. J Neurol Neurosurg Psy-
Frankel JP, Lees AJ, Kempster PA et al. (1990). Subcutaneous chiatry 63: 434440.
apomorphine in the treatment of Parkinsons disease. Green J, McDonald WM, Vitek JL et al. (2002). Cognitive
J Neurol Neurosurg Psychiatry 53: 96101. impairments in advanced PD without dementia. Neurology
Friedman A, Sienkiewicz J (1991). Psychotic complications 59: 13201324.
of long-term levodopa treatment of Parkinsons disease. Gulati A, Alison F, Frauke S et al. (2004). A clinical obser-
Acta Neurol Scand 84: 111113. vational study of the patterns and occurrence of non-motor
Friedman J, Friedman H (1993). Fatigue in Parkinsons symptoms in Parkinsons disease ranging from early to
disease. Neurology 43: 20162018. advanced disease. Mov Disord 19 (Suppl 9): 406.
Friedman JH (1994). Punding on levodopa. Biol Psychiatry Gunal DI, Nurichalichi K, Tuncer N et al. (2002). The clinical
36: 350351. profile of nonmotor fluctuations in Parkinsons disease
Funkiewiez A, Ardouin C, Caputo E et al. (2004). Long term patients. Can J Neurol Sci 29: 6164.
effects of bilateral subthalamic nucleus stimulation on Hadj Tahar A, Gregoire L, Bangassoro E et al. (2000). Sus-
cognitive function, mood, and behaviour in Parkinsons tained cabergoline treatment reverses levodopa-induced
disease. J Neurol Neurosurg Psychiatry 75: 834839. dyskinesias in parkinsonian monkeys. Clin Neuropharma-
Gancher ST, Nutt JG, Woodward WR (1995). Apomorphine col 23: 195202.
infusional therapy in Parkinsons disease: clinical utility Harding AJ, Stimson E, Henderson JM et al. (2002). Clini-
and lack of tolerance. Mov Disord 10: 3743. cal correlates of selective pathology in the amygdala
Giladi N, McMahon D, Przedborski S et al. (1992).Motor of patients with Parkinsons disease. Brain 125:
blocks in Parkinsons disease Neurology 42: 333339. 24312445.
Giladi N, Kao R, Fahn S (1997). Freezing phenomenon in Hardoff R, Sula M, Tamir A et al. (2001). Gastric emptying
patients with parkinsonian syndromes. Mov Disord 12: time and gastric motility in patients with Parkinsons disease.
302305. Mov Disord 16: 10411047.
Giovannoni G, OSullivan JD, Turner K et al. (2000). Hedo- Hagelberg N, Martikainen IK, Mansikka H et al. (2002).
nistic homeostatic dysregulation in patients with Parkin- Dopamine D2 receptor binding in the human brain is
sons disease on dopamine replacement therapies. associated with the response to painful stimulation and
J Neurol Neurosurg Psychiatry 68: 423428. pain modulatory capacity. Pain 99: 273279.
Girotti F, Carella F, Grassi MP et al. (1986). Motor and cog- Hauser RA, Holford NH (2002). Quantitative description
nitive performances of parkinsonian patients in the on and of loss of clinical benefit following withdrawal of
178 S. H. FOX AND A. E. LANG

levodopa-carbidopa and bromocriptine in early Parkinsons of catechol-O-methyltransferase. Gen Pharmacol 25:


disease. Mov Disord 17: 961968. 813824.
Hauser RA, Koller WC, Hubble JP et al. (2000). Time course Kapoor R, Turjanski N, Frankel J et al. (1990). Intranasal
of loss of clinical benefit following withdrawal of levo- apomorphine: a new treatment in Parkinsons disease.
dopa/carbidopa and bromocriptine in early Parkinsons J Neurol Neurosurg Psychiatry 53: 1015.
disease. Mov Disord 15: 485489. Karstaedt PJ, Pincus JH (1992). Protein redistribution diet
Henry B, Brotchie JM (1996). Potential of opioid antagonists remains effective in patients with fluctuating parkinson-
in the treatment of levodopa-induced dyskinesias in Par- ism. Arch Neurol 49: 149151.
kinsons disease. Drugs Aging 9: 149158. Kedia S, Moro E, Tagliati M et al. (2004). Emergence of
Henry B, Crossman AR, Brotchie JM (1999). Effect of restless legs syndrome during subthalamic stimulation for
repeated L-DOPA, bromocriptine, or lisuride administra- Parkinson disease. Neurology 63: 24102412.
tion on preproenkephalin-A and preproenkephalin-B Kempster PA, Lees AJ, Crichton P et al. (1989). Off-period
mRNA levels in the striatum of the 6-hydroxydopamine- belching due to a reversible disturbance of oesophageal
lesioned rat. Exp Neurol 155: 204220. motility in Parkinsons disease and its treatment with apo-
Henry B, Duty S, Fox SH et al. (2003). Increased striatal morphine. Mov Disord 4: 4752.
pre-proenkephalin B expression is associated with dyski- Keshavan MS, David AS, Narayanen HS et al. (1986). On-
nesia in Parkinsons disease. Exp Neurol 183: 458468. off phenomena and manic-depressive mood shifts: case
Herlofson K, Larsen JP (2002). Measuring fatigue in patients report. J Clin Psychiatry 47: 9394.
with Parkinsons diseasethe Fatigue Severity Scale. Eur Keyser DL, Rodnitzky RL (1991). Neuroleptic malignant
J Neurol 9: 595600. syndrome in Parkinsons disease after withdrawal or
Hillen ME, Sage JL (1996). Nonmotor fluctuations in patients alteration of dopaminergic therapy. Arch Intern Med
with Parkinsons disease. Neurology 47: 11801183. 151: 794796.
Hoehn MM, Elton RL (1985). Low dosages of bromocriptine
Khan NL, Graham E, Critchley P et al. (2003). Parkin disease:
added to levodopa in Parkinsons disease. Neurology 35:
a phenotypic study of a large case series. Brain 126:
199206.
12791292.
Holloway RG, Shoulson I, Fahn S et al. (2004). Parkinson
Klintenberg R, Svenningsson P, Gunne L et al. (2002).
Study Group. Pramipexole vs levodopa as initial treatment
Naloxone reduces levodopa-induced dyskinesias and
for Parkinson disease: a 4-year randomized controlled
apomorphine-induced rotations in primate models of
trial. Arch Neurol 61: 10441053.
parkinsonism. J Neural Transm 109: 12951307.
Honey CR, Stoessl AJ, Tsui JK et al. (1999). Unilateral
Koike Y, Takahashi A (1997). Autonomic dysfunction in
pallidotomy for reduction of parkinsonian pain. J Neuro-
Parkinsons disease. Eur Neurol 38 (Suppl 2): 812.
surg 91: 198201.
Horstink MW, Zijlmans JC, Pasman JW et al. (1990). Koller W, Guarnieri M, Hubble J et al. (2005). An open-label
Severity of Parkinsons disease is a risk factor for peak- evaluation of the tolerability and safety of Stalevo (carbi-
dose dyskinesia. J Neurol Neurosurg Psychiatry 53: dopa, levodopa and entacapone) in Parkinsons disease
224226. patients experiencing wearing-off. J Neural Transm 112:
Huber SJ, Shulman HG, Paulson GW et al. (1987). Fluctua- 221230.
tions in plasma dopamine level impair memory in Parkin- Koller WC (1984). Sensory symptoms in Parkinsons dis-
sons disease. Neurology 37: 13711375. ease. Neurology 34: 957959.
Huber SJ, Shulman HG, Paulson GW et al. (1989). Dose- Kompoliti K, Goetz CG, Leurgans S et al. (2000). On
dependent memory impairment in Parkinsons disease. freezing in Parkinsons disease: resistance to visual cue
Neurology 39: 438440. walking devices. Mov Disord 15: 309312.
Hughes AJ, Bishop S, Lees AJ et al. (1991). Rectal apomor- Korchounov A, Kessler KR, Schipper HI (2004). Differential
phine in Parkinsons disease. Lancet 337: 118. effects of various treatment combinations on cardiovascu-
Hughes AJ, Bishop S, Kleedorfer B et al. (1993). Subcuta- lar dysfunction in patients with Parkinsons disease. Acta
neous apomorphine in Parkinsons disease: response to Neurol Scand 109: 4551.
chronic administration for up to five years. Mov Disord Korlipara LV, Cooper JM, Schapira AH (2004). Differences
8: 165170. in toxicity of the catechol-O-methyl transferase inhibitors,
Ilson J, Fahn S, Cote L (1984). Painful dystonic spasms in tolcapone and entacapone to cultured human neuroblas-
Parkinsons disease. Adv Neurol 40: 395398. toma cells. Neuropharmacology 46: 562569.
Isella V, Melzi P, Grimaldi M et al. (2002). Clinical, neurop- Krack P, Pollak P, Limousin P et al. (1999). From off-period
sychological, and morphometric correlates of apathy in dystonia to peak-dose chorea. The clinical spectrum of vary-
Parkinsons disease. Mov Disord 17: 366371. ing subthalamic nucleus activity. Brain 122: 11331146.
Jankovic J, Nour F (1986). Respiratory dyskinesia in Parkin- Krack P, Batir A, Van Blercom N et al. (2003). Five-year
sons disease. Neurology 36: 303304. follow-up of bilateral stimulation of the subthalamic
Kaakkola S, Gordin A, Mannisto PT (1994). General proper- nucleus in advanced Parkinsons disease. N Engl J Med
ties and clinical possibilities of new selective inhibitors 349: 19251934.
MOTOR AND NON-MOTOR FLUCTUATIONS 179

Krupp LB, Pollina DA (1996). Mechanisms and manage- LeWitt PA, Burns RS, Newman RP (1986). Dystonia in
ment of fatigue in progressive neurological disorders. Curr untreated parkinsonism. Clin Neuropharmacol 9: 293297.
Opin Neurol 9: 456460. Lieberman A, Gopinathan G, Miller E et al. (1990). Rando-
Kumar R, Lozano AM, Kim YJ et al. (1998). Double-blind mized double-blind cross-over study of Sinemet-controlled
evaluation of subthalamic nucleus deep brain stimulation release (CR4 50/200) versus Sinemet 25/100 in Parkinsons
in advanced Parkinsons disease. Neurology 51: 850855. disease. Eur Neurol 30: 7578.
Kumar R, Lang AE, Rodriguez-Oroz MC et al. (2000). Deep Lieberman A, Imke S, Muenter M et al. (1993). Multicenter
brain stimulation of the globus pallidus pars interna in study of cabergoline, a long-acting dopamine receptor agonist,
advanced Parkinsons disease. Neurology 55 (12 Suppl 6): in Parkinsons disease patients with fluctuating responses to
S34S39. levodopa/carbidopa. Neurology 43: 19811984.
Kurlan R (2004). Disabling repetitive behaviors in Parkinsons Limousin P, Krack P, Pollak P et al. (1998). Electrical stimu-
disease. Mov Disord 19: 433437. lation of the subthalamic nucleus in advanced Parkinsons
Kurlan R, Rubin AJ, Miller C et al. (1986). Duodenal deliv- disease. N Engl J Med 339: 11051111.
ery of levodopa for on-off fluctuations in parkinsonism: Loher TJ, Burgunder JM, Pohle T et al. (2002). Long-term
preliminary observations. Ann Neurol 20: 262265. pallidal deep brain stimulation in patients with advanced
Kurth MC, Tetrud JW, Irwin I et al. (1993a). Oral levodopa/ Parkinson disease: 1-year follow-up study. J Neurosurg
carbidopa solution versus tablets in Parkinsons patients 96: 844853.
with severe fluctuations: a pilot study. Neurology 43: Luquin MR, Scipioni O, Vaamonde J et al. (1992). Levodo-
10361039. pa-induced dyskinesias in Parkinsons disease: clinical and
Kurth MC, Tetrud JW, Tanner CM et al. (1993b). Double- pharmacological classification. Mov Disord 7: 117124.
blind, placebo-controlled, crossover study of duodenal infu- MacMahon DG, Sachdev D, Boddie HG et al. (1990). A
sion of levodopa/carbidopa in Parkinsons disease patients comparison of the effects of controlled-release levodopa
with on-off fluctuations. Neurology 43: 16981703. (Madopar CR) with conventional levodopa in late Par-
Kurth MC, Adler CH, Hilaire MS et al. (1997). Tolcapone kinsons disease. J Neurol Neurosurg Psychiatry 53:
improves motor function and reduces levodopa require- 220223.
ment in patients with Parkinsons disease experiencing Mancini F, Zangaglia R, Cristina S et al. (2003). Double-
motor fluctuations: a multicenter, double-blind, rando- blind placebo controlled study to evaluate the efficacy
mized, placebo-controlled trial. Tolcapone Fluctuator and safety of botulinum toxin type A in the treatment of
Study Group I. Neurology 48: 8187. drooling in parkinsonism. Mov Disord 18: 685688.
Lagrange E, Krack P, Moro E et al. (2002). Bilateral subtha- Manford M, Andermann F (1998). Complex visual halluci-
lamic nucleus stimulation improves health-related quality nations. Clinical and neurobiological insights. Brain 121:
of life in PD. Neurology 59: 19761978. 18191840.
Lang AE, Johnson K (1987). Akathisia in idiopathic Parkin- Manson AJ, Katzenschlager R, Hobart J et al. (2001). High
sons disease. Neurology 37: 477481. dose naltrexone for dyskinesias induced by levodopa.
Lang AE, Lozano AM (1998). Parkinsons disease. First of J Neurol Neurosurg Psychiatry 70: 554556.
two parts. N Engl J Med 339: 10441053. Manson AJ, Turner K, Lees AJ (2002). Apomorphine mono-
Langston JW, Forno LS (1978). The hypothalamus in Parkin- therapy in the treatment of refractory motor complications
son disease. Ann Neurol 3: 129133. of Parkinsons disease: long-term follow-up study of 64
Lawrence AD, Evans AH, Lees AJ (2003). Compulsive use patients. Mov Disord 17: 12351241.
of dopamine replacement therapy in Parkinsons disease: Maricle RA, Nutt JG, Valentine RJ et al. (1995). Dose-response
reward systems gone awry? Lancet Neurol 2: 595604. relationship of levodopa with mood and anxiety in fluctuat-
Leenders KL, Poewe WH, Palmer AJ et al. (1986). Inhibition ing Parkinsons disease: a double-blind, placebo-controlled
of L-[18F]fluorodopa uptake into human brain by amino study. Neurology 45: 17571760.
acids demonstrated by positron emission tomography. Marras C, Lang AE (2003). Measuring motor complications
Ann Neurol 20: 258262. in clinical trials for early Parkinsons disease. J Neurol
Lees AJ (1987). A sustained-release formulation of L-dopa Neurosurg Psychiatry 74: 143146.
(Madopar HBS) in the treatment of nocturnal and early- Marsden CD (1994). Parkinsons disease. J Neurol Neuro-
morning disabilities in Parkinsons disease. Eur Neurol surg Psychiatry 57: 672681.
27 (Suppl 1): 126134. Marsden CD, Parkes JD (1976). On-off effects in patients
Lees AJ (1989). The on-off phenomenon. J Neurol Neurosurg with Parkinsons disease on chronic levodopa therapy.
Psychiatry (Suppl) Jun: 2937. Lancet 1: 292296.
Lees AJ (1993). Dopamine agonists in Parkinsons disease: a Marsden CD, Parkes JD (1977). Success and problems of
look at apomorphine. Fundam Clin Pharmacol 7 (34): long-term levodopa therapy in Parkinsons disease. Lancet
121128. 1 (8007): 345349.
Lees AJ, Shaw KM, Kohout LJ et al. (1977). Deprenyl in Marsden CD, Parkes JD, Quinn N (1982). Fluctuations of dis-
Parkinsons disease. Lancet 2 (8042): 791795. ability in Parkinsons diseaseclinical aspects. In: CD
Lees AJ, Shaw KM, Stern GM (1978). Baclofen in Parkin- Marsden, S Fahn (Eds.), Movement Disorders. Butterworth
sons disease. J Neurol Neurosurg Psychiatry 41: 707708. Scientific, London, pp. 96122.
180 S. H. FOX AND A. E. LANG

Mathers SE, Kempster PA, Law PJ et al. (1989). Anal Moskovitz C, Moses H 3rd, Klawans HL (1978). Levodopa-
sphincter dysfunction in Parkinsons disease. Arch Neurol induced psychosis: a kindling phenomenon. Am J Psychiatry
46: 10611064. 135: 669675.
Mathias CJ, Kimber JR (1998). Treatment of postural hypo- Mouradian MM, Juncos JL, Fabbrini G et al. (1988). Motor
tension. J Neurol Neurosurg Psychiatry 65: 285289. fluctuations in Parkinsons disease: central pathophysiolo-
Matison R, Mayeux R, Rosen J et al. (1982). Tip-of-the- gical mechanisms, Part II. Ann Neurol 24: 372378.
tongue phenomenon in Parkinson disease. Neurology 32: Movement Disorder Society Task Force on Rating Scales for
567570. Parkinsons Disease (2003). The Unified Parkinsons
McKeith IG, Galasko D, Kosaka K et al. (1996). Consensus Disease Rating Scale (UPDRS): status and recommenda-
guidelines for the clinical and pathologic diagnosis of demen- tions. Mov Disord 18: 738750.
tia with Lewy bodies (DLB): report of the consortium on Muenter MD, Tyce GM (1971). L-dopa therapy of Parkin-
DLB international workshop. Neurology 47: 11131124. sons disease: plasma L-dopa concentration, therapeutic
Meco G, Bonifati V, Bedini L et al. (1991). Relations response, and side effects. Mayo Clin Proc 46: 231239.
between on-off phenomena and cognitive functions in Muenter MD, Sharpless NS, Tyce GM et al. (1977). Patterns of
Parkinson disease. Ital J Neurol Sci 12: 5762. dystonia (I-D-I and D-I-D-) in response to I-dopa therapy
Melamed E (1979). Early morning dystonia. A late side effect in Parkinsons disease. Mayo Clin Proc 52: 163174.
of long-term levodopa therapy in Parkinsons disease. Arch Muller T, Woitalla D, Schulz D et al. (2000). Tolcapone
Neurol 36: 308310. increases maximum concentration of levodopa. J Neural
Melamed E, Bitton V (1984). Delayed onset of responses to Transm 107: 113119.
individual doses of L-dopa in parkinsonian fluctuators: Myllyla VV, Kultalahti ER, Haapaniemi H et al. (2001). FILO-
an additional side effect of long-term L-dopa therapy. MEN Study Group. Twelve-month safety of entacapone in
Neurology 34 (Suppl 2): 270. patients with Parkinsons disease. Eur J Neurol 8: 5360.
Melamed E, Hefti F, Wurtman RJ (1980). Nonaminergic Nausieda PA, Weiner WJ, Kaplan LR et al. (1982). Sleep
striatal neurons convert exogenous L-dopa to dopamine disruption in the course of chronic levodopa therapy: an
in parkinsonism. Ann Neurol 8: 558563. early feature of the levodopa psychosis. Clin Neurophar-
Meltzer HY (1992). The importance of serotonin-dopamine macol 5: 183194.
interactions in the action of clozapine. Br J Psychiatry Ng KY, Colburn RW, Kopin IJ (1971). Effects of L-dopa on
Suppl 17: 2229. efflux of cerebral monoamines from synaptosomes. Nature
Menza MA, Sage J, Marshall E et al. (1990). Mood changes 230: 331332.
and on-off phenomena in Parkinsons disease. Mov Disord Nissenbaum H, Quinn NP, Brown RG et al. (1987). Mood
5: 148151. swings associated with the on-off phenomenon in
Merello M, Lees AJ (1992). Beginning-of-dose motor dete- Parkinsons disease. Psychol Med 17: 899904.
rioration following the acute administration of levodopa Nutt JG (1990). Levodopa-induced dyskinesia:review, obser-
and apomorphine in Parkinsons disease. J Neurol Neuro- vation and speculations. Neurology 40: 340345.
surg Psychiatry 55: 10241026. Nutt JG, Fellman JH (1984). Pharmacokinetics of levodopa.
Merims D, Shabtai H, Korczyn AD et al. (2004). Antiparkin- Clin Neuropharmacol 7: 3549.
sonian medication is not a risk factor for the development Nutt JG, Holford NH (1996). The response to levodopa in
of hallucinations in Parkinsons disease. J Neural Transm Parkinsons disease:imposing pharmacological law and
111: 14471453. order. Ann Neurol 39: 561573.
Mohr E, Fabbrini G, Williams J et al. (1989). Dopamine and Nutt JG, Nygaard TG (2001). Response to L-DOPA in
memory function in Parkinsons disease. Mov Disord DOPA-responsive dystonia. Arch Neurol 58 (6): 905910.
4: 113120. Nutt JG, Woodward WR, Hammerstad JP et al. (1984). The
Molina JA, Sainz-Artiga MJ, Fraile A et al. (2000). Pathologic on-off phenomenon in Parkinsons disease. Relation to
gambling in Parkinsons disease: a behavioral manifestation levodopa absorption and transport. N Engl J Med 310:
of pharmacologic treatment? Mov Disord 15: 869872. 483488.
Molinuevo JL, Valldeoriola F, Tolosa E et al. (2000). Levo- Nutt JG, Gancher ST, Woodward WR (1988). Does an inhi-
dopa withdrawal after bilateral subthalamic nucleus bitory action of levodopa contribute to motor fluctuations?
stimulation in advanced Parkinson disease. Arch Neurol Neurology 38: 15531557.
57: 983988. Nutt JG, Woodward WR, Carter JH et al. (1992). Effect of
Montastruc JL, Rascol O, Senard JM et al. (1991). Sublingual long-term therapy on the pharmacodynamics of levodopa.
apomorphine in Parkinsons disease: a clinical and phar- Relation to on-off phenomenon. Arch Neurol 49: 11231130.
macokinetic study. Clin Neuropharmacol 14: 432437. Nutt JG, Woodward WR, Beckner RM et al. (1994). Effect
Moro E, Scerrati M, Romito LM et al. (1999). Chronic of peripheral catechol-O-methyltransferase inhibition on
subthalamic nucleus stimulation reduces medication the pharmacokinetics and pharmacodynamics of levodopa
requirements in Parkinsons disease. Neurology 53: 8590. in parkinsonian patients. Neurology 44: 913919.
Moro E, Esselink RJ, Benabid AL et al. (2002). Response to Nutt JG, Carter JH, Lea ES et al. (1997). Motor fluctuations
levodopa in parkinsonian patients with bilateral subthala- during continuous levodopa infusions in patients with
mic nucleus stimulation. Brain 125: 24082417. Parkinsons disease. Mov Disord 12: 285292.
MOTOR AND NON-MOTOR FLUCTUATIONS 181

Nutt JG, Obeso JA, Stocchi F (2000). Continuous dopamine- Parkinson Study Group (2003). A controlled trial of rotigotine
receptor stimulation in advanced Parkinsons disease. monotherapy in early Parkinsons disease. Arch Neurol 60:
Trends Neurosci 23 (10 Suppl): S109S115. 17211728.
Nutt JG, Carter JH, Lea ES et al. (2002). Evolution of the Parkinson Study Group (2005). A randomized placebo-con-
response to levodopa during the first 4 years of therapy. trolled trial of rasagiline in levodopa-treated patients with
Ann Neurol 51: 686693. Parkinson disease and motor fluctuations: the PRESTO
Nyholm D, Nilsson Remahl AI, Dizdar N et al. (2005). study. Arch Neurol 62: 241248.
Duodenal levodopa infusion monotherapy vs oral Pearce RK, Heikkila M, Linden IB et al. (2001). L-dopa
polypharmacy in advanced Parkinson disease. Neurology induces dyskinesia in normal monkeys: behavioural and
64: 216223. pharmacokinetic observations. Psychopharmacology
Obeso JA, Rodriguez-Oroz MC, Chana P et al. (2000a). The (Berl) 156: 402409.
evolution and origin of motor complications in Parkin- Peppe A, Pierantozzi M, Bassi A et al. (2004). Stimulation of
sons disease. Neurology 55 (11 Suppl 4): S13S20. the subthalamic nucleus compared with the globus pallidus
Obeso JA, Rodriguez-Oroz MC, Rodriguez M et al. (2000a). internus in patients with Parkinson disease. J Neurosurg
Pathophysiology of the basal ganglia in Parkinsons 101: 195200.
disease. Trends Neurosci 23 (10 Suppl): S8S19. Pfeiffer RF, Sucha EL (1989). On-off-induced lethal
Oh JD, Vaughan CL, Chase TN (1999). Effect of dopamine hyperthermia. Mov Disord 4: 338341.
denervation and dopamine agonist administration on ser- Piccini P, Weeks RA, Brooks DJ (1997). Alterations in opioid
ine phosphorylation of striatal NMDA receptor subunits. receptor binding in Parkinsons disease patients with levo-
Brain Res 821: 433442. dopa-induced dyskinesias. Ann Neurol 42: 720726.
Olanow CW, Koller WC (1998). An algorithm (decision Picconi B, Centonze D, Hakansson K et al. (2003). Loss of
tree) for the management of Parkinsons disease: treatment bidirectional striatal synaptic plasticity in L-DOPA-
guidelines. American Academy of Neurology. Neurology induced dyskinesia. Nat Neurosci 6: 501506.
50 (3 Suppl 3): S1S57. Pincus JH, Barry K (1987). Influence of dietary protein on
Olanow CW, Fahn S, Muenter M et al. (1994). A multicenter motor fluctuations in Parkinsons disease. Arch Neurol
double-blind placebo-controlled trial of pergolide as an 44: 270272.
adjunct to Sinemet in Parkinsons disease. Mov Disord Pinter MM, Pogarell O, Oertel WH (1999). Efficacy, safety,
9: 4047. and tolerance of the non-ergoline dopamine agonist prami-
Olanow CW, Hauser RA, Gauger L et al. (1995). The effect pexole in the treatment of advanced Parkinsons disease:
of deprenyl and levodopa on the progression of Parkin- a double blind, placebo controlled, randomised, multicentre
sons disease. Ann Neurol 38: 771777. study. J Neurol Neurosurg Psychiatry 66: 436441.
Olanow CW, Damier P, Goetz CG et al. (2004). Multicenter, Pluck GC, Brown RG (2002). Apathy in Parkinsons disease.
open-label, trial of sarizotan in Parkinson disease patients J Neurol Neurosurg Psychiatry 73: 636642.
with levodopa-induced dyskinesias (the SPLENDID Poewe W (2003). Psychosis in Parkinsons disease. Mov
Study). Clin Neuropharmacol 27: 5862. Disord 18 (Suppl 6): S80S87.
Ondo WG, Hunter C, Moore W (2004). A double-blind Poewe W, Hogl B (2004). Akathisia, restless legs and periodic
placebo-controlled trial of botulinum toxin B for sialor- limb movements in sleep in Parkinsons disease. Neurology
rhea in Parkinsons disease. Neurology 62: 3740. 63 (8 Suppl 3): S12S16.
Pacchetti C, Albani G, Martignoni E et al. (1995). Off Poewe W, Berger W, Benke T et al. (1991). High-speed
painful dystonia in Parkinsons disease treated with botuli- memory scanning in Parkinsons disease: adverse effects
num toxin. Mov Disord 10: 333336. of levodopa. Ann Neurol 29: 670673.
Papa SM, Chase TN (1996). Levodopa-induced dyskinesias Poewe WH, Lees AJ, Stern GM (1988). Dystonia in Parkin-
improved by a glutamate antagonist in Parkinsonian mon- sons disease: clinical and pharmacological features. Ann
keys. Ann Neurol 39: 574578. Neurol 23: 7378.
Papa SM, Engber TM, Kask AM et al. (1994). Motor fluc- Poewe WH, Deuschl G, Gordin A et al. (2002). Celomen
tuations in levodopa treated parkinsonian rats: relation to Study Group. Efficacy and safety of entacapone in Parkin-
lesion extent and treatment duration. Brain Res 662: sons disease patients with suboptimal levodopa response:
6974. a 6-month randomized placebo-controlled double-blind
Papa SM, Boldry RC, Engber TM et al. (1995). Reversal of study in Germany and Austria (Celomen study). Acta
levodopa-induced motor fluctuations in experimental Neurol Scand 105: 245255.
parkinsonism by NMDA receptor blockade. Brain Res Priano L, Albani G, Brioschi A et al. (2004). Transdermal
701: 1318. apomorphine permeation from microemulsions: a new
Pappert EJ, Goetz CG, Niederman FG et al. (1999). Hallucina- treatment in Parkinsons disease. Mov Disord 19: 937942.
tions, sleep fragmentation, and altered dream phenomena in Quattrone A, Zappia M, Aguglia U et al. (1995). The suba-
Parkinsons disease. Mov Disord 14: 117121. cute levodopa test for evaluating long-duration response
Parkinson Study Group (1997). Entacapone improves motor in Parkinsons disease. Ann Neurol 38: 389395.
fluctuations in levodopa-treated Parkinsons disease Quinn N, Marsden CD (1986). Lithium for painful dystonia
patients. Ann Neurol 42: 747755. in Parkinsons disease. Lancet 1: 1377.
182 S. H. FOX AND A. E. LANG

Quinn NP (1998). Classification of fluctuations in patients Ruottinen HM, Rinne UK (1996b). Entacapone prolongs
with Parkinsons disease. Neurology 51 (2 Suppl 2): levodopa response in a one month double blind study in
S25S29. parkinsonian patients with levodopa related fluctuations.
Quinn NP, Koller WC, Lang AE et al. (1986). Painful Par- J Neurol Neurosurg Psychiatry 60: 3640.
kinsons disease. Lancet 1 (8494): 13661369. Sage JI (2004). Pain in Parkinsons disease. Curr Treat
Rabey JM, Sagi I, Huberman M et al. (2000). Rasagiline Options Neurol 6: 191200.
Study Group. Rasagiline mesylate, a new MAO-B inhibi- Sage JI, Mark MH (1995). Drenching sweats as an off phe-
tor for the treatment of Parkinsons disease: a double-blind nomenon in Parkinsons disease: treatment and relation
study as adjunctive therapy to levodopa. Clin Neurophar- to plasma levodopa profile. Ann Neurol 37: 120122.
macol 23: 324330. Sage JI, Trooskin S, Sonsalla PK et al. (1988). Long-term
Racette BA, Hartlein JM, Hershey T et al. (2002). Clinical duodenal infusion of levodopa for motor fluctuations in
features and comorbidity of mood fluctuations in Parkinsons parkinsonism. Ann Neurol 24: 8789.
disease. J Neuropsychiatry Clin Neurosci 14: 438442. Sage JI, Kortis HI, Sommer W (1990). Evidence for the role
Rajput AH, Martin W, Saint-Hilaire MH et al. (1997). Tolca- of spinal cord systems in Parkinsons disease-associated
pone improves motor function in parkinsonian patients with pain. Clin Neuropharmacol 13: 171174.
the wearing-off phenomenon: a double-blind, placebo- Saint-Cyr JA, Taylor AE, Lang AE (1993). Neuropsychological
controlled, multicenter trial. Neurology 49: 10661071. and psychiatric side effects in the treatment of Parkinsons
Rajput AH, Fenton ME, Di Paolo T et al. (2004). Human disease. Neurology 43 (12 Suppl 6): S47S52.
brain dopamine metabolism in levodopa-induced dyskinesia Samadi P, Gregoire L, Bedard PJ (2003). Opioid antagonists
and wearing-off. Parkinsonism Relat Disord 10: 221226. increase the dyskinetic response to dopaminergic agents in
Rascol O, Fabre N, Blin O et al. (1994). Naltrexone, an opi- parkinsonian monkeys: interaction between dopamine and
ate antagonists, fails to modify motor symptoms in opioid systems. Neuropharmacology 45: 954963.
patients with Parkinsons disease. Mov Disord 9: 437440. Sanchez-Ramos JR, Ortoll R, Paulson GW (1996). Visual
Rascol O, Lees AJ, Senard JM et al. (1996). Ropinirole in hallucinations associated with Parkinson disease. Arch
the treatment of levodopa-induced motor fluctuations in Neurol 53: 12651268.
patients with Parkinsons disease. Clin Neuropharmacol Schrag A, Ben-Shlomo Y, Quinn N (2002). How common are
19: 234245. complications of Parkinsons disease? J Neurol 249: 419423.
Rascol O, Brooks DJ, Korczyn AD et al. (2000). A five-year Seager H (1997). Drug delivery products and the Zydis fast-
study of the incidence of dyskinesia in patients with early dissolving dosage form. J Pharm Pharmacol 50: 375382.
Parkinsons disease who were treated with ropinirole or Seif C, Herzog J, van der Horst C et al. (2004). Effect of
levodopa. 056 Study Group. N Engl J Med 342: subthalamic deep brain stimulation on the function of the
14841491. urinary bladder. Ann Neurol 55: 118120.
Raudino F (2001). Non motor off in Parkinsons disease. Senard JM, Rai S, Lapeyre-Mestre M et al. (1997). Preva-
Acta Neurol Scand 104: 312315. lence of orthostatic hypotension in Parkinsons disease.
Reuter I, Ellis CM, Ray Chaudhuri K (1999). Nocturnal sub- J Neurol Neurosurg Psychiatry 63: 584589.
cutaneous apomorphine infusion in Parkinsons disease Shoulson I, Glaubiger GA, Chase TN (1975). On-off
and restless legs syndrome. Acta Neurol Scand 100: response. Clinical and biochemical correlations during
163167. oral and intravenous levodopa administration in parkinso-
Richard IH, Frank S, McDermott MP et al. (2004). The ups nian patients. Neurology 25: 11441148.
and downs of Parkinson disease: a prospective study of Shulman LM, Taback RL, Bean J et al. (2001). Comorbidity
mood and anxiety fluctuations. Cogn Behav Neurol 17: of the nonmotor symptoms of Parkinsons disease. Mov
201207. Disord 16: 507510.
Riederer P, Youdim MB (1986). Monoamine oxidase acti- Siemers ER, Shekhar A, Quaid K et al. (1993). Anxiety and
vity and monoamine metabolism in brains of parkinsonian motor performance in Parkinsons disease. Mov Disord 8:
patients treated with l-deprenyl. J Neurochem 46: 501506.
13591365. Sieradzan KA, Fox SH, Hill M et al. (2001). Cannabinoids
Riley DE, Lang AE (1993). The spectrum of levodopa- reduce levodopa-induced dyskinesia in Parkinsons
related fluctuations in Parkinsons disease. Neurology 43: disease: a pilot study. Neurology 57: 21082111.
14591464. Silverdale MA, Fox SH, Crossman AR et al. (2003). Potential
Rinne JO, Laihinen A, Lonnberg P et al. (1991). A post-mor- nondopaminergic drugs for Parkinsons disease. Adv
tem study on striatal dopamine receptors in Parkinsons Neurol 91: 273291.
disease. Brain Res 556: 117122. Simon N, Gantcheva R, Bruguerolle B et al. (2004). The
Ruottinen HM, Rinne UK (1996a). Effect of one months effects of a normal protein diet on levodopa plasma
treatment with peripherally acting catechol-O-methyl- kinetics in advanced Parkinsons disease. Parkinsonism
transferase inhibitor, entacapone, on pharmacokinetics Relat Disord 10: 137142.
and motor response to levodopa in advanced parkinsonian Snider SR, Fahn S, Isgreen WP et al. (1976). Primary sen-
patients. Clin Neuropharmacol 19: 222233. sory symptoms in parkinsonism. Neurology 26: 423429.
MOTOR AND NON-MOTOR FLUCTUATIONS 183

Stacy M, Bowron A, Guttman M et al. (2005). Identification Trosch RM, Friedman JH, Lannon MC et al. (1998). Cloza-
of motor and nonmotor wearing-off in Parkinsons pine use in Parkinsons disease: a retrospective analysis
disease: comparison of a patient questionnaire versus a of a large multicentered clinical experience. Mov Disord
clinician assessment. Mov Disord 20 (6): 726733. 13: 377382.
Steiger MJ, Quinn NP, Toone B et al. (1991). Off-period Turjanski N, Lees AJ, Brooks DJ (1997). In vivo studies on
screaming accompanying motor fluctuations in Parkin- striatal dopamine D1 and D2 site binding in L-dopa-trea-
sons disease. Mov Disord 6: 8990. ted Parkinsons disease patients with and without dyskine-
Stein WM, Read S (1997). Chronic pain in the setting of sias. Neurology 49: 717723.
Parkinsons disease and depression. J Pain Symptom Tyne HL, Parsons J, Sinnott A et al. (2004). A 10 year retro-
Manage 14: 255258. spective audit of long-term apomorphine use in Parkin-
Stibe CM, Lees AJ, Kempster PA et al. (1988). Subcutaneous sons disease. J Neurol 251: 13701374.
apomorphine in parkinsonian on-off oscillations. Lancet Uchiyama T, Sakakibara R, Hattori T et al. (2003). Short-
1 (8582): 403406. term effect of a single levodopa dose on micturition distur-
Stolze H, Klebe S, Poepping M et al. (2001). Effects of bilat- bance in Parkinsons disease patients with the wearing-off
eral subthalamic nucleus stimulation on parkinsonian gait. phenomenon. Mov Disord 18: 573578.
Neurology 57: 144146. van Hilten JJ, Weggeman M, van der Velde EA et al. (1993).
Stocchi F, Quinn NP, Barbato L et al. (1994). Comparison Sleep, excessive daytime sleepiness and fatigue in Parkin-
between a fast and a slow release preparation of levodopa sons disease. J Neural Transm Park Dis Dement Sect 5:
and a combination of the two: a clinical and pharmacoki- 235244.
netic study. Clin Neuropharmacol 17: 3844. Varma TR, Fox SH, Eldridge PR et al. (2003). Deep brain
Stocchi F, Nordera G, Marsden CD (1997). Strategies for stimulation of the subthalamic nucleus: effectiveness in
treating patients with advanced Parkinsons disease with advanced Parkinsons disease patients previously reliant
disastrous fluctuations and dyskinesias. Clin Neurophar- on apomorphine. J Neurol Neurosurg Psychiatry 74:
macol 20: 95115. 170174.
Stocchi F, Vacca L, Berardelli A et al. (2001a). Long-duration Vazquez A, Jimenez-Jimenez FJ, Garcia-Ruiz P et al. (1993).
effect and the postsynaptic compartment: study using a Panic attacks in Parkinsons disease. A long-term compli-
dopamine agonist with a short half-life. Mov Disord 16: cation of levodopa therapy. Acta Neurol Scand 87: 1418.
301305. Velasco M, Luchsinger A (1998). Dopamine: pharmacologic
Stocchi F, Vacca L, De Pandis MF et al. (2001b). Subcuta- and therapeutic aspects. Am J Ther 5: 3743.
neous continuous apomorphine infusion in fluctuating Verhagen Metman L, Locatelli ER, Bravi D et al. (1997).
patients with Parkinsons disease: long-term results. Neurol Apomorphine responses in Parkinsons disease and the patho-
Sci 22: 9394. genesis of motor complications. Neurology 48: 369372.
Stocchi F, Ruggieri S, Vacca L et al. (2002). Prospective ran- Verhagen-Metman L, Del Dotto P, van den Munckhof P et al.
domized trial of lisuride infusion versus oral levodopa in (1998). Amantadine as treatment for dyskinesias and motor
patients with Parkinsons disease. Brain 125 (Pt 9): fluctuations in Parkinsons disease. Neurology 50:
20582066. 13231326.
Struck LK, Rodnitzky RL, Dobson JK (1990). Circadian Verhagen Metman L, Konitsiotis S, Chase TN (2000).
fluctuations of contrast sensitivity in Parkinsons disease. Pathophysiology of motor response complications in
Neurology 40: 467470. Parkinsons disease: hypotheses on the why, where, and
Swinn L, Schrag A, Viswanathan R et al. (2003). Sweating what. Mov Disord 15: 38.
dysfunction in Parkinsons disease. Mov Disord 18: Vincken WG, Gauthier SG, Dollfuss RE et al. (1984). Invol-
14591463. vement of upper-airway muscles in extrapyramidal dis-
Syed N, Murphy J, Zimmerman TJr et al. (1998). Ten years orders. A cause of airflow limitation. N Engl J Med 311:
experience with enteral levodopa infusions for motor fluc- 438442.
tuations in Parkinsons disease. Mov Disord 13: 336338. Volkmann J, Allert N, Voges J et al. (2004). Long-term
Tetrud JW, Koller WC (2004). A novel formulation of sele- results of bilateral pallidal stimulation in Parkinsons
giline for the treatment of Parkinsons disease. Neurology disease. Ann Neurol 55: 871875.
63 (7 Suppl 2): S2S6. Waters CH, Sethi KD, Hauser RA et al. (2004). Zydis
Togasaki DM, Tan L, Protell P et al. (2001). Levodopa Selegiline Study Group. Zydis selegiline reduces off time
induces dyskinesias in normal squirrel monkeys. Ann in Parkinsons disease patients with motor fluctuations:
Neurol 50: 254257. a 3-month, randomized, placebo-controlled study. Mov
Torstenson R, Hartvig P, Langstrom B et al. (1997). Differential Disord 19: 426432.
effects of levodopa on dopaminergic function in early and Wakabayashi K, Takahashi H, Ohama E et al. (1993). Lewy
advanced Parkinsons disease. Ann Neurol 41: 334340. bodies in the visceral autonomic nervous system in Parkin-
Tracey I, Becerra L, Chang I et al. (2000). Noxious hot and sons disease. Adv Neurol 60: 609612.
cold stimulation produce common patterns of brain activa- Weiner P, Inzelberg R, Davidovich A et al. (2002). Respira-
tion in humans: a functional magnetic resonance imaging tory muscle performance and the Perception of dyspnea in
study. Neurosci Lett 288: 159162. Parkinsons disease. Can J Neurol Sci 29: 6872.
184 S. H. FOX AND A. E. LANG

Winge K, Werdelin LM, Nielsen KK et al. (2004). Effects of by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP).


dopaminergic treatment on bladder function in Parkinsons Neuropharmacology 32: 315321.
disease. Neurourol Urodyn 23: 689696. Yoshimura N, Mizuta E, Yoshida O et al. (1998). Therapeutic
Witjas T, Kaphan E, Azulay JP et al. (2002). Nonmotor effects of dopamine D1/D2 receptor agonists on detrusor
fluctuations in Parkinsons disease: frequent and disabling. hyperreflexia in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyri-
Neurology 59: 408413. dine-lesioned parkinsonian cynomolgus monkeys. J Phar-
Yeh KC, August TF, Bush DF et al. (1989). Pharmacoki- macol Exp Ther 286: 228233.
netics and bioavailability of Sinemet CR: a summary of Zappia M, Oliveri RL, Montesanti R et al. (1999). Loss of
human studies. Neurology 39 (11 Suppl 2): 2538. long-duration response to levodopa over time in PD:
Yoshimura N, Mizuta E, Kuno S et al. (1993). The dopamine implications for wearing-off. Neurology 52: 763767.
D1 receptor agonist SKF 38393 suppresses detrusor Zenzola A, Masi G, De Mari M et al. (2003). Fatigue in
hyperreflexia in the monkey with parkinsonism induced Parkinsons disease. Neurol Sci 24: 225226.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 40

Levodopa-induced dyskinesias in Parkinsons disease

JOSE A. OBESO1*, MARCELO MERELLO2, MARIA C. RODRIGUEZ-OROZ1, CONCEPCIO MARIN3,


JORGE GURIDI1 AND LAZARO ALVAREZ4

1
Department of Neurology and Neurosurgery, University Clinic and Medical School and Neuroscience Division,
University of Navarra and CIMA, Pamplona, Spain
2
Movement Disorders Section, Raul Carrea Institute for Neurological Research, FLENI, Buenos Aires, Argentina
3
Laboratori de Neurologia Experimental, Fundacio Clnic-Hospital Clnic, Institut dInvestigacions Biomediques August Pi i
Sunyer (IDIBAPS), Hospital Clnic, Barcelona, Spain
4
Movement Disorders Unit, Centro Internacional de Restauracion Neurologica (CIREN), La Habana, Cuba

40.1. Introduction biochemical studies have been conducted with tissue


derived from well-characterized PD patients and the
Hyperkinetic or dyskinetic disorders are characterized revitalization of surgery has permitted the recording
by excessive motor activity that interferes with normal of BG activity in vivo. Thus, a wealth of information
motor control mechanism. The early literature during is now available to put together findings in animal mod-
the 1950s and 1960s used the term hyperkinesia els and in patients, leading to a more comprehensive
to designate wild involuntary movements like chorea, understanding of LID.
ballism, choreoathetosis or action tremor, all of which This review will generically concentrate on LID but
are currently embraced under the global denomina- dopamine agonists are also capable per se of inducing
tion of dyskinesias This comprises chorea and ball- dyskinesias, as shown in MPTP monkeys and de novo
ism, dystonic movements and postures, myoclonus, PD patients.
tics and tremor. The capacity for levodopa to induce
involuntary movements (i.e. levodopa-induced dyski-
nesias or LID) never witnessed before in Parkinsons 40.2. Clinical presentation
disease (PD) patients was established soon after its
introduction in the early 1970s (Cotzias et al., 1969; LID generally occur in patients with motor fluctua-
Barbeau, 1976). tions (i.e. wearing-off or end-of-dose deterioration)
LID are associated with several molecular, biochem- and are therefore better described in relation to the
ical and physiological changes in the basal ganglia motor state achieved in response to levodopa (Marsden
(BG). Among these, molecular and neuronal firing are and Parkes, 1977; Marsden et al., 1981; Obeso et al.,
the ones that have been studied in greater depth. It has 1989; Fahn, 2000). Accordingly, LID may be divided
not yet been possible to decipher which changes into three main groups: (1) peak-dose choreic or dys-
are compensatory, causally linked to LID or induced tonic movements; (2) diphasic dyskinesias; and (3)
as a consequence of the involuntary movements. off-period dystonia. This is a convenient classification
Animal models of PD, mainly the 6-hydroxydopamine that relates phenomenology (i.e. type of dyskinesia) and
(6-OHDA) lesioned rat and the 1-methyl-4-phenyl- temporal onset with the degree of dopaminergic activity
1,2,3,6-tetrayhydropyridine (MPTP)-intoxicated mon- and motor state (Fig. 40.1), thus allowing the character-
key, have been used to elicit LID, allowing the study ization of the underlying pharmacological origin of the
of biochemical changes and modifications of neuro- dyskinesia by clinical inspection. Admittedly, in a large
nal firing activity in the BG. More recently, a few proportion of patients, more than one type of LID is

*Correspondence to: Jose A. Obeso, Neurologia, Clinica Universitaria, Avenida Pio XII 36, Pamplona 31180, Spain. E-mail:
jobeso@unav.es, Tel: 34-948-255400, Fax: +34-948-194700.
186 J. A. OBESO ET AL.

Fig. 40.1. Relationship between onset and type of levodopa-induced dyskinesia and the dopaminergic response. Off-period
dystonia and diphasic dyskinesias occur while dopaminergic activity is low and should, therefore, disappear when higher levels
are reached. In contrast, peak-dose chorea is related to excessive dopaminergic stimulation.

present and the assessment and pharmacological solution DID for dystoniaimprovementdystonia but the
are more problematic. movements are also frequently labeled as ballistic or
choreic. In reality, they typically consist of repetitive
40.2.1. Choreic and dystonic movements stereotypic flexor and extensor movements of the
lower limbs (Fig. 40.2) due to alternating contractions
Choreic and dystonic movements occur during the per- of agonist and antagonist muscles (Obeso et al., 1989;
iod of peak plasma level and maximal clinical benefit Luquin et al., 1992b). This well-established pattern is
(on dyskinesias or peak-dose dyskinesias). On-period incompatible with the definitions of chorea or dysto-
dyskinesias are associated with levodopa in a dose- nia. In severe patients, the repetitive pattern of muscle
related manner. Typically, movements are choreiform contraction may become disrupted and reach large
and involve the face, neck and proximal upper limbs. amplitude, giving rise to a more disorganized move-
A smaller proportion of patients exhibit other move- ment, resembling ballism.
ment disorders such as ballism, myoclonic jerks and
tic-like movements. Involuntary upward saccadic eye 40.2.3. Off-period dystonia
movements are seen in a minority of patients.
Dystonic postures are due to sustained co-contraction of
40.2.2. Diphasic dyskinesias agonist and antagonist muscles, twisting the body into
fixed postures. Off-period dystonia commonly consists
These are repetitive movements, usually of the legs, of focal dystonic postures of a limb, such as extension
that occur at the beginning and end of the levodopa of the foot and big toe or flexion of the toes, but may
dose effect (diphasic) and disappear or transform into also be generalized. Dystonic postures may also have
a choreic (peak-dose) pattern during the on period a diphasic evolution (Luquin et al., 1992b) and may be
(Muenter et al., 1977). Diphasic dyskinesias are asso- the initial manifestation of PD, before treatment is intro-
ciated with low or intermediate levodopa plasma duced. In fact, dystonic postures in PD were well recog-
levels that are insufficient to achieve a full on anti- nized before levodopa became available. Nowadays,
parkinonian effect. Labeling and classifying diphasic however, they are more frequently seen and characteris-
dyskinesias have been permanent sources tic of off periods and therefore readily subside when
of confusion. Muenter et al., (1977) applied the term the on response ensues.
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 187

Fig. 40.2 Phenomenology of diphasic dyskinesias. Movements typically consist of repetitive, kicking of the leg (left). The
characteristic electromyogram pattern, recorded with surface electromyographic electrodes, consists of alternating contraction
of antagonist muscles of the lower limb (right). Note the typical 5 Hz resting tremor in the upper limb. ff, finger flexors; fe,
finger extensors; quad, quadriceps; ham, hamstrings; gastroc, gastronemius; tib ant, tibialis anterior muscles. Modified from
Obeso et al. (1989) and Luquin et al. (1992).

40.2.4. Special presentations 40.2.4.3. Dyskinesias without benefit


Dyskinesias without benefit were described in the
There are some special circumstances when the clinical 1970s and early 1980s but are not so frequently
presentation of LID does not follow the typical patterns encountered nowadays. In this situation patients suffer
described above (see below). dyskinetic movements without having the antiparkin-
sonian benefit of levodopa, similar to off dyskine-
40.2.4.1. Nocturnal myoclonus sias. However, we believe from our experience with
Spontaneous myoclonic jerking of the leg at night occurs chronic infusion of dopamine agonists that such pre-
in the normal population but more frequently in patients sentation mainly corresponds to diphasic dyskinesias
with PD treated with dopaminergic drugs and usually without the characteristic short-lasting and beginning
coincides with nightmares and other parasomnias. The and end-of-dose dual presentation.
importance of nocturnal myoclonus, besides its potential
inconvenience for the spouse, is that it generally heralds 40.2.4.4. Graft-induced off dyskinesias
the beginning of psychiatric complications. It therefore This is a new problem in the field of dyskinesias in
demands early recognition and treatment. PD. Focal or segmental dyskinesias similar to typical
LID have now been reported in two transplantation
40.2.4.2. Dyskinesiasparkinsonism studies and were not observed in any of the placebo-
In patients with severe PD who require large doses of treated patients (Freed et al., 2001; Olanow et al.,
levodopa, motor fluctuations and dyskinesias may not 2003). These dyskinesias thus appear to be specifically
strictly follow the typical patterns described above. The related to the grafted cells. Dyskinesias can persist for
combination of one body part exhibiting dyskinesias days or even weeks following withdrawal of dop-
while the other half is off is one of the most troublesome aminergic medication, hence the use of the term off-
presentations. Typically, the upper half of the body medication dyskinesias. However, careful evaluation
exhibits dyskinesias and coincidentally the patient is indicates that in fact patients are not really off as com-
incapable of walking due to severe freezing of gait. pared to their baseline (pregrafting) state. Indeed, in the
188 J. A. OBESO ET AL.
study by Freed et al. (2001), those patients who dev- The D1 subtype of dopamine receptor was found to
eloped dyskinesias after transplantation greatly im- be increased in the 6-OHDA rat model (Gerfen et al.,
proved, reaching motor scores comparable to those 1990). However, no marked changes induced by levo-
present during on dyskinesia. On the other hand, pat- dopa were encountered in MPTP monkeys and PD
ients in the study by Olanow et al. (2003) showed a very patients (Lee et al., 1978; Graham et al., 1993). Recent
modest improvement and the clinical features and evo- findings suggest that LID may be more related to
lution of the dyskinesias were more compatible with a changes at the level of D1-receptor transmission. There
diphasic-like pattern. is now evidence that downstream D1-mediated signal
transduction cascade (Gerfen, 1995) is abnormal in
40.3. Biochemical and molecular LID. These include increased phosphorylation of cyclic
characteristics adenosine monophosphate-regulated phosphoprotein of
32 kDa (DARPP-32) (Picconi et al., 2003), an increase
Striatal dopaminergic depletion and the discontinuous in D1-agonist-induced G-protein-coupled receptor
or pulsatile activation associated with levodopa ther- kinases and high levels of cyclin-dependent kinase-5
apy are the two major factors determining the develop- (Cdk5) (Aubert et al., 2005; Guigoni et al., 2005a). These
ment of dyskinesias. Together they induce alterations data suggest that LID are closely related to augmented
in the expression of dopamine receptors and in the vast D1-receptor-mediated transmission at the level of the
majority of neurotransmission systems acting at the direct pathway. In addition, D3-receptors may play a
striatal level. The following section summarizes the role in LID. This was initially proposed based on the
major findings. observation that striatal expression of this receptor is
highly dependent on afferent dopamine innervation
40.3.1. Dopaminergic receptors (Sokoloff et al., 1990; Levesque et al., 1995). Thus,
D3-receptor expression is reduced in the striatum of
There are five subtypes of dopaminergic receptors parkinsonian rats and in the caudate nucleus of MPTP
grouped into two main families. D1 and D5 subtypes monkeys (Levesque et al., 1995; Bordet et al., 1997).
constitute the D1 family of dopaminergic receptors Levodopa compensates for this reduction (Morissette
and D2, D3 and D4 subtypes form the D2 family. Many et al., 1998; Quik et al., 2000, Bezard et al., 2003) but
studies have been focused on possible changes in the D3 putaminal receptors are only increased in animals
dopaminergic receptors in the striatum of parkinsonian that developed LID (Bordet et al., 2000; Bezard et al.,
and dyskinetic PD patients and in a possible differen- 2003). Importantly, the overexpression of D3-receptor
tial role of these receptor subtypes in the genesis of was confined to medium spiny striatal neurons, giving
LID. It has been proposed that LID surges as an imbal- rise to the direct striatopallidal projection (Bordet
ance in activity of the two striatal output pathways, et al., 2000). These findings have led to suggest an
possibly through activation of D1 and inhibition of important role of D3 activation and the direct pathway
D2-receptors on the direct and indirect pathways, in the pathophysiology of dyskinesias. On the other
respectively (Obeso et al., 2000b; Bezard et al., hand, other studies have failed to define significant
2001). Despite numerous experimental investigations, abnormalities of D3-receptors in LID in both the MPTP
however, a clear relationship between dyskinesia and monkey model (Hurley et al., 1996a) and PD patients
D1- or D2-receptor expression is not established. (Hurley et al., 1996b) and pharmacological studies
Numerous studies have shown that dopamine using the partial D3-agonist BP 897 induced reduction
denervation causes an increase in striatal D2-receptors of LID, but at the expense of increased parkinsonism
in the postmortem tissue of untreated PD patients and (Hsu et al., 2004).
animal models of parkinsonism (Lee et al., 1978;
Bezard et al., 2001; Aubert et al., 2005; Guigoni 40.3.2. Glutamatergic receptors
et al., 2005a). Chronic levodopa treatment induces a
normalization of the D2-receptor in patients with PD, Striatal medium spiny output neurons receive massive
as assessed in postmortem tissue (Lee et al., 1978) glutamatergic inputs from the cerebral cortex and the
and by positron emission tomography (PET) (Brooks thalamus and are the site of close interaction between
et al., 1992; Shinotoh et al., 1993) and in MPTP dopamine and glutamate receptors (Calabresi et al.,
monkeys (Graham et al., 1993; Herrero et al., 1996a). 2000). Accordingly, striatal glutamate receptors have
However, no difference in the expression of D2-receptors been suspected to be implicated in the development of
was observed between non-dyskinetic and dyskinetic LID (Chase and Oh, 2000; Calabresi et al., 2000; Calon
groups of MPTP-treated monkeys (Aubert et al., 2005; et al., 2002). The actions of glutamate on target neurons
Guigoni et al., 2005a). are mediated by four classes of receptors: three types
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 189
of channel-forming ionotropic receptors and one group Chronic levodopa treatment in 6-OHDA-lesioned
of metabotropic, or G-protein-coupled, glutamate recep- rats, eliciting behavioral sensitization, increases NR1,
tors (Hollmann and Heinemann, 1994; Dingledine et al., NR2A and NR2B subunits in homogenate and synap-
1999). tosomal membrane fractions (Oh et al., 1997; Dunah
et al., 2000; Dunah and Standaert, 2001). In MPTP
40.3.2.1. Glutamatergic ionotropic receptors monkeys, levodopa treatment causing dyskinesias
In the striatum, the majority of fast excitatory synaptic restored or increased the NR2B levels in synaptosomal
transmission is mediated by ionotropic glutamate recep- membranes but in addition increased the abundance of
tors, which are ligand-gated ion channels. These recep- the NR2A subunit (Calon et al., 2002; Hallett et al.,
tors are named according to the synthetic agonists that 2005). Similar studies from postmortem tissue of
are most effective in activating them and classified into patients (treated chronically with levodopa) showed
N-methyl-D-aspartate (NMDA), a-amino-3-hydroxy-5- unchanged (Calon et al., 2003a) or increased (Lange
methylisoxasole-4-propionate (AMPA) and kainate et al., 1997) NR2A binding and increased NR2B
receptors (Dingledine et al., 1999). Striatal dopamine binding (Calon et al., 2003a).
depletion increases the concentration and release of glu- Phosphorylation of ion channels plays a role in
tamate from corticostriatal terminals (Lindefors and mediating synaptic plasticity and serves to increase
Ungerstedt, 1990) but neither intrinsic membrane prop- molecular and functional heterogeneity of NMDA
erties nor the response to exogenous application of glu- receptors (Betarbet et al., 2004). In animal studies
tamate agonists are altered in medium spiny neurons there is a controversy over the phosphorylation state
(Calabresi et al., 1993, 2000). of these receptors either in the parkinsonian state or
in the levodopa-treated condition. Thus, a number of
changes in the rat parkinsonian model, such as
40.3.2.1.1. NMDA receptors increase in the proportion of NR2B subunits that are
NMDA receptors are the more densely expressed sub- tyrosine-phosphorylated (Menegoz et al., 1995; Oh
type in the striatum and their synaptic localization et al., 1997), reduced serine phosphorylation of NR1
appears to be exclusively on the dendritic spines of subunit (Dunah et al., 2000) or increased serine phos-
the medium spiny gamma-aminobutyric acid (GABA)- phorylation of NR2A but not NR2B receptor subunit
ergic neurons. The NMDA receptor is a heterodimeric (Oh et al., 1997), have been described. Chronic levo-
protein essentially composed of two distinct subunits, dopa treatment enhanced serine phosphorylation of
namely NR1 and NR2 (Monyer et al., 1992). The NR1 and NR2A subunits and increasing tyrosine phos-
expression of at least one NR1 subunit is required to phorylation of NR2A and NR2B subunits (Oh et al.,
form a functional channel and the combinations of dis- 1997; Dunah et al., 2000). However, these alterations
tinct NR1 and NR2 subunits confer diversity of NMDA have not been confirmed in parkinsonian MPTP and
receptors. In the striatum, the NMDA receptor subtype dyskinetic monkeys (Hallett et al., 2005).
containing NR2B is predominantly present (Standaert The efficacy of antagonists of the NMDA receptors
et al., 1994). in animal models of PD has been widely examined.
Dopamine depletion in animal models (6-OHDA These studies have revealed the ability of NMDA-
and MPTP monkeys) induces a redistribution of receptor blockade to alleviate the symptoms of parkin-
NMDA receptor subunits in two biochemical separate sonism and augment the effectiveness of dopaminergic
compartments. Thus, in unfractionated homogenates therapy preventing or reversing the induction of LID.
prepared from striatal tissue, the abundance of NR1, For example, the competitive antagonist LY345959
NR2A and NR2B subunit proteins was unchanged and the NR2B-selective antagonists Co101244 and
(Menegoz et al., 1995; Dunah et al., 2000; Calon CI-1041 administered as coadjutants to levodopa
et al., 2002) or minimally increased for the NR2A sub- diminished LID (Papa and Chase, 1996; Blanchet
unit proteins (Oh et al., 1997). In contrast, in striatal et al., 1999; Had Tahar et al., 2004) and CI-1041
synaptosomal membrane fractions, dopamine deple- prevented the development of LID in MPTP monkeys
tion results in decreased NR1 and NR2B subunits (Morissette et al., 2006).
(Dunah et al., 2000; Hallett et al., 2005) and no change In summary, NMDA receptor subunit expression is
in the NR2A subunit. It has been suggested that an altered in both the parkinsonian and dyskinetic states
underlying mechanism for this distribution may be a in the MPTP monkey model and PD patients. These
rapid D1-receptor- and tyrosine kinase-dependent traf- changes may mediate a putative deregulation of corti-
ficking system, which regulates delivery of NMDA costriatal activity underlying LID. The involvement of
receptor to synaptic sites in striatal neurons (Dunah D1 dopamine receptor activation in the cascade of
et al., 2004). intracellular changes summarized above and the fact
190 J. A. OBESO ET AL.
that the NR2A subtype of receptor is mostly expressed transmission in PD (Gubellini et al., 2004). Modulation
in striatal neurons projecting to the substantia nigra of these receptors can alleviate some parkinsonian
pars reticulata (SNpr) in the rat are in keeping with motor features in the 6-OHDA rat (Murray et al.,
the suggested pivotal role of the direct pathway in 2002; Picconi et al., 2002). However, there are no
the pathophysiology of LID (Bezard et al., 2003; direct data at present regarding the possible involve-
Aubert et al., 2005). ment of these receptors in the pathophysiology of LID.
Future research on the possible role of mGluR on the
40.3.2.1.2. AMPA receptors development of dyskinesias is clearly warranted.
The role of AMPA receptors in LID is still controver-
sial. AMPA receptors in the striatum of MPTP monkeys 40.3.3. Neuropeptides and adenosine receptors
with LID have been reported as normal (Silverdale 40.3.3.1. Enkephalin, dynorphin and substantia P
et al., 2002) or moderately increased (Calon et al.,
Enkephalin, dynorphin and substantia P are opiod pep-
2002). No alterations were found in the abundance of
tides acting as cotransmitters in the striatopallidal sys-
striatal GluR2/3 AMPA receptor subunits in synaptoso-
tem. The striatal GABAergic projection neurons are
mal membrane fractions following MPTP lesion and
segregated with respect to the opioid peptides used as
repeated levodopa treatment (Hallett et al., 2005). The
cotransmitters. Thus, striatal neurons projecting to the
absence of changes in AMPA receptor subunit proteins
globus pallidum externa (GPe) in the indirect pathway
in monkeys confirmed previous studies in 6-OHDA-
express enkephalin derived from the precursor pre-
lesioned rats (Dunah et al., 2000; Picconi et al., 2004). proenkephalin A (PPE-A), whereas striatal neurons
In postmortem tissue from levodopa-treated parkinso-
projecting to the globus pallidum interna (GPi) and
nian patients, no change of AMPA binding was
SNpr in the direct pathway express dynorphin derived
observed (Ulas et al., 1994) but recently, a small eleva-
from PPE-B (Henry et al., 1999; Steiner and Gerfen,
tion in AMPA receptors restricted to the lateral putamen
1999) (Fig. 40.3). Dopamine modulates the activity
has been reported (Calon et al., 2003a). The notion that
of striatal neurons and the expression of these neuro-
AMPA receptor-mediated mechanisms are involved in
peptides. Dopamine depletion increases PPE-A mRNA
LID is further suggested by the observation that
and enkephalin peptide levels and reduces PPE-B
AMPA-receptor antagonists can reduce the expression mRNA and dynorphin. These findings have been well
of LID when administered as an adjuvant to levodopa
established in rat and monkey models as well as in tis-
in the MPTP monkeys (Konitsiotis et al., 2000).
sue from patients with PD (Augood et al., 1989; Ger-
fen et al., 1990; Asselin et al., 1994; Herrero et al.,
40.3.2.2. Glutamatergic metabotropic receptors 1995; Nisbet et al., 1995).
In contrast to ionotropic glutamate receptors, the meta- Chronic treatment with levodopa increases PPE-B
botropic glutamate receptors (mGluRs) are coupled to mRNA over basal levels but has little or no effect on
second-messenger systems through G-proteins. They the elevated levels of PPE-A mRNA expression (Her-
have been classified into three groups: group I (mGluR1 rero et al., 1995; Nisbet et al., 1995; Cenci et al., 1998;
and mGluR5), group II (mGluR2 and mGluR3) and Henry et al., 1999; Tel et al., 2002). In LID, PPE-A is
group III (mGluR4, 6, 7 and 8) and have been identified either unchanged or further increased whereas PPE-B
in the striatum. In particular, group I mGluRs have been is increased (Herrero et al., 1995; Morissette et al.,
localized throughout the whole striatum at postsynaptic 1997; Henry et al., 1999; Calon et al., 2000; Zeng
level, whereas group II and III mGluRs have been found et al., 2000; Tel et al., 2002; Henry et al., 2003). Inter-
at presynaptic level of corticostriatal terminals (Pisani estingly, increase of PPE-A mRNA expression in dys-
et al., 2002). Indeed, induction of plastic changes in cor- kinetic patients appears restricted to the lateral part of
ticostriatal synapse depends upon activation of metabo- the putamen, which receives dense projections from
tropic receptors. In particular, long-term potentiation the sensorimotor cortex. In humans, Piccini et al.
(LTP) is partially mediated by mGluR1 and mGluR5 (1997) found significantly reduced striatal and thala-
(Gubellini et al., 2004) and the activation of mGluR2/ mic opioid binding in dyskinetic but not in non-dyski-
3 is required for inducing long-term depression (LTD) netic PD patients. Unexpectedly, there has been no
(Sung et al., 2001). The meaning of these physiological follow-up study to gain further understanding of these
changes with respect to LID is discussed in more detail findings.
in section 40.4 below. Opioid receptor antagonists were shown to reduce
The experimental evidence suggests that metabotro- LID in MPTP monkeys (Henry et al., 2001) but clini-
pic receptors represent another important point in the cal studies have produced controversial results. Two
regulation of the excessive corticostriatal glutamatergic studies have reported an antidyskinetic action of the
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 191
non-selective opioid receptor antagonist naloxone variable and may not even be present in some instances
(Trabucchi et al., 1982; Sandyk and Snider, 1986), (Bove et al., 2005). A2A-receptor blockade did not
whereas oral administration of naltrexone, a longer- attenuate or prevent LID in 6-OHDA-lesioned rats
acting analog of naloxone, was ineffective in alleviating (Lundblad et al., 2003). Similarly, it did not modify
levodopa or apomorphine-induced dyskinesias (Rascol the upregulation in prodynorphin elicited by chronic
et al., 1994; Manson et al., 2001). None of these studies levodopa treatment in rodents (Lundblad et al., 2003).
were placebo-controlled.
40.3.3.3. Early genes
40.3.3.2. Adenosine receptors Early genes and transcription factors that regulate the
The adenosinergic activity in the striatum has also expression of other genes could also be involved in
been investigated in PD and LID. The A2A subtype the increment of PPE and A2A-receptor (Calon et al.,
of adenosine receptor is abundantly expressed in the 2000). PPE and A2A gene expression in LID may be
medium spiny neurons of the striatum (Martinez-Mir amplified by a common transcription factor such as
et al., 1991), particularly in dendrites, and appears to fosB, one of the most thoroughly studied factors. In
signal, in part, through activation of serine/threonine the striatum of the MPTP monkey there is an increment
kinases known to modulate the phosphorylation state in fosB not seen after chronic treatment with very
of ionotropic glutamate receptors (Swope et al., long-acting D2-agonists such as cabergoline or continu-
1999; Carvalho et al., 2000). Adenosine receptors are ous administration of the D1-agonist SKF-82958, which
mainly expressed in GABAergic neurons projecting provided sustained antiparkinsonian efficacy without
to the GPe (indirect pathway) and that also coexpress dyskinesias (Doucet et al., 1996). In animals receiving
PPE-A and D2-receptors (Svenningsson et al., 1998). pulsatile stimulation with the D1-agonist SKF-82958,
Dopaminergic lesion in the 6-OHDA rat model pro- dyskinesias appeared in 2 out of 3 monkeys within 1
duces either no change or 20% increase of A2A receptor week of administration along with a marked induction
in the striatum (Kaelin-Lang et al., 2000; Pinna et al., of fosB in the striatum of dyskinetic animals. Thus,
2002). In a recent study, enhancement of striatal A2A fosB is found to be increased in the striatum of dyski-
adenosine receptors was shown in 6-OHDA-lesioned netic monkeys (Doucet et al., 1996; Calon et al., 2000)
rats and also in MPTP monkeys with LID (Zeng et al., and rats (Cenci, 2002), suggesting it could be a key fac-
2000; Tekumalla et al., 2001), although the finding tor in the development of LID. Interestingly, adminis-
could not be replicated in squirrel monkeys (Quik tration of an antisense directed to fosB blocked LID
et al., 2002). In human studies, adenosine A2A recep- in the rat (Andersson et al., 1999). However, no differ-
tors are increased in the lateral putamen, without ences in fosB expression have been found between
changes in the caudate nucleus, of dyskinetic PD dyskinetic and non-dyskinetic PD patients (Tekumalla
patients compared with non-dyskinetic patients. A2A et al., 2001). Clearly, the meaning and functional
receptors are also increased in the GPe of parkinsonian changes of transcription factors in LID are not fully
patients with and without dyskinesia (Calon et al., understood at present.
2004). There was a positive correlation between the
expression of PPE-A and A2A receptors in the putamen 40.3.4. GABA receptors
of these patients, which represents the motor region of
the striatum (Calon et al., 2004). It is known that PPE- GABA is the major neurotransmitter of the BG. GABA
A is regulated by A2A receptor activity. Thus, A2A- receptors are classified into two major subtypes:
receptor antagonists reverse the PPE-A increment ionotropic (GABAA and GABAC) and metabotropic.
induced in the rat striatum by 6-OHDA lesions and GABAergic neurons are abundantly present in the BG,
dopaminergic drugs (Carta et al., 2002; Lundblad including striatal interneurons, medium spiny neurons,
et al., 2003). This has led to the suggestion that A2A- pallidal and SNpr efferent neurons. They are enriched
receptor activation could precede PPE-A increment by GABAA and GABAB receptor subtypes.
and play a prominent causative role in the origin of LID. GABAA receptors are increased in the GPi of dyski-
Pharmacological studies in animal models suggest netic MPTP monkeys and in PD patients with LID
that the A2A-antagonist KW-6002 has antiparkinso- (Calon et al., 1995, 1999, 2003b; Calon and Di Paulo,
nian activity and potentates the action of levodopa 2002). GABAA receptor upregulation in GPi correlates
(Kanda et al., 1998, 2000; Grondin et al., 1999; Lund- with the development of dyskinesias induced by D1-
blad et al., 2003; Pinna et al., 2005). A2A antagonism and D2-agonists in MPTP monkeys (Calon et al.,
attenuates the overactivity of the striatopallidal 1995, 1999). This was not the case in MPTP monkeys
pathway in the parkinsonian rat, but this effect is chronically treated with dopamine agonists which did
192 J. A. OBESO ET AL.
Motor Cortex

Thalamus

A NORMAL STATE

Motor Cortex

Thalamus

B PARKINSONISM
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 193

Fig. 40.3. Schematic representation of the main basal ganglia circuits. (A) Normal state; (B) Parkinsonian state due to dopa-
minergic striatal depletion. Dopamine depletion induces a dual, opposite effect, on medium spiny neurons. D1-expressing neu-
rons are inhibited whereas D2-containing neurons are overactive. (C) Levodopa-induced dyskinetic state in Parkinsons
disease. This represents the changes elicited by levodopa on a denervated striatum (summarized in B). SNpc-DA, substantia
nigra pars compactadopamine; NMDA, N-methyl-D-aspartate; SP, substance P.

not induce dyskinesias. The main changes in the ex- Under normal circumstances levodopa is decar-
pression of GABAA receptors occurred in the motor boxylated to dopamine in the striatal terminals of
(posteroventral) region of the GPi (Calon et al., 2002). nigrostriatal dopaminegic neurons. However, the site
On the other hand, no significant change was found in of levodopa decarboxylation in parkinsonism remains
GABAA receptors in the putamen of dyskinetic PD obscure, as most dopamine terminals have degen-
patients. Patients with LID had increased expression of erated. There is now evidence that 5-HT neurons are
GABAB receptors in the ventral GPe compared with primarily responsible for the storage and release of
non-dyskinetic patients, but in absolute terms there was levodopa-derived dopamine into the striatum where
no difference from control values (Calon et al., 2003b). dopaminergic terminals are lost (Tanaka et al., 1999;
Overall, these data suggest abnormal deviation of GABA Kannari et al., 2001). In animal models and in pati-
receptors in the motor area of both segments of the GP ents with PD striatal dopaminergic mechanisms can be
but the precise relationship with the parkinsonian and influenced by drugs that selectively interact with seroto-
dyskinetic states is not well depicted. ninergic neurons, including those that stimulate 5-HT1A
receptors (Santiago et al., 1998). Interestingly, it has been
40.3.5. Serotoninergic receptors shown that in 6-OHDA-lesioned rats, pretreatment with
the 5-HT1A receptor agonist 8-hydroxy-2-(di-n-propyla-
The BG receive a rich serotoninergic innervation from mino) tetralin (8-OH-DPAT) attenuates the excessively
the dorsal and medial raphe nuclei which could play a high extracellular dopamine levels that occur in the
role in their motor and behavioral functions (Jacobs dopamine-denervated striatum after levodopa treatment
and Fornal, 1993). The action of serotonin (5-hydroxy- (Kannari et al., 2001). This effect was mediated by stimu-
tryptamine: 5-HT) is mediated by a variety of 5-HT- lation of 5-HT1A-receptors since the co-administration of
receptors, including G-protein-coupled subtypes (5-HT1, WAY-100635, a 5-HT1A-receptor antagonist, attenuated
5-HT2, 5HT47) and a ligand-gated ion channel (5-HT3) the effect of 8-OH-DPAT on dopamine levels (Kannari
(Hoyer et al., 2002). et al., 2001).
194 J. A. OBESO ET AL.
Based on these observations, it has been suggested tions in animal models of PD and dyskinesias
that the administration of 5-HT1A-receptor agonists (Crossman, 1987; Albin et al., 1989; DeLong, 1990).
with levodopa may prevent the deleterious effects of The main idea underlying the model is that corticos-
pulsatile stimulation of dopamine receptors associated triatal afferent activity influences the output of the
with levodopa treatment and reverse LID. Recent BG, the GPi and SNpr, by two relatively segregated
results have shown that the co-administration of striopallidal projections: the indirect pathway and the
8-OH-DPAT prevents behavioral sensitization to levo- direct pathway. The indirect pathway or circuit con-
dopa in the 6-OHDA model (Tomiyama et al., 2005), sists of GABA medium spiny striatal neurons expres-
in association with a decrease in the striatal expression sing D2 and enkephalin and inhibiting the GPe,
of mRNA coding for dynorphin and acid decarboxylase, which in turn inhibits the STN. The STN projection
both of which are established markers of levodopa- is excitatory, i.e. glutamatergic, onto the GPi and
induced motor complications (Tomiyama et al., 2005). SNpr. The direct pathway also arises from GABA
Moreover, the 5-HT1A agonists buspiron and sarizotan striatal neurons bearing D1-receptors and expressing
have been found to reduce LID in two open trials with substance P and dynorphin and projecting monosynap-
PD patients (Bonifati et al., 1994; Bara-Jimenez et al., tically to the GPi/SNpr. These functional arrangements
2005) and MPTP monkeys (Bibbiani et al., 2001). This sustain a dual functional effect of dopamine on striatal
therapeutic effect was not associated with a reduction of medium spiny neurons and on the output of the BG
levodopa efficacy. Moreover, in 6-OHDA-lesioned rats, (Fig. 40.3A). Thus, increased activity in the direct path-
sarizotan reversed the shortening in motor response way and indirect pathway produces inhibition and
duration induced by intermittent administration of excitation respectively of GPi/SNpr neurons. Pauses
levodopa (Bibbiani et al., 2001). of neuronal activity in the GPi/SNpr are associated
The clinical effects observed with sarizotan are most with movement facilitation (Hikosaka and Wurtz,
likely related to the potent ability of the drug to activate 1983; DeLong, 1990) whereas firing is more related
5-HT1A-receptors since its effects disappeared when the to the end and halting of movement (Brotchie et al.,
5-HT1A antagonist WAY-100635 was co-administered 1991; Mink and Thach, 1991). Dopamine striatal defi-
in rat and monkey models (Bibbiani et al., 2001). ciency secondary to loss of the nigrostriatal projection
Other potential mechanisms for the antidyskinetic leads to increased activity in GABA medium spiny
effect of sarizotan may also be taken into account. In neurons expressing D2 and enkephalin and overinhibi-
principle, sarizotans ability to diminish dyskinesias tion of the GPe, resulting in exaggerated activity of the
does not seem to be related to its capability to block STN in the indirect pathway (Fig. 40.3B). In the direct
D2, D3 or D4 dopaminergic receptors (Rabiner et al., pathway dopamine deficiency induces a reduction of
2002; Bartoszyk et al., 2004). Recently, the effects of activity in GABAergic neurons bearing D1-receptors
sarizotan on the corticostriatal glutamate pathways and expressing substance P and dynorphin (Gerfen
have been investigated and the results indicate that et al., 1990). Together, these changes in the indirect
sarizotan produces a reduction in cortical and striatal and direct pathways combine to produce a state of
glutamate levels when systemically administered or abnormally increased activity in the inhibitory output
perfused into the motor cortex (Antonelli et al., 2005). of the BG (Fig. 40.3B), which is a main pathophysiolo-
Of particular interest are recent reports that indicate gical feature of the parkinsonian state. The opposite
the influence of 5-HT-receptors on subthalamic nucleus functional picture (Fig. 40.3C), on the other hand,
(STN) activity. The intrasubthalamic administration of characterizes dyskinesias.
5-HT-induced contralateral rotational behavior sug- Initial evidence indicated a paramount role of the
gests an inhibitory effect of 5-HT on STN neurons indirect pathway in the induction of dyskinesias. In nor-
(Bellforte and Pazo, 2004). Furthermore, extracellular mal monkeys, choreic dyskinesias are produced by dis-
single-unit recordings in mouse brain slices demon- inhibition of the GPe and blockade or lesion of the STN
strates that 5-HT elicits two distinct effects in STN neu- (Carpenter et al., 1950; Crossman et al., 1988; Hamada
rons, the first being an excitation and the second an and DeLong, 1992). Thus, blockade of the striato-GPe
inhibition; the latter effect is attenuated by the adminis- inhibitory projection (with bicuculline) leads to
tration of WAY-100635, indicating a 5-HT1A-receptor- increased GPe efferent activity, which results in overin-
mediated inhibition (Stanford et al., 2005). hibition of the STN (Crossman et al., 1988; Mitchell
et al., 1989, Grably et al., 2004), and reduction in STN
40.4. Pathophysiology glutamatergic efferent activity induces hypoactivity of
40.4.1. The classic model the GPi, both causing dyskinesias (Mitchell et al.,
1989; Robertson et al., 1989; Hamada and DeLong,
In the late 1980s a pathophysiological model of the 1992). Hypoactivity of the GPi may also occur by
BG was established (Figure 40.3) based on observa- increased GABAergic input from the GPe and the direct
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 195
striatopallidal pathway. Direct administration of the ingly, abnormal synaptic plasticity at corticostriatal
GABA agonist muscimol into the GPi has mainly been synapses may play a role in the pathophysiology of
associated with dystonic co-contraction of antagonist parkinsonism and LID (Calabresi et al., 1996, 2000;
muscles and secondary slowness of movement initiation Picconi et al., 2003; Pisani et al., 2005).
(Mink and Thach, 1991; Kato and Kimura, 1992; Inase In parkinsonian rats after dopaminergic denervation
et al., 1996). There is only one report of choreic dyski- with 6-OHDA or in animals treated with a D1-selective
nesias of the contralateral limbs provoked by overinhi- antagonist, LTP is blocked (Calabresi et al., 2000; Pisani
bition (with muscimol) of the GPi (Burbaud et al., et al., 2005). Chronic levodopa treatment restores this
1998). In addition, permanent lesion of the GPi (Horak response in dyskinetic and non-dyskinetic animals. After
and Anderson, 1984; Mink and Thach, 1991) is not induction of LTP, the delivery of low-frequency stimula-
associated with any dyskinetic movement; on the con- tion in normal rats produces a reversal of synaptic
trary (see below) it eliminates dyskinesias. strength to pre-LTP levels, named depotentiation. Par-
Hypoactivity of the GPi leads to disinhibition of kinsonian rats chronically treated with levodopa but
the thalamocortical projection in the motor circuit, without dyskinesias exhibited the same response as
releasing the motor thalamic nuclei and thereby facil- normal rats, whereas dyskinetic rats did not show the
itating the abnormal recruitment of cortical motor depotentiation response (Picconi et al., 2003). Thus,
areas and the development of involuntary move- LTP cannot be reversed by low-frequency stimulation
ments (Crossman, 1987; Albin et al., 1989; DeLong, in the 6-OHDA animals that developed LID after chronic
1990; Obeso et al., 2000b). This summary reflects treatment, in contrast to levodopa-treated rats without
the underlying notion of the classic model. A similar LID. Synaptic depotentiation is thought to increase the
sequence of events is supposed to take place due to efficiency of information storage in neuronal networks
excessive striatal dopaminergic stimulation in the and may play a prominent role in eliminating irrelevant
levodopa-treated parkinsonian state (Crossman, 1990; information underlying the process of forgetting. Thus,
Obeso et al., 2000b). In recent years, newer evidence the lack of depotentiation in the dyskinetic animals can
has arisen which has forced the revision and adapta- represent a failure in erasing inadequate motor informa-
tion of several assumptions of the original model. tion, leading to the development of abnormal involuntary
We discuss in detail all these different developments movements (Picconi et al., 2003).
in the subsections below. Striatal LTP mainly depends on activation of
NMDA and D1-receptors (Calabresi et al., 1996) but
40.4.2. Synaptic plasticity it is also modulated by mGluR2/3 metabotropic recep-
tors and endocannainoids (Genderman et al., 2002). As
LTP and LTD are forms of synaptic plasticity that may already indicated in section 40.3 above, NR2A recep-
be critically involved in learning and memory pro- tors are overexpressed in the striatum of MPTP mon-
cesses and are mediated by glutamatergic NMDA keys with LID (Calon et al., 2002; Hallett et al.,
receptors. In addition to the hippocampus, where it 2005) whereas NR1 and NR2B are normalized by
was described for the first time, this phenomenon is levodopa treatment. This set of changes could be
also present in a variety of brain structures, including responsible for a reinforcement of LTP leading to aug-
the striatum. Repetitive stimulation of the corticostria- mented threshold for its reversion, or depotentiation, in
tal pathway can induce both LTP (in response to the dyskinetic animals.
high-frequency stimulation) and LTD (in response to Dendritic spines are the locus of the interaction
low-frequency stimulation) of synaptic transmission between glutamate and dopamine responsible for the
in the striatum. Glutamatate released by corticostriatal LTP and LTD. It could be that dopaminergic deple-
stimulation excites striatal medium spiny neurons, tion induces a redistribution of the subtypes of NMDA
which are modulated by the nigrostriatal dopaminergic receptors, along the cellular membrane, that might
projection. Dendritic spines of striatal neurons are the cause an impairment of corticostriatal synaptic plasti-
anatomical locus of interaction between glutamate city. An increase in the percentage of glutamatergic
and dopamine with the dopaminergic terminal loca- synapses has been found (Anglade et al., 1996) in
lized at the necks and the glutamatergic terminals at the striatum of rats with 6-OHDA-induced lesion
the head of the spines respectively (Smith et al., and in PD patients. Moreover, the same studies have
1996). LID exhibit some features that are highly sug- shown that dendritic spines of striatal neurons are
gestive of plastic changes. The gradual development, reduced numerically and show abnormal length, size
persistency and dependence of glutamate receptor and shape (Anglade et al., 1996; Meshul et al.,
mechanism suggest that LID may be aberrant forms 1999; Stephens et al., 2005). At present, there are no
of motor learning (Centonze et al., 1999). Accord- data either in the MPTP monkey model or in PD
196 J. A. OBESO ET AL.
patients regarding the synaptic or extrasynaptic alloca- GPe neuronal activity and the presence of LID are
tion of the NR2A and NR2B subtypes of NMDA strikingly limited (Filion et al., 1991).
receptors in the dyskinetic state. The exact meaning Firing rate in the STN and GPi is therefore, appar-
of the changes so far encountered needs further stu- ently well correlated with the parkinsonian and dyski-
dies. In perirhinal cortex, the NR2B NMDA receptors netic motor states (Fig. 40.4). At odds with the model
responsible for LTD are allocated extrasynaptically, is the fact that the lesion of the GPi (pallidotomy),
whereas NR2A NMDA receptors, responsible for instead of aggravating LID, is associated with the
LTP and depotentiation, are located synaptically suppression of these undesired movements. A more
(Massey et al., 2004). Thus, it is possible that LID detailed analysis of neuronal activity in the GPi dur-
in PD could similarly involve relocation of NR2 ing LID has shown that not every single neuron
receptors underlying the formation of abnormal decreased its firing rate during LID (Filion et al.,
changes in synaptic plasticity. This could be a promi- 1991; Boraud et al., 2001; Levy et al., 2001). Simi-
nent feature of striatal physiology and LID. larly, in the STN (Levy et al., 2001), there was a
reduction in the overall firing rate of the nucleus but
40.4.3. Neurophysiology: neuronal firing a proportion of neurons had no change. Importantly,
the proportion of neurons discharging in bursts and
The parkinsonian state is associated with increased fir- irregularly is increased in the STN and GPi during
ing rate in the STN and GPi (Filion and Tremblay, LID (Filion et al., 1991; Merello et al., 1999a;
1991; Bergman et al., 1994; Merello et al., 1999a; Lozano et al., 2000; Boraud et al., 2001). Accord-
Vitek and Giroux, 2000). LID are associated with ingly, the averaged firing frequency is not the only
reduction in the STN and GPi firing rate in the MPTP physiological determinant of the dyskinetic state with
monkey model (Filion et al., 1991; Papa et al., 1999) respect to the parkinsonian motor condition. Thus, it
and in patients with PD (Merello et al., 1999a; Lozano is now believed that it is the pattern of neuronal activ-
et al., 2000; Boraud et al., 2001; Levy et al., 2001). ity in the output of the BG that is relevant to the
Levodopa treatment is associated with an increment development of dyskinesias (Vitek et al., 1999; Vitek
in the firing rate of GPe neurons (Filion et al., 1991; and Giroux, 2000; Obeso et al., 2000b; Bezard et al.,
Boraud et al., 1998). However, precise data regarding 2001; Boraud et al., 2001).

120
100
90
80
70
60
Hertz

50
80 40
30
20
Hertz

10
0
Off On On with
Dyskinesias

40

0
0 2 23 24 30 34
Off On On with Dyskinesias Minutes
Fig. 40.4. Recording of neuronal extracellular activity in the globus pallidus internal segment of one representative 1-methyl-
4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) monkey. In the off state there is increased firing rate, which is reduced when the
animal turns on after administration of a dopaminergic drug. The rate decreases to near zero, coinciding with the ensuing of
dyskinesias. Reproduced from Papa et al. (1999) with permission from the American Neurological Association.
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 197

Oscillatory Activity in the STN

5-10 Hz
2
1 qa
0

2,5
10-20 Hz

1,5
0,5

0,25
20-30 Hz

b 1,2
0,15
0,05

0,05
30-40 Hz

0,03
0,01 g1

0,07
>60 Hz

0,05
0,03
g2
0,01
(mV2) L-DOPA

OFF DD ON-Dyskinesias

Fig. 40.5. Recording oscillatory activity in the subthalamic nucleus of a patient with Parkinsons disease through implanted
electrodes. In the off state there is a predominant peak of activity in the beta band. The onset of diphasic dyskinesias (left vertical
line) is associated with drastic attenuation of beta activity and onset of theta activity (upper row). At the time of on dyskinesias
(right vertical line), there is an additional peak in the gamma band.

The importance of oscillations and patterns of neuro- and found precisely the same correlation between dyski-
nal activity in LID is starting to be clarified (Fig. 40.5). nesias and slow oscillations. The peak activity in the
In patients submitted to surgery for deep brain stimula- theta/alpha band coincided with a reduction in neuronal
tion (DBS) it is possible to record local field potentials firing rate in the SNpr and increased dopamine levels in
(LFP) from the implanted electrodes in the STN or GPi. the striatum. It appears, therefore, that reduction in
Typically, in the off state there is a predominant peak single-cell neuronal activity allows synchronization at
in the 1130 Hz (beta) band and, when patients are trea- around 48 Hz, which facilitates the onset of dyskinesias.
ted with dopaminergic drugs, the beta rhythm is drasti- Interestingly, a similar predominance around 48 Hz
cally attenuated and activity in the 6080 Hz gamma coinciding with LID has been recorded in the GPi in 1
band predominates (Levy et al., 2002; Brown, 2003). A patient with PD (Foffani et al., 2005) and in a group of
recent study has shown that the dyskinetic state is asso- patients with torsion dystonia (Silberstein et al., 2003).
ciated with a predominant oscillatory activity at 410 How this pattern of activity in the output nuclei of the
Hz in the STN of PD patients who developed LID BG releases involuntary movements is not understood.
(Alonso-Frech et al., 2006) and that such theta/alpha
peaks are absent in patients without dyskinesias. Indeed, 40.4.4. Anatomofunctional basis
stimulating the STN at this 48 Hz frequency in patients
treated with DBS in the STN elicited dyskinesias (Liu Loss of nigrostriatal terminals is associated with large
et al., 2002). In the 6-OHDA rat with LID the Bordeaux and uncontrolled oscillations in dopamine levels after
group (Meissner et al., 2006) recorded from the SNpr exogenous administration of levodopa (Miller and
198 J. A. OBESO ET AL.
Abercrombie, 1999; Brotchie et al., 2005), which previously installed LID in MPTP monkeys who actu-
increases with disease progression (De la Fuente- ally deteriorated further (Blanchet et al., 1994). Thus,
Fernandez et al., 2004; Obeso et al., 2004). The stria- the prediction of the model whereby levodopa induces
tum has been classically assumed to be the site where hyperactivity of the GPe leading to the expected over-
abnormal dopaminergic activation takes place in inhibition of the STN is not supported by metabolic
patients chronically treated with levodopa, but direct data. In this regard, it is worth noticing that there is a
evidence had not existed until recently. This has come direct dopaminergic innervation of the STN, GPe and
from experience regarding fetal mesencephalic trans- GPi (Hedreen, 1999) which is impaired in animal models
plants in patients with PD. Two double-blind studies and patients with PD (Francois et al., 2000; Jan et al.,
have conclusively shown that increasing dopaminergic 2000). It is therefore possible that modulation of STN
levels in the putamen (revealed by PET) is associated output activity occurs not only from the GPe but also
with dyskinesias, which are otherwise identical to the by a direct effect of dopaminergic drugs.
ones provoked by levodopa (Ma et al., 2002; Olanow The involvement of the direct pathway in the devel-
et al., 2003). opment of LID has been substantiated in recent years,
In line with the prevailing pathophysiological particularly by studies related with modifications of
explanation for LID discussed in the previous section, D1 striatal receptors signal transduction, as discussed
excessively large amounts of dopamine induce plastic in section 40.3 above. Animal models gave variable
changes in medium spiny striatal (i.e. putaminal) neu- results regarding the expression of D1-receptors in
rons, giving rise to the indirect and direct pathways. the striatum but importantly, no modification was pre-
The relative importance of either striatopallidal projec- sent in PD patients (Lee et al., 1978). A recent study
tion is not well defined and, in fact, it has changed concluded that the expression of D1 striatal receptors
over the years. is unchanged in relation to LID in MPTP monkeys
Direct data for the participation of the indirect but recognized an increment in binding of [(35)S]
pathway in LID are relatively scarce. In normal pri- GTP gamma, indicating functionally active receptors.
mates dyskinesias are evoked by reducing STN activ- Moreover, levels of Cdk5 and DARPP-32, two pivotal
ity (see above) and 2-deoxyglucose uptake is players in the D1 signal transduction pathway, are
increased in the STN and GPi of MPTP monkeys with increased in the striatum of dyskinetic monkeys
LID (Mitchell et al., 1992). Neuronal activity in the (Aubert et al., 2005). These findings were not present
STN from PD patients with dyskinesias elicited by in monkeys with a similar degree of parkinsonism
apomorphine during surgery is reduced compared with and dopaminergic depletion and without dyskinesias,
the off parkinsonian state (Lozano et al., 2000; Vitek despite the fact that they received the same regimen
and Giroux, 2000). Interestingly, the distribution of the of levodopa treatment. The severity of LID also corre-
410 Hz oscillatory activity recently associated with lated linearly with D3-receptor binding levels. Thus,
LID in PD patients (Alonso-Frech et al., 2006) and two markers of the medium spiny neurons at the origin
the increment in glucose uptake in dyskinetic MPTP of the direct pathway, such as D1- and D3-receptors,
monkeys predominate over the ventral region of the indicate a prominent role of the direct pathway in LID.
STN (Mitchell et al., 1992; Guigoni et al., 2005b). In contrast, levodopa treatment reduced and normalized
These results are in keeping with the classic model the increased expression of D2-receptors in MPTP mon-
and the role of the indirect pathway in the pathophy- keys with and without dyskinesias (Herrero et al.,
siology of LID. However, there is controversy over 1996a; Bezard et al., 2001). PET studies in PD patients
the functional state of the GPe and its role in the showed a similar pattern of striatal D2-receptor after
reduction of STN activity that mediates dyskinesias receiving levodopa treatment (Brooks et al., 1992).
(Obeso et al., 1997). On the one hand, recording neu- We believe that the dichotomy between the direct and
ronal activity in the GPe of dyskinetic animals has indirect pathways in the pathophysiology of LID is fruit-
shown increased firing rate (Filion et al., 1991; Boraud less and unrealistic. Manipulation of either pathway at
et al., 2001). However, metabolic markers are not in the level of the GPi can induce dyskinesias. Thus, in nor-
keeping with such findings. Expression of mRNA for mal monkeys dyskinesias can be elicited either by injec-
striatal met-enkephalin remains increased (Herrero tion of glutamatergic antagonists into the GPi, blocking
et al., 1995) and expression of glutamic acid decarbox- the STN glutamatergic excitation (Robertson et al.,
ylase (GAD) mRNA (an index of GABA activity) and 1989), or by local injection of the GABAergic agonist
cytochrome oxidase mRNA (an index of cellular muscimol (Burbaud et al., 1998), mimicking inhibition
activation) were not above normal in the GPe of dyski- from the GPe and the direct pathway. There are several
netic monkeys (Vila et al., 1997; Herrero et al., other sources of evidence indicating participation of both
1996b). Importantly, lesions of the GPe did not block striatopallidal projections in the 6-OHDA rat and MPTP
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 199
monkeys. For instance, to restore striatal LTP in the isms involved in the suppression of unwanted actions
denervated striatum activation of both D1- and D2-recep- and the running of automatic movements (Marsden
tors is necessary (Calabresi et al., 2000). Dyskinesias and Obeso, 1994; Obeso et al., 2000a). Normally,
may be elicited by a D2-agonist (i.e. PHNO, 4-pro- actions are limited to a few movements at a time.
pyl-3,4,4a,5,6,10b-hexahydro-2H-naphtho(1,2-b)(1,4) The motor apparatus guarantees that other body seg-
oxazin-9-ol) after co-administration of a D1-antago- ments are not recruited. The BG are thought to contri-
nist (i.e. SCH-233390) and vice versa, a D1-agonist bute fundamentally to this operation by setting the
may evoke dyskinesias in the presence of a D2- right balance between movement facilitation and inhi-
antagonist (Luquin et al., 1992a). Finally, and more bition (Mink and Thach, 1991; Marsden and Obeso,
importantly, administration of a D1-agonist in the 1994; Obeso et al., 2000a; Mink, 2003). Dyskinesias
primed MPTP monkey induces dyskinesias and represent a deviation of this essential mechanism
causes changes in neuronal activity in the GPi which whereby voluntary motor acts are performed with rela-
are the same as those caused by a D2-agonist that also tive accuracy, but unwanted, involuntary fragments of
provokes dyskinesias (Boraud et al., 2001). This sug- movements are also simultaneously recruited. It is as
gests that reduction of GPi firing frequency and slow if the motor system has lost the capacity to focus and
oscillatory synchronization responsible for LID may appropriately select just the correct and desired move-
occur through functional changes in several circuits ment pattern, a typical function of the BG (Denny-
influencing GPi output activity. In fact, recent anato- Brown and Yanagisawa, 1976; Marsden, 1982).
mical data strongly indicate that the concept of the
direct or indirect pathways cannot be sustained based 40.5. Treatment
on connectivity among the different BG nuclei (Lev-
esque and Parent, 2005). Our own view is, therefore, LID are highly prevalent in the overall PD population
that the available data do not suggest the exclusive and may be very disabling, interfering with quality of
participation of a given striatopallidal pathway in life (Chapuis et al., 2005). This is especially relevant
the pathophysiology of LID. More likely, changes in in patients with younger age at disease onset. For
afferent activity to the GPi from the GPe, STN and example, in PD beginning before the age of 45 years
putamen are all involved in setting the firing patterns old, LID are severe and present in almost all patients
and oscillatory activity that give rise to LID. after 5 years of treatment.
Lesions of the GPi and the pallidal receiving area of The optimal therapeutic approach for LID is to avoid
the thalamus may eradicate LID. This conclusively their development. Advances in such an approach have
indicates that the abnormal signals are conveyed been gained over the last few years but the outcome is
through the pallidothalamic projection to the cortical still quite limited. This is due to the fact that LID are
motor areas. Indeed, regional cerebral blood flow mea- directly related to disease progression, a problem that
sured by PET or single-photon emission computed has no realistic therapy yet and the inevitable need to
tomography (SPECT) was increased in the motor use levodopa during the evolution of PD.
thalamus in patients with LID (Hershey et al., 1998)
or hemichorea (Kim et al., 2002), indicating increased 40.5.1. Pharmacological treatments
input, presumably from the GPi. Two other studies
compared regional activation during the performance Three main therapeutic strategies are used to manage LID
of manual activity. A PET study with O15H2O com- in parkinsonian patients: (1) prevention of the sensitiza-
pared levodopa-treated PD patients with and without tion or priming phenomenon by early use of a dopamine
dyskinesia and found increased activation of the BG, agonist; (2) symptomatic treatment, once LID have
area 4, area 6, supplementary motor area and dorsolat- developed, with putative antidyskinetic interventions;
eral prefrontal cortex at rest and during a joystick task and (3) continuous dopaminergic stimulation to reverse
coinciding with on dyskinesias (Brooks et al., 2000). the functional changes underlying dyskinesias.
Using 133Xe SPECT to measure blood flow during a
finger movement task, Rascol et al. (1998) described
similar findings at the cortical level. 40.5.1.1. Prevention of priming
In conclusion, it appears that LID depend upon The use of neuroprotective drugs to slow disease pro-
uncontrolled dopaminergic signaling at the putaminal gression has been extensively explored. L-deprenyl
level, leading to abnormal generation of firing patterns (in an extension of the Deprenyl and Tocopherol Anti-
and oscillations in the output of the BG, which is con- oxidative Therapy of Parkinsons disease (DATATOP)
veyed via the thalamus to the cortical motor areas. LID study) failed to produce a significant reduction in the
may represent the dysfunction of primary BG mechan- incidence of dyskinesias (Shoulson, 1998).
200 J. A. OBESO ET AL.
The only group of drugs that has convincingly levodopa is added to the regimen. In addition, several
demonstrated a reduction in the risk of developing dys- issues related to the design of the studies have been
kinesias is the dopamine agonists. Three placebo-con- raised by critical voices. Our own view is that the
trolled studies comparing the evolution of patients severity of LID observed in clinical practice has been
initiated with a dopamine agonist (ropinirol, pramipexol considerably reduced in the last decade or so, coincid-
and cabergoline) and standard levodopa have shown ing with the earlier use of dopamine agonist and the
that the former provided significant symptomatic relief, associated possibility of reducing levodopa daily dose.
although slightly less than levodopa, with less risk In addition, neuroimaging studies with PET and
for developing dyskinesia after a follow-up period of SPECT suggested that dopamine agonists may be
25 years. associated with a reduction in the decline of dopami-
Rascol et al. (2000), in a comprehensive, double- nergic striatal terminals when used in the early stages
blind parallel study, compared the efficacy of ropinirol of the disease, raising the possibility of a neuroprotec-
and levodopa over a period of 5 years in 268 patients tive effect (Marek et al., 2002; Whone et al., 2003).
with early PD. The analysis of the time to onset of Thus, while sufficient and definitive data are being
dyskinesia showed a significant difference in favor compiled, we favor the prevailing concept of starting
of ropinirol. At 5 years, the cumulative incidence of therapy with a dopamine agonist, particularly in
dyskinesia, regardless of levodopa supplementation, patients who are 65 years old or younger, at the time
was 20% in the ropinirol group and 45% in the levo- of diagnosis.
dopa group. This confirmed clinically experimental
data in the MPTP monkey (Maratos et al., 2001), 40.5.1.2. Dyskinesias in patients already primed:
showing that ropinirol alone or in combination with
symptomatic treatments
low-dose levodopa delayed dyskinesia onset while
improving motor performance. The mean daily dose This is the commonest clinical scenario. Patients have
of ropinirol was 15 mg but the majority of patients already developed LID and the clinician has to attempt
enrolled in that arm required supplementary treatment to control the dyskinesias by adjusting the antiparkin-
with levodopa (Rascol et al., 2000). sonian drugs or adding agents capable of reducing
Holloway et al. (2004) compared the incidence of LID without increasing motor disability. The difficulty
motor complications between pramipexole and levodopa in achieving therapeutic efficacy is directly related to
as initial treatment in early PD. Patients allocated to pra- the severity and complexity of PD in each individual
mipexole treatment showed a significant reduction in subject. Thus, LID are relatively easy to control when
the risk of developing dyskinesias (24.5% versus 54%; they are mild and occur in patients with a wide thera-
P < 0.001). Cabergoline is an ergoline derivative with a peutic window but may be very difficult (or impossi-
very long half-life ( 72 hours) that can therefore be ble) to treat pharmacologically in severe patients who
administered once daily. In a double-blind multicenter exhibit all forms of LID and fall into severe off
trial on 419 patients naive to treatment, comparing caber- episodes when they are not dyskinetic. We shall
goline and levodopa as initial therapy for PD, motor review here the different individual pharmacologi-
complications were significantly delayed and occurred cal approaches available to treat LID but, in many
less frequently in cabergoline-treated patients compared instances of clinical practice, one needs to combine
to levodopa-treated patients (Bracco et al., 2004). several options.
An evidence-based review (Inzelberg et al., 2003)
compared the results of studies published on early 40.5.1.2.1. Dopamine agonists
treatment of PD with dopamine agonists (cabergoline, A dopamine agonist is added with the intention of
ropinirol or pramipexole) with similar studies using reducing levodopa dose and avoiding peak of dose
levodopa. Cabergoline, pramipexole and ropinirol on-dyskinesias associated with high levodopa plasma
were similarly effective in reducing the risk for dyski- levels. Belanger et al. (2003) examined the possibility
nesia relative to levodopa. Dyskinesia risk reduction of reducing LID by using a small dose of cabergoline.
was slightly greater for pramipexole and ropinirol than During treatment, they observed LID in the levodopa
for cabergoline. A concern encountered in the three stu- group but not in the levodopa cabergoline group,
dies was that, whereas treatment with a dopamine ago- which suggests that a small dose of a long-acting D2-
nist reduced the risk of dyskinesia, this was associated agonist combined with low doses of levodopa could
with less antiparkinsonian benefit. reduce the incidence of LID in patients with PD. This
It remains open to future analysis if the initial ben- study supports a commonly applied clinical strategy.
efit on LID of treatment with a dopamine agonist is Another approach is to achieve an antidyskinetic
carried forward over the long-term evolution, once effect with a partial dopamine agonist. These drugs,
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 201
characterized by having lower intrinsic activity at the Katzenschlager et al. (2005) assessed the effect of
receptor level than full agonists, act as either func- quetiapine on dyskinesias in a double-blind placebo-
tional agonist or antagonist, depending on the levels controlled cross-over study in 9 patients with PD,
of endogenous dopamine. A partial D2-receptor ago- receiving 25 mg quetiapine or placebo at night, for 2
nist may represent an interesting alternative for the weeks, in prerandomized fashion, with a 1-week wash-
treatment of PD and dyskinesias. out between treatment periods. Patients subsequently
Preclamol, the ()enantiomer of 3(3-hydroxyphenyl)- went on open-label quetiapine at 50 mg/day for a mean
N-n-propyl piperidine (3-PPP), has a selective dopamine duration of 30 days. During the double-blind phase,
mixed agonistantagonist profile for both pre and post- dyskinesias remained unmodified with either 25 mg
synaptic receptors. Its action in patients with disabling quetiapine or with placebo. On 50 mg/day quetiapine,
onoff fluctuations was compared against placebo a slight reduction in dyskinesia severity was observed
and subcutaneous apomorphine (Pirtosek et al., 1993). on a visual analog scale. This improvement was not
Preclamol had a mild but unequivocal antiakinetic effect, reflected in the patients overall impression of treat-
less than that achieved with subcutaneous apomorphine, ment effect. Thus, no antidyskinetic effect was per-
but caused less dyskinesia. Aripiprazole is a new ceived with the low dose (25 mg) of quetiapine, but
antipsychotic drug showing partial agonist activity for a possible antidyskinetic effect was described with
D2 and 5-HT1A-receptors and antagonist activity for higher doses.
5-HT2A-receptors. Lieberman (2004) postulated that Durif et al. (2004) investigated the efficacy and
this drug may be able to reduce dyskinesias without safety of clozapine in the treatment of LID in 50
producing akinesia, but further studies are required to patients during a 10-week, double-blind, parallel-
investigate its antidyskinetic capacity. group, placebo-controlled, multicenter trial. Mean clo-
zapine dose was 39.4  4.5 mg/day. The maximal LID
score at rest during the levodopa challenge was signif-
40.5.1.2.2. Dopamine antagonists
icantly decreased in the clozapine group. The authors
The use of drugs that block the dopaminergic system has concluded that clozapine is effective in the treatment
been a classical approach for the treatment of dyskinesias of LID in severe PD. In another study, Manson et al.
in general. D2-antagonists, like haloperidol, tiapride and (2000) assessed the usefulness of 2.57.5 mg/day of
sulpiride, and presynaptic dopamine-depleting drugs, olanzapine versus placebo for LID in 10 patients with
like reserpine and tetrabenazine, have all proven useful PD during a 2-week course. There was a 41% dyskine-
in the management of hemichorea-ballism (HCB), tard- sia reduction in the olanzapine-treated group compared
ive dyskinesias and tics. These same drugs are also effec- to placebo as measured by objective dyskinesia rating,
tive in reducing or stopping LID in PD, but this effect but it resulted in a significant increment in off time
is invariably associated with marked motor worsening as measured by patient diaries (30% versus 2%) and
after a variable period (ranging from hours to weeks). increased parkinsonism. These authors therefore con-
In clinical practice, therefore, they are not useful. cluded that olanzapine is effective in reducing dys-
Recent observations increasingly suggest that atypi- kinesias in PD but, even at a very low dose, it
cal neuroleptic drugs, which are able to block D3- may lead to unacceptable increases in parkinsonism
receptors preferentially, can be beneficial for patients and off time.
with movement disorders. Oh et al. (2002) evaluated
the effects of quetiapine, an atypical antipsychotic
with 5-HT2A/C and D2/3-antagonistic activity, on motor 40.5.1.2.3. Drugs acting on the opioid system
behavior in the 6-OHDA-lesioned rat and in MPTP The opioid striatal neurons may play a role in the
monkeys. In unilaterally lesioned rats, quetiapine induction of dyskinesias. Samadi et al. (2004) investi-
(5 mg/kg p.o.) reversed the shortening of the motor gated the effect of different doses of the opioid recep-
response to levodopa challenge produced by treatment tor antagonists naloxone and naltrexone on the
during 3 weeks with levodopa twice daily. Quetiapine dyskinetic response to the D1-agonist SKF-82958, the
(5 mg/kg p.o.) also normalized the short-duration D2-agonist quinpirole and levodopa in MPTP cyno-
response to acute injection of either a D1-receptor molgus monkeys. They found that conjunct adminis-
agonist (SKF-38392) or a D2-agonist (quinpirole) in tration of naloxone or naltrexone together with
rats that had received chronic levodopa treatment. dopaminergic agents led to a significant reduction in
Quetiapine had no effect on parkinsonian manifesta- the severity of dyskinesias without losing antiparkinso-
tions when given alone to 6-OHDA-lesioned rats or nian efficacy. Carroll et al. (2004) conducted a rando-
MPTP monkeys but did substantially reduce LID when mized, double-blind, placebo-controlled cross-over
administered together with levodopa. trial to examine the hypothesis that cannabis may have
202 J. A. OBESO ET AL.
a beneficial effect on dyskinesias in PD. Seventeen LID and is therefore worth trying in the absence of con-
patients completed the trial and cannabis was well toler- traindications. The antidyskinetic effect is probably
ated with no pro- or antiparkinsonian action, but no evi- exerted at the level of the STN as amantandine failed
dence of a treatment effect on LID as assessed by to control dyskinesias evoked by subthalamotomy in
Unified Parkinsons Disease Rating Scale (UPDRS) or patients who had previously responded markedly well
any secondary outcome measurements. Thus, despite (Merello et al., 2006).
many experimental suggestions, there is no drug Merello et al. (1999b) evaluated the efficacy of
currently employed clinically to manipulate the opioid memantine (1-amino 3,5-dimethyl-adamantane hydro-
system for the treatment of LID (Manson et al., 2001). chloride) on cardinal symptoms of PD, the pharmaco-
logical response to levodopa (latency, duration and
40.5.1.2.4. Glutamatergic antagonists magnitude) and the induction of dyskinesias. In 12
patients and contrary to recent findings with amantadine,
The use of NMDA antagonists in humans has been no effect on LID was observed. However, in a recent
generally limited because of adverse effects associated
report by Lokk (2004), memantine was given to 3 PD
with non-selective NMDA receptor blockade.
patients with cognitive impairment and LID and 2 of
Verhagen Metman et al. (1998b) in a double-blind
them benefited with regard to dyskinesia control.
cross-over study showed that 3 weeks treatment with
Riluzole is an inhibitor of glutamatergic transmis-
dextrometorphan was able to reduce dyskinesias by
sion in the central nervous system currently given to
3040% while maintaining the response to levodopa.
patients with lateral amyotrophic sclerosis in an
In recent years, amantadine has become popular as a
attempt to improve prognosis. Braz et al. (2004) eval-
possible antidyskinetic drug based on its putative uated the effect of riluzole on dyskinesia in 16 patients
anti-NMDA action (Verhagen Metman et al., 1998a).
with PD and found no effect against apomorphine-
However, it is worth noticing that there is no definitive
induced dyskinesias.
evidence indicating such a mechanism of action. Del
In general, the high expectations (Chase et al.,
Dotto et al. (2001) evaluated the effect of a 2-hour intra-
2000) that were raised with the potential therapeutic
venous amantadine (200 mg) or placebo infusion against
impact of antiglutamatergic drugs for PD have failed
LID in 9 PD patients with motor fluctuations and
to become a reality.
severely disabling peak-dose dyskinesias. Intravenous
amantadine acutely improved LID by 50%, without los- 40.5.1.2.5. Drugs acting on the serotoninergic system
ing the antiparkinsonian benefit of levodopa in a 5-week,
double-blind cross-over trial. In another study Luginger The serotoninergic system projects quite profusely to
et al. (2000) assessed dyskinetic severity following oral the striatum and also to other key BG nuclei (i.e.
levodopa challenges as well as with self-scoring dyskine- STN, GPe, GPi) and exerts an inhibitory control on
sia diaries and found them to be reduced by approxi- dopamine striatal transmission. Durif et al. (1995)
mately 50% after amantadine treatment compared with found a 47% improvement in the severity of dyskine-
baseline or placebo control. Snow et al. (2000) evaluated sias evoked by apomorphine in 7 patients with PD
the effect of amantadine on LID in a double-blind, pla- treated with fluoxetine. There was no reduction in
cebo-controlled study and found a 24% reduction in the levodopa antiparkinsonian benefit.
total dyskinesia score. Buspirone has a complex mechanism of action,
The above clinical data indicate that amantadine is which aside from its 5-HT1A properties includes par-
an effective treatment for LID. However, the duration tial dopamine agonism and mild opiate and noradre-
and magnitude of its antidyskinetic effect remain to be nergic antagonism (Kleedorfer et al., 1991). Bonifati
established. Thomas et al. (2004) performed a 12-month et al. (1994), in a double-blind, placebo-controlled,
double-blind study including 40 patients suffering peak- cross-over study, found that buspirone (20 mg) signif-
dose and diphasic dyskinesias in an attempt to elucidate icantly lessened the severity of LID in 5 out of 7
the duration of the antidyskinetic effect. After 15 days patients. Mirtazapine is an a2-antagonist, 5-HT1A ago-
of amantadine treatment, a reduction of 45% in the total nist and 5-HT2 antagonist, potentially useful for LID.
dyskinesia scores was detected, but the benefit lasted Meco et al. (2003) found in an open-label study
less than 8 months. Moreover, in a systematic review including 20 parkinsonian patients that mirtazapine
on the efficacy and safety of amantadine for LID treat- may be effective in reducing LID.
ment, Crosby et al. (2003) concluded that there was
not enough evidence to determine whether amantadine 40.5.1.2.6. Noradrenergic drugs
was effective. Our view is that, on an individual basis, The close relationship between the dopaminergic,
amantandine may result in a drastic amelioration of adrenergic and noradrenergic systems has led to the
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 203
assessment of a possible antidyskinetic effect of a few infusion of a levodopa/carbidopa gel (Nyholm et al.,
drugs acting on those systems. Two studies have 2005). The time spent in the on state was increased
shown how the a2-adrenoreceptor antagonist idazoxan 81100% by infusion therapy with no increase in
can significantly reduce LID in MPTP monkeys as dyskinesias. This was associated with improvement in
well as in patients with advanced PD (Grondin et al., quality-of-life scores. This approach is now commer-
2000). Rascol et al. (2001) reported improvement of cially available and may be a valid option for some
LID without reappearance of parkinsonian symptoms patients, despite the obvious practical limitations.
in 18 patients treated with idazoxan. Carpentier et al. Similarly, continuous delivery of dopamine ago-
(1996) found a significant 40% improvement in dyski- nists by the subcutaneous route, such as lisuride and
nesia scores in PD patients treated with a low dose of apomorphine, is associated with a reduction in LID.
propranolol (1020 mg/day). The majority of trials used infusions during the day-
time but stopped at nighttime to reduce the risk of
40.5.1.2.7. Practical considerations severe psychiatric complications.
There appear to be many drugs that are capable of Manson et al. (2002) reviewed their experience in
reducing the severity of LID. In some occasional 64 patients treated with apomorphine infusions.
patients, the therapeutic impact of any one of the treat- Forty-five patients were successfully converted to
ments summarized above may be strikingly positive, monotherapy and discontinued all other dopaminergic
but in the majority it is limited to mild or short-lasting drugs during the daytime treatment period with apo-
improvement. Nevertheless, they are generally well morphine. Patients were followed for a mean of 33.8
tolerated and worth trying in patients in whom other months, with a mean maintenance dose of apomor-
therapeutic measurements cannot be afforded. In our phine of 98 mg/day. Dyskinesias were reduced by
experience, the degree of symptomatic control of 64% in the monotherapy group compared to 30%
LID mainly depends upon the complexity of dyskine- in those on polytherapy. These results confirmed that
sias and severity of off periods. This may be schema- subcutaneous apomorphine monotherapy can reset
tically summarized as follows: (1) in patients with peak-dose dyskinesia threshold in levodopa-treated
mild but bothersome peak-dose dyskinesias, read- patients, while further reducing off-period disability.
just the levodopa schedule, and consider adding a Katzenschlager et al. (2005) prospectively assessed
dopamine agonist and any one of the drugs discussed the antidyskinetic effect of continuous subcutaneous
above; (2) for patients with severe peak-dose dyskine- apomorphine using subjective and objective measures
sias, consider switching treatment to provide continu- and the response to a levodopa challenge. At 6 months,
ous dopaminergic stimulation; and (3) patients with the mean levodopa dose had been reduced by 55% and
severe peak-dose dyskinesias and diphasic dyskinesias the daily off time in patients diaries was reduced
probably require surgical treatment. by 38%. Levodopa challenge showed a reduction of
4044% in the dyskinesia scores. Patients self-assess-
40.5.1.3. Reversing priming: continuous ment scores reflected these significant changes. The
dopaminergic stimulation improvement correlated with a reduction in oral
Since the introduction of the concept of continuous drug therapy and with final apomorphine dose. This
dopaminergic stimulation in the 1980s (Horowski prospective study confirmed a marked reduction in
et al., 1988; Chase et al., 1989; Obeso et al., 1989, dyskinesia with continuous subcutaneous apomorphine
1994), it was realized that constant delivery of dopami- therapy, paralleled by reduced dyskinesias during
nergic drugs was associated with a reduction in the dopaminergic challenge tests. Similarly, Stocchi et al.
severity of LID. Over the past decade, further evidence (2002) compared the long-term incidence of dyskine-
has accumulated to support the notion that continuous sias in patients treated with subcutaneous infusion of
stimulation of dopamine receptors may even reverse lisuride (plus supplementary oral levodopa as needed)
part of the changes induced by chronic pulsatile levo- versus patients treated with standard levodopa orally
dopa administration. The initial pivotal study in this and showed that patients receiving lisuride infusions
regard was conducted by Mouradian et al. (1990), experienced a significant reduction in the incidence of
who continuously delivered levodopa intravenously both motor fluctuations and dyskinesia, compared with
for 712 days to a small group (n 12) of patients with patients receiving standard dopaminergic therapies.
severe PD. They found a progressive attenuation of LID Benefits persisted for the 4-year duration of the study.
and amelioration of the onoff fluctuations. More This study also confirmed earlier results indicating that
recently, 24 patients with motor fluctuations and dyski- continuous lisuride infusion can be fairly well tolerated
nesia were randomized in a cross-over design to com- and beneficial for patients motor complications (Obeso
pare conventional oral treatments and intraduodenal et al., 1986), provided they have not previously
204 J. A. OBESO ET AL.
developed severe psychiatric complications (Vaa- Motor function and UPDRS motor score are not
monde et al., 1991). improved in the on medication condition following
Overall, these results support the concept that repla- pallidotomy (Bronstein et al., 1999; Alkhani and
cement of short-acting oral antiparkinsonian medication Lozano, 2001). Pallidotomy conveys a marked and
with drugs capable of providing a more continuous permanent benefit against LID. The lesion practically
dopamine receptor stimulation may at least partially abolishes peak-dose dyskinesias, diphasic dyskinesias
avoid or reverse the sensitization process believed to and off-period dystonia on the contralateral side to
mediate the development of LID. In theory, therefore, the lesion. This effect is permanent, in our experience
therapy with infusions capable of providing continuous lasting 10 years postoperatively, and has been shown
dopaminergic stimulation might be the pharmacological to remain stable between 1 and 4 years after surgery
treatment of choice for advanced PD patients. by controlled studies (Baron et al., 2000). A beneficial
Nevertheless, the degree of control of LID achieved effect in the ipsilateral side of the surgery has also
with infusions is not complete in many patients. Phar- been reported but became not significant after 12
macological tolerance appears in a large proportion years (Lang et al., 1997; Baron et al., 2000). The
after some time on treatment. It occurs more readily meta-analysis of Alkhani and Lozano (2001) found
the more severe the underlying disease is, leading to 71 patients properly assessed 1 year after pallidotomy.
off episodes or exacerbation of diphasic dyskinesias. In such a selected patient population, LID were
The latter may cause a very troublesome dyskinetic reduced by 73.5% and 86.4% at 6 months and 12
status (Vaamonde et al., 1991). Thus, surgery may still months respectively postsurgery. Bilateral pallidotomy
be the only and best therapeutic option for a proportion is, despite its potentially larger antiparkinsonian effect,
of patients with severe LID. a procedure with high risk of speech (Favre et al.,
2000), gait and cognitive deterioration (Intemann
40.5.2. Surgery et al., 2001; Merello et al., 2001). Thus, it is not parti-
cularly indicated nowadays when deep brain stimula-
Early surgical procedures to treat hyperkinesias, such tion (DBS) is available throughout the world.
as choreoathetosis or hemiballism, were aimed to DBS of the GPi (GPi DBS) has a significant anti-
interrupt the peripheral and spinal motor pathways, dyskinetic effect, about 7080% reduction, which is
the cerebral peduncles and cerebral cortex with subpial associated with improvement in activities of daily liv-
extirpations of motor areas. Subsequently, during the ing (ADL) score. The effect of GPi DBS against LID
early 1950s, the GPi became the target of choice for appears stable after several years in spite of maintain-
the surgical treatment of any movement disorder, ing levodopa daily dosage unchanged. A recent study
whether bradykinetic or hyperkinetic. showed a reduction in LID severity of 76% (P
The introduction of levodopa modified the need for < 0.0001) with no change in the levodopa dose equiva-
the surgical treatment of PD. For many years thalam- lents in 20 patients with severe PD followed by 34
otomy was the only surgical procedure performed in years in the Cooperative Study of DBS for advanced
patients with PD. Failure to control appropriately the PD (Rodriguez-Oroz et al., 2005). Volkmann et al.
tremor or levodopa-induced side-effects were the prin- (2004) followed 11 patients for 5 years after bilateral
cipal reasons for elective surgery during the 1970s and GPi DBS and reported that LID remained significantly
up to the early 1990s. In the last decade, there has been reduced (58% at 1 year, 63% at 3 and 64% at 5 years)
a revitalization of surgery for PD in the light of the in parallel with a worsening in the degree of benefit in
newer pathophysiological concepts (DeLong 1990; the UPDRS motor scale (56% at 1 year, 43% at 3 and
Obeso et al., 1997), described in section 40.4 above. 24% improvement at 5 years). This study therefore
showed a differentiated response, being therapeutically
40.5.2.1. Surgery of the globus pallidum very positive against LID but showing no sustained
Unilateral pallidotomy induces a significant alleviation benefit against the parkinsonian condition, similar to
of the cardinal motor features of PD in the side con- what occurs with pallidotomy (Volkmann et al., 2004).
tralateral to the surgery, reducing the severity of dis- Two studies described that GPi DBS may have both
ability in the off motor state and the UPDRS motor a prodyskinetic and an antidyskinetic effect depending
score. Alkhani and Lozano (2001) encountered 1735 upon electrode placement, suggesting two different
pallidotomies in 85 papers published between1992 and targets for parkinsonism and LID in the same anatomi-
1999 from 40 different centers. The UPDRS motor cal structure (Bejjani et al., 1997; Krack et al., 1998).
score improved by about 4045% at 6 and 12 months The dorsal contacts of the GPi conveyed the prodyski-
after pallidotomy, the benefit being mainly contralateral netic effect whereas the antidyskinetic action was due
to the lesion (Alkhani and Lozano, 2001). to activation of the ventral contacts. Yelnik et al.,
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 205
(2003) described that dyskinesias induced by stimula- PD by its striking capacity to reduce off medica-
tion in the GPi may be correlated with the placement tion severity and disability and because it is usually
of the dorsal contacts within or near the GPe. In mon- associated with a 3060% reduction of levodopa daily
keys, inactivation of the motor region of the GPe (by dose, unlike pallidal surgery. The experience regarding
local administration of bicuculline) is associated with the effect of STN DBS on LID is relatively homoge-
choreatic movements (Crossman et al., 1988; Grabli neous in different studies. Most surgical groups have
et al., 2004). It is possible therefore that activation of reported a significant antidyskinetic effect, closely
GABAergic fibers from the GPe or blockade of the correlated with reduction in daily levodopa dose
GPe itself from dorsal contacts is responsible for the (Table 40.1) (Krack et al., 2003; Moro et al., 1999;
reported induction of dyskinesias. Interestingly, we Molinuevo et al., 2000; Obeso et al., 2001; Simuni
never encountered this problem with GPi DBS (Obeso et al., 2002). Subthalamic stimulation appears to
et al., 2001) and it has not been reported after long- improve the whole spectrum of LID, namely peak
term follow-up of 20 patients from eight centers dose, diphasic dyskinesias and off-dystonia (Krack
(Rodriguez-Oroz et al., 2005). et al., 1999). Two independent studies summing
together 98 patients reported a significant reduction of
40.5.2.2. Surgery of the subthalamic nucleus LID severity after 45 years of follow-up (Krack
Nowadays, bilateral stimulation of the STN (STN et al., 2003; Rodriguez-Oroz et al., 2005) (Table 40.1).
DBS) is the surgical procedure most often used for Levodopa was reduced from a mean of 1309 to 859

Table 40.1
Efficacy of bilateral deep brain stimulation (DBS) of the globus pallidum pars interna (GPi) and the subthalamic
nucleus (STN) against levodopa-induced dyskinesias in Parkinsons disease: summary of the literature

Levodopa
Reference Patients Surgery Dyskinesia reduction reduction Follow-up

Ghika et al. (1998) 6 GPi DBS P < 0.01 Unchanged 6 months


Houeto et al. (2000) 23 STN DBS 77% (P < 0.001) 61% (P < 0.001) 6 months
DBSPDSG 36 GPi DBS P < 0.01 Unchanged 6 months
96 STN DBS P < 0.001 (P < 0.001)
16 (6
Loher et al. (2002) unilateral) GPi DBS 71% Unchanged 12 months
Patel et al. (2003) 16 STN DBS Duration (P < 0.002) 48% (P < 0.002) 12 months
Disability (P < 0.01)
Simuni et al. (2002) 12 STN DBS 64% 55% 12 months
Hamel et al. (2003) 25 STN DBS 50% 50% 12 months
Ford et al. (2004) 30 STN DBS P < 0.001 22% 12 months
Burchiel et al. (1999) 6 STN DBS 67% (P < 0.003) 51% (P < 0.03) 12 months
Bejjani et al. (2000) 12 STN DBS 83% (P < 0.05) 70% (P < 0.05) 12 months
Kleiner-Fisman et al.
(2003) 25 STN DBS 46.4% (P < 0.007) 38% (P < 0.001) 24 months
Herzog et al. (2003) 48 STN DBS 85% 67.8% 24 months
Rodriguez-Oroz et al.
(2005) 11 STN DBS 53% (P < 0.01) 50% (P < 0.01) 4 years
Rodriguez-Oroz et al.
(2005) 20 GPi DBS 76% (P < 0.0001) Unchanged 4 years
49 STN DBS 59% (P < 0001) (P < 0.001)
Visser-Vanderwalle 75% (P < 0.001)3 4 years and
et al. (2003) 26 unilateral GPi DBS months 53% increased 3 months
28% (P < 0.03)15
months
Volkman et al. (2004) 11 GPi DBS 64% (P < 0.05) Unchanged 5 years
Krack et al. (2003) 49 STN DBS Duration (P < 0.001) 74% (P < 0.001) 5 years
Disability (P < 0.001)
206 J. A. OBESO ET AL.
mg/day (P < 0.001) and in 6 patients levodopa was of PD on the side contralateral to the surgery and on
stopped altogether (Rodriguez-Oroz et al., 2005). axial features up to 7 years postoperatively (Alvarez
Hamani et al. (2005) in a recent survey encountered 38 et al., 2001, 2005). Immediately after surgery, the
studies from 34 neurosurgical centers with 737 patients great majority of patients exhibit dyskinesias on the
involved. They found reduction of LID (UPDRS dyskine- operated side but these are transient and self-resolving
sias scores) by 73% and 94% at 6 and 12 months in the in most instances, mimicking what happened after pal-
on-stimulation on medication compared with preopera- lidotomy (Merello et al., 1997). The incidence of
tive on medication scores. symptomatic, i.e. severe, HCB secondary to lesion of
For a large majority of authors the LID response to the STN is not as large as expected by classic neuro-
STN DBS is directly related to a reduction of dopami- surgical concepts. A review of the literature looking
nergic drugs (Molinuevo et al., 2000; Moro et al., for HCB in PD secondary to mistargeting during thala-
2002; Vingerhoets et al., 2002). Recently, a study motomy in the pre-levodopa era indicated a low inci-
separated patients into two groups after STN DBS dence of cases (Guridi and Obeso, 2001). In fact, we
according to the possibility of reducing levodopa. have observed symptomatic HCB in 7% of 170 conse-
The group that required medication after surgery had cutive patients with unilateral STN lesions operated in
a moderate alleviation in LID (47% reduction) and the CIREN. In such instances, pharmacological man-
the group that did not receive levodopa after surgery agement of the HCB is not simple. Stopping all anti-
had a dramatic reduction in LID (90%; P < 0.003) parkinsonian drugs does not immediately abolish
(Vingerhoets et al., 2002). On the other hand, some the dyskinesias and may result in worsening of the
authors have suggested that the antidyskinetic non-operated side (Su et al., 2002; Alvarez et al.,
response after STN DBS could be due to a direct effect 2005). Amantandine is not useful either, even in
of continuous high-frequency electrical stimulation in patients in whom this drug has been previously useful
the target (Krack et al., 1999). Another possible for LID (Merello et al., 2005). Pallidotomy is higly
mechanism could be that stimulation of fibers running efficacious in eliminating the HCB in those few
above the dorsal border of the STN (fasciculus lenticu- patients in whom the dyskinesia persists and is dis-
laris and zona incerta) would block impulse trans- abling. This corroborates the principle established in
mission in the GPi-thalamic projection. This would the monkey by Carpenter et al. (1950) and recently
be in agreement with the findings that the STN target applied in patients with HCB of vascular origin (Vitek
for stimulation is dorsal to the upper border of the et al., 1999). In our experience, pallidotomy abolishes
nucleus and therefore the electrical current may spread the HCB secondary to subthalamotomy without aggra-
to fibers in the fasciculus lenticularis passing through vation of parkinsonian features or indeed any other
the Forel field. In such instances, the mechanism neurological complication.
would be similar to a pallidotomy-like effect. Experience with bilateral, simultaneous subthala-
Lesion of the STN, i.e. subthalamotomy, as a sur- motomy is much more limited but it seems that HCB
gical approach for PD was first considered in the may be more frequent with such an approach, which
early 1990s (Guridi et al., 1993). This relied on the requires further evaluation before being recommended
striking improvement induced in the MPTP monkey for routine application (Alvarez et al., 2005).
model by unilateral ablation of the STN. However, The factors governing the onset and persistence of
the fear of inducing hemiballism moved attention severe HCB after subthalamotomy are not yet well
towards pallidotomy, despite the fact that there was defined. Extension of the lesion dorsally to interrupt
no experimental basis, because of the important surgi- the fasciculus lenticularis, producing a pallidotomy-
cal experience accumulated in patients by Laitinen like effect, has been suggested as an explanation of
et al. (1992). Currently, subthalamotomy is performed the low incidence of HCB after a lesion of the STN
by only a few teams, around the world. Probably, this is (Lozano, 2001; Guridi and Obeso, 2001; Chen et al.,
due to the greater methodological difficulty, in com- 2002). However, almost all instances of severe HCB
parison with DBS surgery, in establishing a lesion have been associated with dorsal lesions, usually large
large enough to have a permanent and marked benefit enough to extend well within the zona incerta, which
without side-effects. The largest published experience should have also lesioned the pallidothalamic projec-
corresponds to the Centro Internacional Restauracion tion. Guridi and Obeso (2001) suggested, based on their
Neurologica (CIREN) in La Habana (Cuba) as part of experience with lesions of the STN in MPTP monkeys
an international collaborative effort (Alvarez et al., and the organization of the BG, that the dyskinetic
2001, 2005). threshold is increased in the parkinsonian state, thus
Subthalamotomy is associated with a significant reducing the probability of developing severe HCB
and maintained improvement in all cardinal features after interrupting the STN efferent activity.
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 207
The striking antiparkinsonian effect of subthalamot- Benabid in Grenoble introduced DBS for PD for the
omy per se or the presence of HCB has resulted in a treatment of tremor instead of thalamotomy (Benabid
drastic reduction in levodopa daily dose in most et al., 1989, 1996). Generally, it is agreed that the
reported studies, making assessment of the impact on effect of Vim-DBS on LID is minor or nil. Limousin
LID almost impossible. However, Alvarez et al. et al. (1999) reported 73 patients enrolled in a multi-
(2001) kept the levodopa dose unchanged up to a year center cooperative study for the treatment of tremor
after unilateral subthalamotomy and found a signifi- and described that LID were slightly, but not signifi-
cant reduction in LID. Those patients nevertheless cantly decreased by DBS at 12 months postopera-
were chosen for surgery precisely because of the tively. Similarly, Tasker et al. (1997) found no
absence of severe LID preoperatively. Su et al. significant difference between Vim-thalamotomy and
(2002) also reported that LID were reduced by 75% Vim-DBS regarding LID. On the other hand, the Lille
at 18 months postoperatively in 3 out of 4 patients group described in 12 parkinsonian patients LID aboli-
who suffered severe bilateral LID before surgery, even tion after Vim-DBS. They suggested that electrodes
though the levodopa dose was unchanged during the placed near the centromedian-parafascicular nucleus
initial 3 months. Recently, Alvarez et al. (2005) of the thalamus may have a more marked effect
reported the effects of bilateral subthalamotomy in against LID, but this has not been further substantiated
18 patients, 7 staged lesions and 11 simultaneous (Caparros-Lefebre et al., 1999).
surgery, followed for a minimum period of 3 years
and up to 7 years. In both groups, the reduction in
40.5.2.4. Mechanism of action of surgery
LID was significant after subthalamotomy together against LID
with a reduction in daily levodopa dose.
Hassler in Freiburg and a few years later Cooper in
New York considered that hyperkinesias were asso-
40.5.2.3. Thalamic surgery ciated with uncontrolled discharges in the GPi, leading
Hassler and Riechert in 1954 pioneered thalamotomy to to excessive impulses in the pallidal projection to the
improve tremor control in PD. The optimal region within Voa of the thalamus (Hassler et al., 1960; Cooper,
the thalamus was later defined by Ohye et al. (1976) as 1969). Hassler reported that hyperkinesias of various
the ventralis intermedius (Vim) and became the estab- etiologies and in particular HCB following surgery
lished surgical target for tremor up to the 1970s, when for PD could be abolished by a lesion placed in the
levodopa was introduced as the treatment of choice for GPi, by coagulation of the Forel H2 field or by an
PD. Subsequently, a few papers described a possible pre- extensive lesion performed in the Voa/Vop of the tha-
ventive effect on the development of LID in patients lamus. This approach was supported by earlier work in
with a prior thalamotomy (Narabayashi et al., 1984; the monkey performed by Mettlers group (Whittier
Derome et al., 1986; Tasker, 1990) or the successful and Mettler, 1949; Carpenter et al., 1950). They
amelioration of LID after surgery (Kelly and Gilling- described the induction of HCB by destruction of the
ham, 1980; Fox et al., 1991; Jankovic et al., 1995). How- STN (at least 20% of its volume) and how a second
ever, there is no consistent report in the literature truly lesion placed in the globus pallidum or the lenticu-
assessing the impact of Vim-thalamotomy against LID, lar fasciculus abolished the choreoballic movements
with the exception of the work of Narabayashi et al. (Carpenter et al., 1950).
(1984). This report described that patients with surgical According to current pathophysiological concepts
lesions in Voa/Vop-thalamotomy (ventralis oralis ante- of PD, surgical destruction or inactivation of pallidal
rior and posterior, which are the pallidal receiving terri- neuronal activity reduces excessive inhibitory output
tory in the thalamus) prior to the introduction of to the thalamus (DeLong, 1990; Obeso et al., 1997).
levodopa did not develop LID. On the other hand, Pallidotomy has been shown by PET, functional mag-
patients with Vim-thalamotomy for tremor developed netic resonance imaging and physiological studies to
dyskinesias when levodopa was introduced for PD restore thalamocortical excitability functionally
(Narabayashi et al., 1984). They concluded that the (Bakay et al., 1992; Eidelberg et al., 1996; Samuel
GPi-Voa/Vop pathway mediated LID, in agreement with et al., 1997). On the other hand, reduction of GPi out-
Hasslers previous concept about the origin of hyperki- put activity is the essential physiological hallmark of
nesias (Hassler, 1978). Page et al. (1993) reported simi- LID (Filion and Tremblay, 1991; Papa et al., 1999;
lar results in MPTP monkeys with LID in whom a lesion Boraud et al., 2001), which is superficially in contra-
in the pallidal territory of the thalamus abolished diction with the profound antidyskinetic effect of palli-
dyskinesias but lesion of the nigral or cerebellar terminal dotomy. In other words, according to the classic
territories had no benefit. pathophysiological model of the BG, pallidotomy
208 J. A. OBESO ET AL.

should induce a worsening rather than amelioration of the BG through its dense glutamatergic innerva-
LID. Recent findings may shed some light on this see- tion of the GPi, GPe and SNpr and, to a lesser
mingly paradoxical finding. extent, of the substantia nigra pars compacta and
In the rat with unilateral lesion of the nigrostriatal striatum. It could be that a given subregion of
pathway by 6-OHDA, LID are associated with a the STN is required to maintain synchrony in the
reduction in neuronal firing rate in the SNpr, the main BG at 48 Hz. Consequently, blockade of the
BG output in rodents. Simultaneously, the same ani- STN removes an essential component of the net-
mals exhibited a peak in the theta/alpha band (410 work engaged in the generation of the abnormal
Hz) recorded from the SNpr by LFPs. Thus, reduced signals, leading to LID. In this regard, it is note-
firing rate of single neuronal activity and oscillatory worthy that LID in the MPTP monkey model is
activity in the theta band in the SNpr coincided with associated with a significant increase in 2-deoxy-
excessive striatal dopamine release (Meissner et al., glucose uptake in the ventromedial region of the
2006). Moreover, recording of LFP from the GPi and STN (Mitchell et al., 1992; Guigoni et al., 2005b).
STN of PD patients who were implanted with electro- 2. STN blockade functionally restores the BG,
des for DBS have shown that LID are associated with making it relatively insensible to the prodyskinetic
a specific oscillatory activity at around 48 Hz effects of levodopa. Reduction of STN effer-
(Brown, 2003; Foffani et al., 2005; Alonso-Frech ent activity, immediately after a lesion or at the
et al., 2006). These two complementary experimental beginning of DBS treatment, may be associated
and clinical studies strongly suggest that LID coin- with dyskinesias because of drastic reduction in
cides with an abnormally slow synchronization of neu- excitatory drive on to the GPi. However, this gives
ronal activity in the output of the BG, which is the way over the following weeks to a functional
code or signal conveyed to the cortex via the thalamus. restoration of output (Hirsch et al., 2000). Thus,
Indeed, neuroimaging studies using PET and SPECT molecular markers such as mRNA expression of
data showed increased activity and regional blood flow cytocrome oxidase and succinate dehydrogenase
in the supplementary motor area, premotor area and (as an index of cellular activation) or mRNA
ipsilateral and contralateral primary motor areas expression of GAD become reduced or even nor-
(Rascol et al., 1998). Accordingly, pallidotomy and malized in the output BG after subthalamotomoty
Voa/Vop-thalamotomy would remove the effect in the rat and monkey (Guridi et al., 1996; Blan-
of slow oscillatory activity on the thalamocortical dini et al., 1997). Moreover, subthalamotomy
motor projection, stopping the delivery of wrong reverses the increase in corticostriatal glutamater-
signals or noisy patterns to the cortex. Whether gic activity and increased GAD mRNA expression
GPi-DBS acts by a similar mechanism is unknown at in entopeduncular nucleus (GPi equivalent in the
present. It seems likely that GPi-DBS will interfere rat) associated with dopamine depletion, reverses
with the theta rhythm associated with LID, either by the abnormalities in striatal excitatory synaptic
blocking GPi neuronal activity or by driving it into a transmission (Delfs et al., 1995; Touchon et al.,
different (i.e. non-prodyskinetic) oscillatory activity. 2004; Centonze et al., 2005) and ameliorates the
The improvement of LID in patients treated with wearing-off response (Marin et al., 2004). It
STN surgery may have a more complex interpretation. appears therefore that STN surgery leads to func-
Reduction of levodopa daily dose is not the sole expla- tional normalization of the BG output nuclei. This
nation, as a proportion of patients treated with STN in turn could reduce the sensitivity of the BG net-
surgery clearly had an improvement in LID despite work to pulsatile dopaminergic stimulation with
maintaining levodopa dose unchanged. A pallidot- standard levodopa (Obeso et al., 2004), thus
omy-like effect, by dorsal extension of the lesion or increasing the threshold for dyskinesias. We envi-
the electrical field, to block pallidothalamic transmis- sage that STN blockade stabilizes the BG network,
sion does not seem to account for all observations. removing the generation of abnormal signals in
We may suggest here that blockade of STN activity the parkinsonian off state and in response to
may have by itself an anti-LID effect. This could take levodopa.
place through two principal mechanisms:
40.6. Conclusions
1. The abnormal rhythms and oscillation patterns
associated with LID represent neuronal firing syn- The origin of LID in PD is mainly related to the impact
chrony in various nuclei of the BG and possibly on striatal physiology of nigrostriatal dopaminergic
elsewhere. The STN is the major driving force of depletion and discontinuous levodopa replacement ther-
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 209
apy. These combine to induce abnormal expression of Augood SJ, Emson PC, Mitchell IJ et al. (1989). Cellular
dopamine-receptor signaling and neuropeptides, leading localisation of enkephalin gene expression in MPTP-
to deranged corticostriatal plasticity. Avoiding pulsatile treated cynomolgus monkeys. Brain Res Mol Brain Res
dopaminergic stimulation (i.e. high dose of standard 6: 8592.
levodopa administration) has already resulted in a reduc- Bakay RAE, DeLong MR, Vitek JL (1992). Posteroventral
pallidotomy for Parkinsons disease. Letter. J Neurosurg
tion in the incidence and severity of LID in the general
77: 487488.
PD population. Treatment of severe LID requires contin-
Bara-Jimenez W, Bibbiani F, Morris MJ et al. (2005). Effects
uous dopaminergic stimulation by means of infusions. of serotonin 5-HT1A agonist in advanced Parkinsons
Nevertheless, it is worth trying simpler and more conven- disease. Mov Disord 20: 932936.
tional treatments as a first approach. Pallidotomy is radi- Barbeau A (1976). Pathophysiology of the oscillations in
cally useful against LID but nowadays rarely indicated performance after long-term therapy with L-DOPA. In:
because of the limited unilateral effect. DBS of the GPi W Birkmayer, O Hornykiewicz (Eds.), Advances in Par-
or the STN is also highly efficacious in the treatment of kinsonism. Roche Editions, Basel. pp. 424428.
LID, perhaps through different mechanisms than simple Baron MS, Vitek JL, Bakay RAE et al. (2000). Treatment of
blockade of BG output. In patients with very severe and advanced Parkinsons disease by unilateral posterior GPi
complex LID, GPi-DBS may be a safer indication, even pallidotomy: 4 years results of a pilot study. Mov Disord
15: 230237.
when pallidal surgery is not associated with a significant
Bartoszyk GD, van Amsterdam C, Greiner HE et al.
reduction in daily levodopa intake.
(2004). Sarizotan, a serotonin 5-HT1A receptor agonist
Advances in neuroprotection and better means of and dopamine receptor ligand 1. Neurochemical profile.
replacing the dopaminergic striatal deficit should lead J Neural Transm 111: 113126.
to a gradual reduction of LID in PD in the near future. Bejjani B, Damier P, Arnoulf I et al. (1997). Pallidal stimu-
Eventually, LID will become an obsolete neurological lation for Parkinsons disease. Two targets? Neurology 49:
complication. 15641569.
Bejjani B, Gervais D, Arnulf I et al. (2000). Axial parkinso-
References nian symptoms can be improved: the role of levodopa and
bilateral subthalamic stimulation. J Neurol Neurosurg
Albin RL, Young AB, Penney JB (1989). The functional Psychiatry 68: 595600.
anatomy of basal ganglia disorders. Trends Neurosci 12: Belanger N, Gregoire L, Hadj Tahar A et al. (2003). Chronic
366375. treatment with small doses of cabergoline prevents dopa-
Alkhani A, Lozano AM (2001). Pallidotomy for Parkinsons induced dyskinesias in parkinsonian monkeys. Mov Disord
disease: a review of contemporary literature. J Neurosurg 18: 14361441.
94: 4349.
Bellforte JE, Pazo JH (2004). Turning behaviour induced by
Alonso-Frech F, Zamarbide I, Alegre M et al. (2006). Slow
stimulation of the 5-HT receptors in the subthalamic
oscillatory activity and levodopa-induced dyskinesias in
nucleus. Eur J Neurosci 19: 346355.
Parkinsons disease. Brain 129: 17481757.
Benabid AL, Pollak P, Hommel M et al. (1989). Traitement
Alvarez L, Macias R, Guridi J et al. (2001). Dorsal subthala-
du tremblement parkinsonien par stimulation chronique du
motomy for Parkinsons disease. Mov Disord 16: 7278.
noyau ventral intermediere du thalamus. Rev Neurol 145:
Alvarez L, Macias R, Lopez G et al. (2005). Bilateral subtha-
320323.
lamotomy in Parkinsons disease: initial and long term
Benabid AL, Pollak P, Gao JM et al. (1996). Chronic electri-
response. Brain 128: 570583.
cal stimulation of the ventralis intermedius nucleus of the
Andersson M, Hilbertson A, Cenci MA (1999). Striatal fosB
thalamus as a treatment of movement disorders. J Neuro-
expression is causally linked with L-Dopa-induced abnor-
surg 84: 203214.
mal involuntary movements and the associates upregula-
Bergman H, Wichmann T, Karmon B et al. (1994). The
tion of striatal prodynorphin mRNA in a rat model of
primate subthalamic nucleus. II. Neuronal activity in the
Parkinsons disease. Neurobiol Dis 6: 461474.
MPTP model of parkinsonism. J Neurophysiol 72:
Anglade P, Mouatt-Pringent A, Agid Y et al. (1996). Synaptic
507520.
plasticity in the caudate nucleus of patients with Parkin-
sons disease. Neurodegeneration 5: 121128. Betarbet R, Poisik O, Sherer TB et al. (2004). Differential
Antonelli T, Fuxe K, Tomasini MC et al. (2005). Effects of expression and ser897 phosphorylation of striatal N-
sarizotan on the corticostriatal glutamate pathways. methyl-d-aspartate receptor subunit NR1 in animal models
Synapse 58: 193199. of Parkinsons disease. Exp Neurol 187: 7685.
Asselin MC, Soghomonian JJ, Cote PY et al. (1994). Striatal Bezard E, Brotchie JM, Gross CE (2001). Pathophysiology
changes in preproenkephalin mRNA levels in parkinsonian of levodopa-induced dyskinesia: potential for new thera-
monkeys. Neuroreport 5: 21372140. pies. Nat Rev Neurosci 2: 577588.
Aubert I, Guigoni C, Hakansson K et al. (2005). Increased Bezard E, Ferry S, Mach U et al. (2003). Attenuation of
D1 dopamine receptor signaling in levodopa-induced levodopa-induced dyskinesia by normalizing dopamine
dyskinesia. Ann Neurol 57: 1726. D3 receptor function. Nat Med 9: 762767.
210 J. A. OBESO ET AL.

Bibbiani F, Oh JO, Chase TN (2001). Serotonin 5-HT1A Brotchie JM, Lee J, Venderova K (2005). Levodopa-induced
improves motor complications in rodent and primate par- dyskinesia in Parkinsons disease. J Neural Transm 112:
kinsonian models. Neurology 57: 18291834. 359391.
Blanchet PJ, Boucher R, Bedard PJ (1994). Excitotoxic lateral Brotchie P, Iansek R, Horne MK (1991). Motor function of
pallidotomy does not relieve L-dopa-induced dyskinesia in the monkey globus pallidus. 2. Cognitive aspects of move-
MPTP parkinsonian monkeys. Brain Res 65: 3239. ment and phasic neuronal activity. Brain 114: 16851702.
Blanchet PJ, Konitsiotis S, Whittemore ER et al. (1999). Diif- Brown P (2003). Oscillatory nature of human basal ganglia
fering effects of N-methyl-D-aspartate-receptor subtype activity: relationship to the pathophysiology of Parkin-
selective antagonists on dyskinesias in levodopa-treated sons disease. Mov Disord 18: 357363.
1-methyl-4-phenyl-tetrahydropyridine monkeys. J Phar- Burbaud P, Bonnet B, Guehl D et al. (1998). Movement dis-
macol Exp Ther 290: 10341040. orders induced by gamma-aminobutyric agonist and
Blandini F, Garcia-Osuna M, Greenamyre JT (1997). Subthala- antagonist injections into the internal globus pallidus and
mic ablation reverses changes in basal ganglio oxidative substantia nigra pars reticulata of the monkey. Brain Res
metabolism and motor response to apomorphine induced by 780: 102107.
nigrostriatal lesion in rats. Eur J Neurosci 9: 14071413. Burchiel K, Anderson V, Favre J et al. (1999). Comparison
Bonifati V, Fabrizio E, Cipriani R et al. (1994). Buspirone in of pallidal and subthalamic nucleus deep brain stimulation
levodopa-induced dyskinesias. Clin Neuropharmacol 17: for advanced Parkinsons disease: results of a randomized,
7382. blinded pilot study. Neurosurgery 45: 13751384.
Boraud T, Bezard E, Guehl D et al. (1998). Effects of L- Calabresi P, Mercuri NB, Sancesario G et al. (1993). Elec-
DOPA on neuronal activity of the globus pallidus externa- trophysiology of dopamine-denervated striatal neurons.
lis (GPe) and globus pallidus internalis (GPi) in the Implications for Parkinsons disease. Brain 116: 433452.
MPTP-treated monkey. Brain Res 787: 157160. Calabresi P, Pisani A, Mercuri NB et al. (1996). The corti-
Boraud T, Bezard E, Bioulac B et al. (2001). Dopamine costriatal projection: from synaptic plasticity to dysfunc-
agonist-induced dyskinesias are correlated to both firing tions of the basal ganglia. Trends Neurosci 19: 1924.
pattern and frequency alterations of pallidal neurones in Calabresi P, Giacomini P, Centonze D et al. (2000). Levodopa-
the MPTP-treated monkey. Brain 124: 546557. induced dyskinesia: a pathological form of striatal synaptic
Bordet R, Ridray S, Carboni S et al. (1997). Induction of plasticity? Ann Neurol 47: S60S68.
dopamine D3 receptor expression as a mechanism of beha- Calon F, Di Paolo T (2002). Levodopa response motor
vioral sensitization to levodopa. Proc Natl Acad Sci USA complications-GABA receptors and preproenkephalin
94: 33633367. expression in human brain. Parkinsonism Relat Disord 8:
Bordet R, Ridray S, Schwartz JC et al. (2000). Involvement 449454.
of the direct striatonigral pathway in levodopa-induced Calon F, Goulet M, Blanchet PJ et al. (1995). Levodopa or
sensitization in 6-hydroxydopamine-lesioned rats. Eur J D2 agonist induced dyskinesia in MPTP monkeys: corre-
Neurosci 12: 21172123. lation with changes in dopamine and GABA-A receptors
Bove J, Serrats J, Mengod G et al. (2005). Neuroprotection in the striatopallidal complex. Brain Res 680: 4352.
induced by the adenosine A (2A) antagonist CSC in the Calon F, Morissette M, Goulet M et al. (1999). Chronic D1 and D2
6-OHDA rat model of parkinsonism: effect on the activity dopaminomimetic treatment of MPTP-denervated monkeys:
of striatal output pathways. Exp Brain Res 165: 362374. effects on basal ganglia GABA(A)/benzodiazepine receptor
Bracco F, Battaglia A, Chouza C et al. (2004). PKDS009 complex and GABA content. Neurochem Int 35: 8191.
Study Group. The long-acting dopamine receptor agonist Calon F, Hadj Tahar A, Blanchet PJ et al. (2000). Dopamine-
cabergoline in early Parkinsons disease: final results of receptor stimulation: behavioral and biochemical conse-
a 5-year, double-blind, levodopa-controlled study. CNS quences. Trends Neurosci 23 (Suppl): S92S100.
Drugs 18: 733746. Calon F, Morissette M, Ghribi O et al. (2002). .Alteration of
Braz CA, Borges V, Ferraz HB (2004). Effect of riluzole on glutamate receptors in the striatum of dyskinetic 1-methyl-4-
dyskinesia and duration of the on state in Parkinson dis- phenyl-1,2,3,6-tetrahydropyridine-treated monkeys following
ease patients: a double-blind, placebo-controlled pilot dopamine agonist treatment. Prog Neuropsychopharmacol
study. Clin Neuropharmacol 27: 2529. Biol Psychiatry 26: 127138.
Bronstein JM, DeSalles A, DeLong MR (1999). for the Calon F, Rajput AH, Hornykiewicz O et al. (2003a). Levodopa-
Workshop Participants. Stereotactic pallidotomy in the induced motor complications are associated with alterations
treatment of Parkinsons disease. Arch Neurol 56: of glutamate receptors in Parkinsons disease. Neurobiol
10641069. Dis 14: 404416.
Brooks DJ, Ibanez V, Sawle GV et al. (1992). Striatal D-2 Calon F, Morissette M, Rajput AH et al. (2003b). Changes of
receptor status in patients with Parkinsons disease, stria- GABA receptors and dopamine turnover in the postmor-
tonigral degeneration, and progressive supranuclear palsy, tem brains of parkinsonians with levodopa-induced motor
measured with 11C-raclopride and positron emission complications. Mov Disord 18: 241253.
tomography. Ann Neurol 31: 184192. Calon F, Dridi M, Hornykiewicz O et al. (2004). Increased
Brooks DJ, Piccini P, Turjanski N et al. (2000). Neuroima- adenosine A2A receptors in the brain of Parkinsons
ging of dyskinesia. Ann Neurol 47 (Suppl 1): S154S158. disease patients with dyskinesias. Brain 127: 10751084.
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 211
Caparros-Lefebre D, Blond S, Feltin M-P et al. (1999). Improve- Crossman AR (1987). Primate models of dyskinesia: the
ment of levodopa induced dyskinesias by thalamic deep brain experimental approach to the study of basal ganglia-
stimulation is related to slight variation in electrode place- related involuntary movement disorders. Neuroscience
ment: possible involvement of the center median and parafas- 21: 140.
cicular complex. J Neurol Neurosurg Psychiatry 67: 308314. Crossman AR (1990). A hypothesis on the pathophysio-
Carpenter MB, Witthier JR, Mettler FA (1950). Analysis of logical mechanisms that underlie levodopa- or dopamine
choreoid hyperkinesias in the rhesus monkey. J Comp agonist-induced dyskinesia in Parkinsons disease: impli-
Neurol 92: 293322. cations for future strategies in treatment. Mov Disord 5:
Carpentier AF, Bonnet AM, Vidailhet M et al. (1996). Improve- 100108.
ment of levodopa-induced dyskinesia by propranolol in Crossman AR, Mitchell IJ, Sambrook MA et al. (1988). Chorea
Parkinsons disease. Neurology 46: 15481551. and myoclonus in the monkey induced by gamma-aminobu-
Carroll CB, Bain PG, Teare L et al. (2004). Cannabis for tyric acid antagonist in the lentiform complex: the site of
dyskinesia in Parkinson disease: a randomized double- drug action and a hypothesis for the neural mechanism of
blind crossover study. Neurology 63: 12451250. chorea. Brain 111: 12111233.
Carta AR, Pinna A, Cauli O et al. (2002). Differential regulation de la Fuente-Fernandez R, Sossi V, Huang Z et al. (2004).
of GAD67, enkephalin and dynorphin mRNAs by chronic- Levodopa-induced changes in synaptic dopamine levels
intermittent L-dopa and A2A receptor blockade plus L-dopa increase with progression of Parkinsons disease: implica-
in dopamine-denervated rats. Synapse 44: 166174. tions for dyskinesias. Brain 127: 27472754.
Carvalho AL, Duarte CB, Carvalho AP (2000). Regulation Del Dotto P, Pavese N, Gambaccini G et al. (2001). Intrave-
of AMPA receptors by phosphorylation. Neurochem Res nous amantadine improves levadopa-induced dyskinesias:
25: 12451255. an acute double-blind placebo-controlled study. Mov
Cenci MA (2002). Transcription factors involved in the Disord 16: 515520.
pathogenesis of L-DOPA-induced dyskinesia in a rat Delfs JM, Vivian M, Ciaramitaro M et al. (1995). Subthala-
model of Parkinsons disease. Amino Acids 23: 105109. mic nucleus lesions. Widespread effects on changes in
Cenci MA, Lee CS, Bjorklund A (1998). L-DOPA-induced gene expression induced by nigrostriatal dopamine deple-
dyskinesia in the rat is associated with striatal overexpres- tion in rats. J Neurosci 15: 65626575.
sion of prodynorphin- and glutamic acid decarboxylase DeLong MR (1990). Primate models of movement disorders
mRNA. Eur J Neurosci 10: 26942706. of basal ganglia origin. Trends Neurosci 13: 281285.
Centonze D, Calabresi P, Giacomini P et al. (1999). Neu- Denny-Brown D, Yanagisawa N (1976). The role of the
rophysiology of Parkinsons disease: from basic research basal ganglia in the initiation of movement. Res Publ
to clinical correlates. Clin Neurophysiol 110: 20062013. Assoc Res Nerv Ment Dis 55: 115149.
Centonze D, Gubellini P, Rossi S et al. (2005). Subthalamic Derome PJ, Jedynak CD, Visot A et al. (1986). Traitement
nucleus lesions reverses motor abnormalities and striatal des mouvements anormaux par lesions thalamiques. Rev
glutamatergic overactivity in experimental parkinsonism. Neurol 142: 391397.
Neuroscience 133: 831840. Dingledine R, Borges K, Bowie D et al. (1999). The gluta-
Chapuis S, Ouchchane L, Metz O et al. (2005). Impact of the mate receptor ion channels. Pharmacol Rev 51: 761.
motor complications of Parkinsons disease on the quality Doucet JP, Nakabeppu Y, Bedard PJ et al. (1996). Chronic
of life. Mov Disord 20: 224230. alterations in dopaminergic neurotransmission produce a
Chase TN, Oh JD (2000). Striatal dopamine- and glutamate- persistent elevation of deltaFosB-like protein(s) in both
mediated dysregulation in experimental parkinsonism. the rodent and primate striatum. Eur J Neurosci 8:
Trends Neurosci 23 (10 Suppl): S86S91. 365381.
Chase TN, Baronti F, Fabbrini G et al. (1989). Rationale for Dunah AW, Standaert DG (2001). Dopamine D1 receptor-
continuous dopaminomimetic therapy of Parkinsons dis- dependent trafficking of striatal NMDA glutamate receptors
ease. Neurology 39 (Suppl 2): 710. to the postsynaptic membrane. J Neurosci 21: 55465558.
Chase TN, Oh JD, Konitsiotis S (2000). Antiparkinsonian and Dunah AW, Wang Y, Yasuda RP et al. (2000). Alterations in
antidyskinetic activity of drugs targeting central glutamatergic subunit expression, composition, and phosphorylation of
mechanisms. J Neurol 247 (Suppl 2): 11361142. striatal N-methyl-D-aspartate glutamate receptors in a rat
Chen CC, Lee ST, Wu T et al. (2002). Hemiballism after sub- 6-hydroxydopamine model of Parkinsons disease. Mol
thalamotomy in patients with Parkinsons disease: report of Pharmacol 57: 342352.
2 cases. Mov Disord 17: 13671371. Dunah AW, Sirianni AC, Fienberg AA et al. (2004). Dopa-
Cooper IS (1969). Hemiballismus and hemichorea. In: IS mine D1-dependent trafficking of striatal N-methyl-D-
Cooper (Ed.), The Vital Probe: My life as a brain surgeon. aspartate glutamate receptors requires Fyn protein tyrosine
Morton and Company, New York, pp. 293315. kinase but not DARPP-32. Mol Pharmacol 65: 121129.
Cotzias GC, Papavasiliou PS, Gellene R (1969). Modification Durif F, Vidailhet M, Bonnet AM et al. (1995). Levodopa-
of parkinsonism-chronic treatment with L-dopa. N Engl J induced dyskinesias are improved by fluoxetine. Neurology
Med 280: 337345. 45: 18551858.
Crosby NJ, Deane KH, Clarke CE (2003). Amantadine for Durif F, Debilly B, Galitzky M et al. (2004). Clozapine
dyskinesia in Parkinsons disease. Cochrane Database Syst improves dyskinesias in Parkinson disease: a double-blind,
Rev CD003467. placebo-controlled study. Neurology 62: 381388.
212 J. A. OBESO ET AL.

Eidelberg D, Moeller JR, Ishikawa T et al. (1996). Regional Grondin R, Hadj Tahar A, Doan VD et al. (2000). Noradreno-
metabolic correlates of surgical outcome following unilat- ceptor antagonism with idazoxan improves L-dopa-
eral pallidotomy for Parkinsons disease. Ann Neurol 39: induced dyskinesias in MPTP monkeys. Naunyn Schmie-
450459. debergs Arch Pharmacol 361: 181186.
Fahn S (2000). The spectrum of levodopa-induced dyskine- Gubellini P, Pisani A, Centonze D et al. (2004). Metabotropic
sias. Ann Neurol 47 (Suppl 1): 211. glutamate receptors and striatal synaptic plasticity: implica-
Favre J, Burchiel KJ, Taha JM et al. (2000). Outcome of uni- tions for neurological diseases. Prog Neurobiol 74: 271300.
lateral and bilateral pallidotomy for Parkinsons disease: Guigoni C, Dovero S, Aubert I et al. (2005a). Levodopa-
patient assessment. Neurosurgery 46: 344355. induced dyskinesia in MPTP-treated macaques is not
Filion M, Tremblay L (1991). Abnormal spontaneous activ- dependent on the extent and pattern of nigrostriatal lesion-
ity of globus pallidus neurons in monkeys with MPTP- ing. Eur J Neurosci 22: 283287.
induced parkinsonism. Brain Res 547: 142151. Guigoni C, Li Q, Aubert I et al. (2005b). Involvement of sen-
Filion M, Treblay L, Bedard PJ (1991). Effects of dopamine sorimotor, limbic, and associative basal ganglia domains
agonist on the spontaneous activity of the globus pallidus in L-3,4-dihydroxyphenylalanine-induced dyskinesia.
neurons in monkeys with MPTP-induced parkinsonism. J Neurosci 25: 21022107.
Brain Res 547: 152161. Guridi J, Obeso JA (2001). The subthalamic nucleus, hemi-
Foffani G, Ardolino G, Meda B et al. (2005). Altered ballismus and Parkinsons disease: reappraisal of a surgi-
subthalamo-pallidal synchronisation in parkinsonian cal dogma. Brain 124: 519.
dyskinesias. J Neurol Neurosurg Psychiatry 76: 426428. Guridi J, Luquin MR, Herrero MT et al. (1993). The subtha-
Ford B, Winfield L, Pullman SL et al. (2004). Subthalamic lamic nucleus: a possible target for stereotaxic surgery in
nucleus stimulation in advanced Parkinsons disease: Parkinsons disease. Mov Disord 8: 421429.
blinded assessment at one year follow up. J Neurol Neuro- Guridi J, Herrero MT, Luquin MR et al. (1996). Subthalamot-
surg Psychiatry 75: 12551259. omy in parkinsonian monkeys. Behavioural and biochem-
Fox MW, Ahlskog JE, Kelly PJ (1991). Stereotactic ventro- ical analysis. Brain 119: 17171727.
lateralis thalamotomy for medically refractory tremor in Hallett PJ, Dunah AW, Ravenscroft P et al. (2005). Altera-
post-levodopa era Parkinsons disease patients. J Neuro- tions of striatal NMDA receptor subunits associated with
surg 75: 723730. the development of dyskinesia in the MPTP-lesioned pri-
Francois C, Savy C, Jan C et al. (2000). Dopaminergic inner- mate model of Parkinsons disease. Neuropharmacology
vation of the subthalamic nucleus in the normal state, in 48: 503516.
MPTP-treated monkeys, and in Parkinsons disease Hamada I, DeLong MR (1992). Excitotoxic acid lesions of
patients. J Comp Neurol 425: 121129. the primate subthalamic nucleus result in transient dyski-
Freed CR, Greene PE, Breeze RE et al. (2001). Transplanta- nesias of the contralateral limbs. J Neurophysiol 68:
tion of embryonic dopamine neurons for severe Parkin- 18501858.
sons disease. N Engl J Med 344: 710719. Hamani C, Richter E, Schwalb J et al. (2005). Bilateral sub-
Genderman GL, Ronesi J, Lovinger DM (2002). Postsynaptic thalamic nucleus stimulation for Parkinsons disease: a
endocannabinoid release is critical to long-term depression systematic review of the clinical literature. Neurosurgery
in the striatum. Nat Neurosci 5: 445451. 56: 13131324.
Gerfen CR (1995). Molecular effects of dopamine on striatal Hamel W, Fietzek U, Morsnowski A et al. (2003). Deep
projections pathways. Trends Neurosci 23: S64S70. brain stimulation of the subthalamic nucleus in Parkin-
Gerfen CR, Engber TM, Mahan LC et al. (1990). D1 and D2 sons disease: evaluation of active electrode contacts.
dopamine receptor-regulated gene expression of striatoni- J Neurol Neurosurg Psychiatry 74: 10361046.
gral and striatopallidal neurons. Science 250: 14291432. Hassler R (1978). Striatal control of locomotion, intentional
Ghika J, Villemure J-G, Fankhauser H et al. (1998). Efficiency actions and of integrating and perceptive activity. J Neurol
and safety of bilateral contemporaneous pallidal stimula- Sci 36: 187224.
tion (deep brain stimulation) in levodopa-responsive Hassler R, Riechert T (1954). Indikationen und lokalisations
patients with Parkinsons disease with severe fluctuations: methode der gezieten hirnoperationen. Nervenarzt 25:
a 2-year follow-up review. J Neurosurg 89: 713718. 441447.
Grabli D, McCairn K, Hirsch EC et al. (2004). Behavioural Hassler R, Riechert T, Mundinger F et al. (1960). Physiologi-
disorders induced by external globus pallidus dysfunction cal observations in stereotaxic operations in extrapyramidal
in primates: I. Behavioural study. Brain 127: 20392054. motor disturbances. Brain 83: 337349.
Graham WC, Sambrook MA, Crossman AR (1993). Differen- Hedreen JC (1999). Tyrosine hydroxylase-immunoreactive
tial effect of chronic dopaminergic treatment on dopamine elements in the human globus pallidus and subthalamic
D1 and D2 receptors in the monkey brain in MPTP- nucleus. J Comp Neurol 409: 400410.
induced parkinsonism. Brain Res 602: 290303. Henry B, Crossman AR, Brotchie JM (1999). Effect of
Grondin R, Bedard PJ, Hadj Tahar A et al. (1999). Anti- repeated L-DOPA, bromocriptine, or lisuride administra-
parkinsonian effect of a new selective adenosine A2A tion on preproenkephalin-A and preproenkephalin-B
receptor antagonist in MPTP-treated monkeys. Neurology mRNA levels in the striatum of the 6-hydroxydopamine-
52: 16731677. lesioned rat. Exp Neurol 155: 204220.
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 213

Henry B, Fox SH, Crossman AR et al. (2001). Mu- and marmoset and following MPTP and L-DOPA treatment.
delta-opioid receptor antagonists reduce levodopa-induced Brain Res 709: 259264.
dyskinesia in the MPTP-lesioned primate model of Parkin- Hurley MJ, Stubbs CM, Jenner P et al. (1996b). D3 receptor
sons disease. Exp Neurol 171: 139146. expression within the basal ganglia is not affected by
Henry B, Duty S, Fox SH et al. (2003). Increased striatal pre- Parkinsons disease. Neurosci Lett 214: 7578.
proenkephalin B expression is associated with dyskinesia Inase M, Buford JA, Anderson ME (1996). Changes in the
in Parkinsons disease. Exp Neurol 183: 458468. control of arm position, movement, and thalamic dis-
Herrero MT, Augood SJ, Hirsch EC et al. (1995). Effects of charge during local inactivation in the globus pallidus of
L-DOPA on preproenkephalin and preprotachykinin gene the monkey. J Neurophysiol 75: 10871104.
expression in the MPTP-treated monkey striatum. Neu- Intemann PM, Masterman D, Subamanian I et al. (2001).
roscience 68: 11891198. Staged bilateral pallidotomy for treatment of Parkinson
Herrero MT, Augood SJ, Asensi H et al. (1996a). Effects of disease. J Neurosurg 94: 437444.
L-dopa therapy on dopamine D-2 receptor mRNA expres- Inzelberg R, Schechtman E, Nisipeanu P (2003). Cabergoline,
sion in the striatum of MPTP-intoxicated parkinsonian pramipexole and ropinirole used as monotherapy in early
monkeys. Brain Res Mol Brain Res 42: 149155. Parkinsons disease: an evidence-based comparison. Drugs
Herrero MT, Levy R, Ruberg M et al. (1996b). Consequence Aging 20: 847855.
of nigrostriatal denervation and L-dopa therapy on the Jacobs BL, Fornal CA (1993). 5-HT and motor control: a
expression of glutamic acid decarboxylase messenger hypothesis. Trends Neurosci 16: 346352.
RNA in the pallidum. Neurology 47: 219224. Jan C, Francois C, Tande D et al. (2000). Dopaminergic
Hershey T, Black KJ, Stambuk MK et al. (1998). Altered innervation of the pallidum in the normal state, in
thalamic response to levodopa in Parkinsons patients with MPTP-treated monkeys and in parkinsonian patients. Eur
dopa-induced dyskinesias. Proc Natl Acad Sci USA 95: J Neurosci 12: 45254535.
1201612021. Jankovic J, Cardoso F, Grossman RG et al. (1995). Outcome
Herzog J, Volkmann J, Krack P et al. (2003). Two-year after stereotactic thalamotomy for parkinsonism, essential,
follow-up of subthalamic deep brain stimulation in Parkin- and others types of tremor. Neurosurgery 37: 680687.
sons disease. Mov Disord 18: 13321337. Kaelin-Lang A, Liniger P, Probst A et al. (2000). Adenosine
Hikosaka O, Wurtz RH (1983). Visual and oculomotor func- A2A receptor gene expression in the normal striatum and after
tions of monkey substantia nigra pars reticulata. I. Relation 6-OH-dopamine lesion. J Neural Transm 107: 851859.
of visual and auditory responses to saccades. J Neurophysiol Kanda T, Jackson MJ, Smith LA et al. (1998). Adenosine
49: 12301253. A2A antagonist: a novel antiparkinsonian agent that does
Hirsch E, Perier C, Orieux G et al. (2000). Metabolic effects not provoke dyskinesia in parkinsonian monkeys. Ann
of nigrostriatal denervation in basal ganglia. Trends Neu- Neurol 43: 507513.
rosci 23: (Suppl): 7885. Kanda T, Jackson MJ, Smith LA et al. (2000). Combined use
Hollmann M, Heinemann S (1994). Cloned glutamate recep- of the adenosine A (2A) antagonist KW-6002 with L-
tors. Annu Rev Neurosci 17: 31108. DOPA or with selective D1 or D2 dopamine agonists
Holloway RG, Shoulson I, Fahn S et al. (2004). Parkinson increases antiparkinsonian activity but not dyskinesia in
Study Group Pramipexole vs levodopa as initial treatment MPTP-treated monkeys. Exp Neurol 162: 321327.
for Parkinson disease: a 4-year randomized controlled Kannari K, Yamato H, Shen H et al. (2001). Activation of 5-
trial. Arch Neurol 61: 10441053. HT1A, but not 5-HT1 B receptors attenuates an increase in
Horak FB, Anderson ME (1984). Influence of globus palli- extracellular dopamine derived from exogenously admi-
dus on arm movements in monkeys. I. Effects of kainic nistered L-Dopa in the striatum with nigrostriatal denerva-
acid-induced lesions. J Neurophysiol 52: 290304. tion. J Neurochem 76: 13461353.
Horowski R, Marsden CD, Obeso JA (1988). Continuous dopa- Kato M, Kimura M (1992). Effects of reversible blockade of
minergic stimulation for Parkinsons disease: state of the art basal ganglia on a voluntary arm movement. J Neurophy-
and outlook. J Neural Trans 27 (Suppl): 249252. siol 68: 15161534.
Houeto JL, Damier P, Bejjani PB et al. (2000). Suthalamic Katzenschlager R, Hughes A, Evans A et al. (2005). Contin-
stimulation in Parkinson disease. A multidisciplinary uous subcutaneous apomorphine therapy improves dyski-
approach. Arch Neurol 57: 461465. nesias in Parkinsons disease: a prospective study using
Hoyer D, Hannon JP, Martin GR (2002). Molecular, pharma- single-dose challenges. Mov Disord 20: 151157.
cological and functional diversity of 5-HT receptors. Phar- Kelly PJ, Gillingham FJ (1980). The long term of stereotaxic
macol Biochem Behav 71: 533554. surgery and L-Dopa therapy in patients with Parkinsons
Hsu A, Togasaki DM, Bezard E et al. (2004). Effect of the disease. J Neurosurg 53: 332337.
D3 dopamine receptor partial agonist BP897 [N-[4- Kim JS, Lee KS, Lee KH et al. (2002). Evidence of thalamic
(4-(2-methoxyphenyl)piperazinyl)butyl]-2-naphthamide] disinhibition in patients with hemichorea: semiquantitative
on L-3,4-dihydroxyphenylalanine-induced dyskinesias analysis using SPECT. J Neurol Neurosurg Psychiatry 72:
and parkinsonism in squirrel monkeys. J Pharmacol Exp 329333.
Ther 311: 770777. Kleedorfer B, Lees AJ, Stern GM (1991). Buspirone in the
Hurley MJ, Jolkkonen J, Stubbs CM et al. (1996a). Dopa- treatment of levodopa induced dyskinesias. J Neurol
mine D3 receptors in the basal ganglia of the common Neurosurg Psychiatry 54: 376377.
214 J. A. OBESO ET AL.

Kleiner-Fisman, Fisman D, Sime E et al. (2003). Long- Lokk J (2004). Memantine can relieve certain symptoms in
term follow-up of bilateral deep brain stimulation of the Parkinson disease. Improvement achieved in two out of
subthalamic nucleus in patients with advanced three described cases with dyskinesia and cognitive fail-
Parkinsons disease. J Neurosurg 99: 489495. ure. Lakartidningen 101: 20032006.
Konitsiotis S, Blanchet PJ, Verhagen L et al. (2000). AMPA Lozano AM (2001). The subthalamic nucleus: myth and
receptor blockade improves levodopa-induced dyskinesia opportunities. Mov Disord 16: 183184.
in MPTP monkeys. Neurology 54: 15891595. Lozano AM, Lang AE, Levy R et al. (2000). Neuronal
Krack P, Pollak P, Limousin P et al. (1998). Opposite motor recordings in Parkinsons disease patients with dyskinesias
effects of pallidal stimulation in Parkinsons disease. Ann induced by apomorphine. Ann Neurol 47 (4 Suppl 1):
Neurol 43: 180192. S141S146.
Krack P, Pollak P, Limousin P et al. (1999). From off period Luginger E, Wenning GK, Bosch S et al. (2000). Beneficial
dystonia to peak dose chorea: the clinical spectrum of vary- effects of amantadine on L-dopa-induced dyskinesias in
ing subthalamic nucleus activity. Brain 122: 11331146. Parkinsons disease. Mov Disord 15; 873878.
Krack P, Batir A, Van Blercom N et al. (2003). Five-year follow- Lundblad M, Vaudano E, Cenci MA (2003). Cellular and
up of bilateral stimulation of the subthalamic nucleus in behavioural effects of the adenosine A2a receptor antago-
advanced Parkinsons disease. N Eng J Med 349: 19251934. nist KW-6002 in a rat model of l-DOPA-induced dyskine-
Laitinen LV, Bergerheim T, Hariz MI (1992). Leksells pos- sia. J Neurochem 84: 13981410.
teroventral pallidotomy in the treatment of Parkinsons Luquin MR, Laguna J, Obeso JA (1992a). Selective D2
disease. J Neurosurg 76: 5361. receptor stimulation induces dyskinesia in parkinsonian
Lang AE, Lozano AM, Montgomery E et al. (1997). Postero- monkeys. Ann Neurol 31: 551554.
ventral pallidotomy in advanced Parkinsons disease. N Luquin MR, Scipioni O, Vaamonde J et al. (1992b). Levodopa-
Engl J Med 337: 10361042. induced dyskinesias in Parkinsons disease. Clinical and
Lange KW, Kornhuber J, Riederer P (1997). Dopamine/ pharmacological classification. Mov Disord 7: 117124.
glutamate interactions in Parkinsons disease. Neurosci Ma Y, Feigin A, Dhawan V et al. (2002). Dyskinesia after
Biobehav Rev 21: 393400. fetal cell transplantation for parkinsonism: a PET study.
Lee T, Seeman P, Rajput A et al. (1978). Receptor basis for Ann Neurol 52: 628634.
dopaminergic supersensitivity in Parkinsons disease. Manson AJ, Schrag A, Lees AJ (2000). Low-dose olanzapine
Nature 273: 5961. for levodopa induced dyskinesias. Neurology 55: 795799.
Levesque M, Parent A (2005). The striatofugal fiber system Manson AJ, Katzenschlager R, Hobart J et al. (2001). High
in primates: a reevaluation of its organization based on dose naltrexone for dyskinesias induced by levodopa.
single-axon tracing studies. Proc Natl Acad Sci USA J Neurol Neurosurg Psychiatry 70: 554556.
102: 18881893. Manson AJ, Turner K, Lees AJ (2002). Apomorphine mono-
Levesque D, Martres MP, Diaz J et al. (1995). A paradoxical therapy in the treatment of refractory motor complications
regulation of the dopamine D3 receptor expression sug- of Parkinsons disease: long-term follow-up study of 64
gests the involvement of an anterograde factor from dopa- patients. Mov Disord 17: 12351241.
mine neurons. Proc Natl Acad Sci USA 92: 17191723. Maratos EC, Jackson MJ, Pearce RK et al. (2001). Anti-
Levy R, Dostrovsky JO, Lang AE et al. (2001). Effects of parkinsonian activity and dyskinesia risk of ropinirole
apomorphine on subthalamic nucleus and globus pallidus and L-DOPA combination therapy in drug naive MPTP-
internus neurons in patients with Parkinsons disease. J lesioned common marmosets. Mov Disord 16: 631641.
Neurophysiol 86: 249260. Marin C, Jimenez A, Tolosa E et al. (2004). Bilateral subtha-
Levy R, Ashby P, Hutchison WD et al. (2002). Dependence lamic nucleus lesion reverses L-dopa-induced motor
of subthalamic nucleus oscillations on movement and fluctuations and facilitates dyskinetic movements in hemi-
dopamine in Parkinsons disease. Brain 125: 11961209. parkinsonian rats. Synapse 51: 140150.
Lieberman JA (2004). Dopamine partial agonists: a new Marek K, Jennings D, Seibyl J (2002). Do dopamine agonists
class of antipsychotic. CNS Drugs 18: 251267. or levodopa modify Parkinsons disease progression? Eur
Limousin P, Speelman JD, Gielen F et al. (1999). study col- J Neurol 9 (Suppl 3): 1522.
laborators. Multicentre European study of thalamic stimu- Marsden CD (1982). The mysterious motor function of basal
lation in parkinsonian and essential tremor. J Neurol ganglia. Neurology 32: 514539.
Neurosurg Psychiatry 66: 289296. Marsden CD, Obeso JA (1994). The functions of the basal
Lindefors N, Ungerstedt U (1990). Bilateral regulation of gluta- ganglia and the paradox of stereotaxic surgery in Parkin-
mate tissue and extracellular levels in caudate-putamen by sons disease. Brain 117: 877897.
midbrain dopamine neurons. Neurosci Lett 115: 248252. Marsden CD, Parkes J (1977). Success and problems of long-
Liu X, Ford-Dunn HL, Hayward GN et al. (2002). The oscil- term levodopa therapy in Parkinsons disease. The Lancet
latory activity in the parkinsonian subthalamic nucleus 1: 345349.
investigated using the macro-electrodes for deep brain Marsden CD, Parkes JS, Quinn NP (1981). Fluctuations of
stimulation. Clin Neurophysiol 113: 16671672. disability in Parkinsons disease clinical aspects. In: CD
Loher T, Burgunder JM, Pohle T et al. (2002). Long-term pallidal Marsden and S Fahn (Eds.), Neurology 2: Movement
deep brain stimulation in patients with advanced Parkinsons Disorders. Butterworth Scientific, London-New York, pp.
disease: 1-year follow-up study. J Neurosurg 96: 844853. 96122.
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 215

Martinez-Mir MI, Probst A, Palacios JM (1991). Adenosine A2 Mitchell IJ, Boyce S, Sambrook MA et al. (1992). A 2-deox-
receptors: selective localization in the human basal ganglia yglucose study of the effects of dopamine agonists on the
and alterations with disease. Neurosci 42: 697706. parkinsonian primate brain. Implications for the neural
Massey PV, Johnson BE, Moult PR et al. (2004). Differential mechanisms that mediate dopamine agonist-induced dys-
roles of NR2A and NR2B-containing NMDA receptors in kinesias. Brain 115: 808824.
cortical long-term potentiation and long-term depression. J Molinuevo J, Valldeoriola F, Tolosa E et al. (2000). Levodo-
Neurosci 24: 78217828. pa withdrawal alter bilateral subthalamic nucleus stimula-
Meco G, Fabrizio E, Di Rezze S et al. (2003). Mirtazapine in tion in advanced Parkinsons disease. Arch Neurol 57:
L-dopa-induced dyskinesias. Clin Neuropharmacol 26: 983988.
179181. Monyer H, Sprengel R, Schoepfer R et al. (1992). Hetero-
Meissner W, Ravenscroft P, Reese R et al. (2006). Increased meric NMDA receptors: molecular and functional distinc-
slow oscillatory activity in substantia nigra pars reticulata tion of subtypes. Science 256: 12171221.
triggers abnormal involuntary movements in the 6-OHDA- Morissette M, Goulet M, Soghomonian JJ et al. (1997). Pre-
lesioned rat in the presence of excessive extracellular proenkephalin mRNA expression in the caudate-putamen
striatal dopamine. Neurobiol Dis 22: 586598. of MPTP monkeys after chronic treatment with the D2
Menegoz M, Lau LF, Herve D et al. (1995). Tyrosine phos- agonist U91356A in continuous or intermittent mode of
phorylation of NMDA receptor in rat striatum: effects of administration: comparison with L-DOPA therapy. Brain
6-OH-dopamine lesions. Neuroreport 7: 125128. Res Mol Brain Res 49: 5562.
Merello M, Cammarota A, Betti O et al. (1997). Involuntary Morissette M, Goulet M, Grondin R et al. (1998). Associative
movements during thermolesion predict a better outcome and limbic regions of monkey striatum express high levels of
after microelectrode guided posteroventral pallidotomy. dopamine D3 receptors: effects of MPTP and dopamine ago-
J Neurol Neurosurg Psychiatry 63: 210213. nist replacement therapies. Eur J Neurosci 10: 25652573.
Merello M, Balej J, Delfino M et al. (1999a). Apomorphine Morissette M, Dridi M, Calon F et al. (2006). Prevention
induces changes in GPi spontaneous outflow in patients of dyskinesia by an NMDA receptor antagonist in MPTP
with Parkinsons disease. Mov Disord 14: 4549. monkeys: effect on adenosine A2A receptors. Synapse
Merello M, Nouzeilles MI, Cammarota A et al. (1999b). 60: 239250.
Effect of memantine (NMDA antagonist) on Parkinsons Moro E, Scerrati M, Romito LM et al. (1999). Chronic sub-
disease: a double-blind crossover randomized study. Clin thalamic stimulation requirements in Parkinsons disease.
Neuropharmacol 22: 273276. Neurology 53: 8590.
Merello M, Starkstein S, Nouzeilles MI et al. (2001). Bilat- Moro E, Esselink R, Benabid AL et al. (2002). Response to
eral pallidotomy for treatment of Parkinsons disease levodopa in parkinsonian patients with bilateral subthala-
induced corticobulbar syndrome and psychic akinesia mic nucleus stimulation. Brain 125: 24082417.
avoidable by globus pallidus lesion combined with con- Mouradian MM, Heuser IJ, Baronti F et al. (1990). Modifica-
tralateral stimulation. J Neurol Neurosurg Psychiatry 71: tion of central dopaminergic mechanisms by continuous
611614. levodopa therapy for advanced Parkinsons disease. Ann
Merello M, Perez Lloret S, Antico J et al. (2006). Dyskinesias Neurol 27: 1823.
induced by subthalamotomy in Parkinsons disease are Muenter MD, Sharpless NS, Tyce GM et al. (1977). Patterns
unresponsive to amantadine. J Neurol Neurosurg Psychiatry of dystonia (I-D-I and D-I-D) in response to L-dopa
77: 172174. therapy of Parkinsons disease. Mayo Clinic Proc 52:
Meshul CK, Emre N, Nakamura CM et al. (1999). Time- 163174.
dependent changes in striatal glutamate synapses follow- Murray TK, Messenger MJ, Ward MA et al. (2002). Evalua-
ing a 6-hydroxydopamine lesion. Neurosci 88: 116. tion of the mGluR2/3 agonist LY379268 in rodent models
Miller DW, Abercrombie ED (1999). Role of high-affinity of Parkinsons disease. Pharmacol Biochem Behav 73:
dopamine uptake and impulse activity in the appearance 455466.
of extracellular dopamine in striatum after administration Narabayashi H, Yokochi F, Nakajima Y (1984). Levodopa-
of exogenous L-DOPA: studies in intact and 6-hydroxydo- induced dyskinesia and thalamotomy. J Neurol Neurosurg
pamine-treated rats. J Neurochem 72: 15161522. Psychiatry 47: 831839.
Mink JW (2003). The basal ganglia and involuntary move- Nisbet AP, Foster OJ, Kingsbury A (1995). Preproenkephalin
ments: impaired inhibition of competing motor patterns. and preprotachykinin messenger mRNA expression in nor-
Arch Neurol 60: 13651368. mal human basal ganglia and in Parkinsons disease. Neu-
Mink JW, Thach WT (1991). Basal ganglia motor control III. roscience 66: 361376.
Pallidal ablation: normal reaction time, muscle contrac- Nyholm D, Nilsson Remahl AI, Dizdar N et al. (2005). Duo-
tion, and slow movement. J Neurophysiol 65: 330351. denal levodopa infusion monotherapy vs oral polyphar-
Mitchell IJ, Jackson A, Sambrook MA et al. (1989). The role macy in advanced Parkinson disease. Neurology 64:
of the subthalamic nucleus in experimental chorea. 216223.
Evidence from 2-deoxyglucose metabolic mapping and Obeso JA, Luquin MR, Martinez-Lage JM (1986). Lisuride
horseradish peroxidase tracing studies. Brain 112: infusion pump: a device for the treatment of Parkinsons
15331548. disease. Lancet 1: 467470.
216 J. A. OBESO ET AL.

Obeso JA, Grandas F, Vaamonde J et al. (1989). Motor com- parkinsonism and chronic levodopa treatment. Brain 125:
plications associated with chronic levodopa therapy in 26352645.
Parkinsons disease. Neurology 39 (Suppl 2): 1118. Picconi B, Centonze D, Hakansson K et al. (2003). Loss of
Obeso JA, Grandas F, Herrero MT et al. (1994). The role of bidirectional striatal synaptic plasticity in L-DOPA-
pulsatile versus continous dopamine receptor stimulation induced dyskinesia. Nat Neurosci 6: 501506.
for functional recovery in Parkinsons disease. Eur J Neu- Picconi B, Gardoni F, Centonze D et al. (2004). Abnormal
rosci 6: 889897. Ca2-calmodulin-dependent protein kinase II function
Obeso JA, Rodriguez MC, DeLong MR (1997). Basal ganglia mediates synaptic and motor deficits in experimental par-
pathophysiology. A critical review. Adv Neurol 74: 318. kinsonism. J Neurosci 24: 52835291.
Obeso JA, Rodriguez-Oroz MC, Rodriguez M et al. (2000a). Pinna A, Corsi C, Carta AR et al. (2002). Modification of
Pathophysiology of the basal ganglia in Parkinsons dis- adenosine extracellular levels and adenosine A (2A)
ease. Trends Neurosci 23 (10 Suppl): S8S19. receptor mRNA by dopamine denervation. Eur J Pharma-
Obeso JA, Rodrguez-Oroz MC, Rodrguez M et al. (2000b). col 446: 7582.
Current problems in understanding the basal ganglia Pinna A, Wardas J, Simola N et al. (2005). New therapies for
model for Parkinsons disease and levodopa-induced dys- the treatment of Parkinsons disease: Adenosine A (2A)
kinesias. Ann Neurol 47 (Suppl 1): 2232. receptor antagonists. Life Sci 77: 32593267.
Obeso JA, Olanow CW, Rodriguez-Oroz MC et al. (2001). Pirtosek Z, Merello M, Carlsson A et al. (1993). Preclamol
Deep Brain Stimulation for Parkinsons Disease Study and parkinsonian fluctuations. Clin Neuropharmacol 16:
Group. Deep brain stimulation of the subthalamic nucleus 550554.
or the pars interna of the globus pallidus in Parkinsons Pisani A, Bonsi P, Catania MV et al. (2002). Metabotropic
disease. N Engl J Med 345: 956963. glutamate 2 receptors modulate synaptic inputs and cal-
Obeso JA, Rodriguez-Oroz M, Marin C et al. (2004). The cium signals in striatal cholinergic interneurons. J Neu-
origin of motor fluctuations in Parkinsons disease: impor- rosci 22: 61766185.
tance of dopaminergic innervation and basal ganglia Pisani A, Centonze D, Bernardi G et al. (2005). Striatal
circuits. Neurology 62 (Suppl 1): S17S30. synaptic plasticity: implications for motor learning and
Oh JD, Del Dotto P, Chase TN (1997). Protein kinase A inhi- Parkinsons disease. Mov Disord 20: 395402.
bitor attenuates levodopa-induced motor response altera- Quik M, Police S, He L et al. (2000). Expression of D(3)
tions in the hemi-parkinsonian rat. Neurosci Lett 228: 58. receptor messenger RNA and binding sites in monkey
Oh JD, Bibbiani F, Chase TN (2002). Quetiapine attenuates striatum and substantia nigra after nigrostriatal degenera-
levodopa-induced motor complications in rodent and pri- tion: effect of levodopa treatment. Neurosci 98: 263273.
mate parkinsonian models. Exp Neurol 177: 557564. Quik M, Police S, Langston JW et al. (2002). Increases
Ohye C, Maeda T, Narabayashi H (1976). Physiologically in striatal preproenkephalin gene expression are associa-
defined Vim nucleus, its special reference to the control ted with nigrostriatal damage but not L-DOPA-induced
of tremor. Appl Neurophysiol 39: 285295. dyskinesias in the squirrel monkey. Neurosci 113:
Olanow CW, Goetz CG, Kordower JH et al. (2003). A dou- 213220.
ble blind placebo-controlled trial of bilateral fetal nigral Rabiner EA, Gunn RN, Wilkins MR et al. (2002). Evaluation
transplantation in Parkinsons disease. Ann Neurol 54: of EMD128130 occupancy of the 5HT1A and the D2
403414. receptor: a human PET study with [11C]WAY-100365
Page RD, Sambrook MA, Crossman AR (1993). Thalamot- and [11C]-raclopride. J Psychopharmacol 16: 195199.
omy for the alleviation of levodopa-induced dyskinesia: Rascol O, Fabre N, Blin O et al. (1994). Naltrexone, an opi-
experimental studies in the 1-methyl-4-phenyl-1,2,3,6-tet- ate antagonist, fails to modify motor symptoms in patients
rahydropyridine treated parkinsonian monkey. Neurosci with Parkinsons disease. Mov Disord 9: 437440.
55: 147165. Rascol O, Sabatini U, Brefel C et al. (1998). Cortical motor
Papa SM, Chase TN (1996). Levodopa-induced dyskinesias overactivation in parkinsonian patients with L-Dopa
improved by a glutamate antagonist in Parkinsonian mon- induced peak-dose dyskinesia. Brain 121: 527533.
keys. Ann Neurol 39: 574578. Rascol O, Brooks DJ, Korczyn AD et al. (2000). A five-year
Papa SM, Desimone R, Fiorani M et al. (1999). Internal glo- study of the incidence of dyskinesia in patients with early
bus pallidus discharge is nearly suppressed during levo- Parkinsons disease who were treated with ropinirole or
dopa-induced dyskinesias. Ann Neurol 46: 732738. levodopa. 056 Study Group. N Engl J Med 342:
Patel N, Plaha P, OSullivan K et al. (2003). MRI directed 14841491.
bilateral stimulation of the subthalamic nucleus in patients Rascol O, Arnulf I, Peyro-Saint Paul H et al. (2001). Idazox-
with Parkinsons disease. J Neurol Neurosurg Psychiatry an, an alpha-2 antagonist, and L-DOPA-induced dyskine-
74: 16311637. sias in patients with Parkinsons disease. Mov Disord
Piccini P, Weeks RA, Brooks DJ (1997). Alterations in opioid 16: 708713.
receptor binding in Parkinsons disease patients with levo- Robertson RG, Farmery SM, Sambrook MA et al. (1989).
dopa-induced dyskinesias. Ann Neurol 42: 720726. Dyskinesia in the primate following injection of an excita-
Picconi B, Pisani A, Centonze D et al. (2002). Striatal metabo- tory amino acid antagonist into the medial segment of the
tropic glutamate receptor function following experimental globus pallidus. Brain Res 476: 317322.
LEVODOPA-INDUCED DYSKINESIAS IN PARKINSONS DISEASE 217

Rodriguez-Oroz MR, Obeso JA, Lang AE et al. (2005). Bilat- Stocchi F, Ruggieri S, Vacca L et al. (2002). Prospective
eral deep brain stimulation in Parkinsons disease: a multi- randomized trial of lisuride infusion versus oral levo-
centre study with 4 years follow-up. Brain 128: 22402249. dopa in patients with Parkinsons disease. Brain 125:
Samadi P, Gregoire L, Bedard PJ (2004). The opioid agonist 20582066.
morphine decreases the dyskinetic response to dopaminergic Su P, Tseng HM, Liu HM et al. (2002). Subthalamotomy for
agents in parkinsonian monkeys. Neurobiol Dis 16: 246253. advanced Parkinsons disease. J Neurosurg 97: 598606.
Samuel M, Ceballos Boumann AO, Turjanski N et al. Sung KW, Choi S, Lovinger DM (2001). Activation of group
(1997). Pallidotomy in Parkinsons disease increases sup- ImGluRs is necessary for long-term depression in striatal
plementary motor area and prefrontal activation during synapses. J Neurophysiol 86: 24052412.
performance of volitional movements. An H2O PET Svenningsson P, Le Moine C, Aubert I et al. (1998). Cellular
study. Brain 12: 13011313. distribution of adenosine A2A receptor mRNA in the
Sandyk R, Snider SN (1986). Naloxone treatment of primate striatum. J Comp Neurol 399: 299240.
L-Dopa-induced dyskinesias in Parkinsons disease. Am Swope SL, Moss SJ, Raymond LA et al. (1999). Regulation
J Psychiatry 143: 118. of ligand-gated ion channels by protein phosphorylation.
Santiago M, Matarredona ER, Machado A et al. (1998). Influ- Adv Second Messenger Phosphoprotein Res 33: 4978.
ence of serotoninergic drugs on in vivo dopamine extracel- Tanaka H, Kannari K, Maeda T et al. (1999). Role of serotoniner-
lular output in rat striatum. J Neurosci Res 52: 591598. gic neurons in L-Dopa derived extracellular dopamine in the
Shinotoh H, Hirayama K, Tateno Y (1993). Dopamine D1 and striatum of 6-OHDA-lesioned rats. Neuroreport 10: 631634.
D2 receptors in Parkinsons disease and striatonigral degen- Tasker RR (1990). Thalamotomy. Neurosurg Clin N Am 1:
eration determined by PET. Adv Neurol 60: 488493. 841864.
Shoulson I (1998). DATATOP: a decade of neuroprotective Tasker RR, Munz M, Junn FSCK et al. (1997). Deep brain
inquiry. Parkinson Study Group. Deprenyl And Tocopherol stimulation and thalamotomy for tremor compared. Acta
Antioxidative Therapy Of Parkinsonism. Ann Neurol Neurochir Suppl 68: 4953.
44 (Suppl 1): S160S166. Tekumalla PK, Calon F, Rahman Z et al. (2001). Elevated
Silberstein P, Kuhn AA, Kupsch A et al. (2003). Patterning levels of DeltaFosB and RGS9 in striatum in Parkinsons
of globus pallidus local field potentials differs between disease. Biol Psychiatr 50: 813816.
Parkinsons disease and dystonia. Brain 126: 25972608. Tel BC, Zeng BY, Cannizzaro C et al. (2002). Alterations in
Silverdale MA, Crossman AR, Brotchie JM (2002). Striatal striatal neuropeptide mRNA produced by repeated admin-
AMPA receptor binding is unaltered in the MPTP- istration of L-Dopa, ropinirole or bromocriptine correlate
lesioned macaque model of Parkinsons disease and dyski- with dyskinesia induction on MPTP-treated common mar-
nesia. Exp Neurol 174: 2128. mosets. Neuroscience 115: 10471058.
Simuni T, Jaggi JL, Mulholland H et al. (2002). Bilateral Thomas A, Iacono D, Luciano AL et al. (2004). Duration of
stimulation of the subthalamic nucleus in patients with amantadine benefit on dyskinesia of severe Parkinsons
Parkinsons disease: a study of efficacy and safety. disease. J Neurol Neurosurg Psychiatry 75: 141143.
J Neurosurg 96: 666672. Tomiyama M, Kimura T, Maeda T et al. (2005). A serotonin
Smith Y, Charara A, Parent A (1996). Synaptic innervation of 5-HT1A receptor agonist prevents behavioral sensitization
midbrain dopaminergic neurons by glutamate-enriched term- to levodopa in a rodent model of Parkinsons disease.
inals in the squirrel monkey. J Comp Neurol 364: 231253. Neurosci Res 52: 185194.
Snow BJ, Macdonald L, Mcauley D et al. (2000). The effect Touchon JC, Moore C, Frederickson C et al. (2004). Lesion
of amantadine on levodopa-induced dyskinesias in Parkin- of subthalamic or motor thalamic nucleus in 6-hydroxydo-
sons disease: a double-blind, placebo-controlled study. pamine-treated rats: effects on striatal glutamate and apo-
Clin Neuropharmacol 23: 8285. morphine-induced contralateral rotations. Synapse 15:
Sokoloff P, Giros B, Martres MP et al. (1990). Molecular 287298.
cloning and characterization of a novel dopamine receptor Trabucchi M, Bassi S, Frattola L (1982). Effect of naloxone
(D3) as a target for neuroleptics. Nature 347: 146151. on the on-off syndrome in patients receiving long-term
Standaert DG, Testa CM, Penney JB et al. (1994). Organization levodopa therapy. Arch Neurol 39: 120121.
of N-methyl-D-aspartate glutamate receptor gene expression Ulas J, Weihmuller FB, Brunner LC et al. (1994). Selective
in the basal ganglia of the rat. J Comp Neurol 64: 1123. increase of NMDA-sensitive glutamate binding in the
Stanford IM, Kantaria MA, Chahal HS et al. (2005). 5-Hydro- striatum of Parkinsons disease, Alzheimers disease, and
xytryptamine induced excitation and inhibition in the subtha- mixed Parkinsons disease/Alzheimers disease patients:
lamic nucleus: action at 5-HT2c, 5-HTA4 and 5-HT1A an autoradiographic study. J Neurosci 14: 63176324.
receptors. Neuropharmacology 49: 12281234. Vaamonde J, Luquin MR, Obeso JA (1991). Subcutaneous
Steiner H, Gerfen CR (1999). Enkephalin regulates acute D2 lisuride infusion in Parkinsons disease: response to chronic
dopamine receptor antagonist-induced immediate early administration in 34 patients. Brain 114: 601614.
gene expression in striatal neurons. Neurosci 88: 795810. Verhagen Metman L, Del Dotto P, Blanchet PJ et al. (1998a).
Stephens B, Mueller AJ, Shering AF et al. (2005). Evidence Blockade of glutamatergic transmission as treatment for
of a breakdown of corticostriatal connections in Parkin- dyskinesias and motor fluctuations in Parkinsons disease.
sons disease. Neurosci 132: 741754. Amino Acids 14: 7582.
218 J. A. OBESO ET AL.

Verhagen Metman L, Del Dotto P, Natte R et al. (1998b). Whone AL, Watts RL, Stoessl AJ et al. (2003), REAL-PET
Dextromethorphan improves levodopa-induced dyskine- Study Group. Slower progression of Parkinsons disease
sias in Parkinsons disease. Neurology 51: 203206. with ropinirole versus levodopa: the REAL-PET study.
Vila M, Levy R, Herrero MT et al. (1997). Consequences of Ann Neurol 54: 93101.
nigrostriatal denervation on the functioning of the basal Yelnik J, Damier P, Demeret S et al. (2003). Localization of
ganglia in human and nonhuman primates: an in situ stimulation electrodes in patients with Parkinson disease
hybridization study of cytochrome oxidase subunit I by using a three-dimensional atlas-magnetic resonance
mRNA. J Neurosci 17: 765773. coregistration method. J Neurosurg 99: 8999.
Vingerhoets FJG, Villemure JG, Temperli P et al. (2002). Zeng BY, Pearce RK, MacKenzie GM et al. (2000). Alterations
Subthalamic DBS replace levodopa in Parkinsons dis- in preproenkephalin and adenosine-2a receptor mRNA,
ease: two year follow-up. Neurology 58: 396401. but not preprotachykinin mRNA correlate with occurrence
Visser-Vanderwalle V, Van der Linden C, Temel Y et al. of dyskinesia in normal monkeys chronically treated with
(2003). Long term motor effect of unilateral pallidal sti- L-DOPA. Eur J Neurosci 12: 10961104.
mulation in 26 patients with advanced Parkinson disease.
J Neurosurg 99: 701707.
Vitek JL, Giroux M (2000). Physiology of hypokinetic and Further Reading
hyperkinetic movement disorders: model for dyskinesia.
Ann Neurol 47 (Suppl 1): S131S140. Bonifati V, Fabrizio E, Cipriani R et al. (1992). Buspirone
Vitek JL, Chockkan V, Zhang IY et al. (1999). Neuronal in levodopa-induced dyskinesias. Clin Neuropharmacol
activity in the basal ganglia in patients with generalized 17: 7382.
dystonia and hemiballismus. Ann Neurol 46: 2235. Hadj Tahar A, Gregoire L, Darre A et al. (2004). Effect of a
Volkmann J, Allert N, Voges J et al. (2004). Long term selective glutamate antagonist on L-dopa-induced dyski-
results of bilateral pallidal stimulation in Parkinsons nesias in drug-naive parkinsonian monkeys. Neurobiol
disease. Ann Neurol 55: 871875. Dis 15: 171176.
Whittier JR, Mettler FA (1949). Studies of the subthalamus in Katzenschlager R, Manson AJ, Evans A et al. (2004).
rhesus monkey. II. Hyperkinesia and other physiological Low dose quetiapine for drug induced dyskinesias in Par-
effects of the subthalamic lesion, with special reference to kinsons disease: a double blind cross over study. J Neurol
the subthalamic nucleus of Luys. J Comp Neurol 90: 319372. Neurosurg Psychiatry 75: 295297.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 41

Treatment-induced mental changes in Parkinsons disease

KELVIN L. CHOU AND JOSEPH H. FRIEDMAN*

Department of Clinical Neurosciences, Brown University Medical School and NeuroHealth Parkinsons Disease and
Movement Disorders Center, Warwick, RI, USA

41.1. Introduction This chapter provides an overview of the various


adverse behavioral and cognitive changes seen as a
Although Parkinsons disease (PD) is defined by the result of the pharmacological and surgical treatment
presence of its motor features alone, it has become of PD. The phenomenology and treatment of drug-
increasingly clear that PD is not just a neurological induced psychosis will be discussed in detail, and
disorder, but a neurobehavioral syndrome with well- mood fluctuations, as well as repetitive and compul-
documented mood, cognitive and behavioral symptoms. sive behaviors due to dopaminergic medications, will
Unfortunately, many of the behavioral abnormalities be reviewed. Recent literature highlighting the nega-
associated with PD are due to or exacerbated by the med- tive behavioral and cognitive changes from ablative
ications used to treat the myriad symptoms of surgeries and deep brain stimulation (DBS) for PD will
the disease. In addition, evidence is accumulating to also be covered.
show that surgical therapies may worsen or even cause
behavioral or cognitive decline in patients with PD.
41.2. Mental changes due to pharmacologic
Because these mental changes may occur late in the
treatment of Parkinsons disease
course of the illness, long after antiparkinsonian
medications have been started, it can be difficult to 41.2.1. Drug-induced psychosis
determine whether these changes are treatment-induced,
treatment-exacerbated or intrinsic to the disease process. Although psychosis used to be defined as a major
Further complicating the problem is the issue of mental disorder in which reality testing is impaired,
dementia with Lewy bodies (DLB) and our limited the current definition describes psychosis as a disorder
understanding of the dementia syndrome that often characterized by hallucinations, delusions or disorga-
occurs in PD (see Ch. 60). Whereas the presence of visual nized thinking (American Psychiatric Association,
hallucinations in an untreated state is one of the clinical 1994). Hallucinations are perceptions without any
hallmarks of DLB, visual hallucinations in DLB are basis in reality, i.e. seeing, hearing, smelling, tasting
indistinguishable from drug-induced hallucinosis in PD. or feeling things that are not present. These need to
Moreover, DLB is diagnosed clinically when the demen- be distinguished from illusions, which are distorted
tia occurs before or soon after the onset of parkinsonism perceptions, such as seeing an animal in a shadow,
(McKeith et al., 1996), yet it has become clear that the mistaking a distant tree for a person, seeing faces in
dementing aspect may develop years later (Apaydin flowers or misperceiving a motor sound for a voice.
et al., 2002). Finally, all of the dementing disorders, A delusion is a false and irrational belief that has no
including the dementia of PD and DLB, are associated foundation. Common examples include believing that
with a variety of psychotic and other behavioral symp- strangers are living in or moving objects around the
toms (Cahn-Weiner et al., 2002). These overlapping house, thinking that family members have been
clinical symptoms make it challenging to distinguish replaced by imposters and believing that one is being
between the different disorders. investigated by the Federal Bureau of Investigation.

*Correspondence to: Joseph H. Friedman, NeuroHealth Parkinsons Disease and Movement Disorders Center, 227 Centerville
Road, Warwick, RI 02886, USA. E-mail: Joseph_Friedman@brown.edu, Tel: 1-401-732-3332, Fax: 1-401-737-3623.
220 K. L. CHOU AND J. H. FRIEDMAN
Psychosis is not a feature of untreated PD. Whereas Table 41.1
visual hallucinations are typical of untreated DLB and
Types of delusions in 160 Parkinsons disease patients
delusions may occur in demented PD patients who are
with drug-induced psychosis
not taking medication, the vast majority of PD patients
who develop psychotic symptoms do so on PD Type of delusion Number of patients
medications and return to their non-psychotic baseline
if the PD medications are discontinued (Kofman, Stealing 53
1984; Friedman, 1985). Not my house 46
Psychosis affects 510% of drug-treated PD Abandonment 41
patients (Dewey and OSuilleabhain, 2000). There Spouse imposter 32
are three explanations for the occurrence of psychosis Infidelity 26
in this population. Most commonly, the medications Other 38 paranoid 21 non-paranoid
themselves are responsible. However, many psychoses
also undoubtedly occur because of the presence of imagined guests and water and food bowls are placed
dementia, whether due to PD dementia, DLB or some for dog and cat visitors. The police may be called to
other pathology (see Ch. 60). Additionally, a tiny per- catch burglars. The phenomenology of the delusions
centage of patients have a premorbid primary psycho- overlaps with those seen in other dementing illnesses
sis, such as schizophrenia, and later develop PD. The (Cahn-Weiner et al., 2002), as assessed in the Neuropsy-
importance of recognizing and treating psychosis in chiatric Inventory (Cummings, 1997). Family members
PD is illustrated by studies which have shown that or other people may be stealing money or discussing
psychosis, not motor dysfunction, is the single greatest things about the patient, strangers may be living in the
precipitant for nursing-home placement in PD patients house, patients may accuse the spouse of being an
(Goetz and Stebbins, 1993; Aarsland et al., 2000). imposter, trips are supposedly being taken without the
As might be deduced from this observation, the single patient and nursing-home placement is planned none
greatest stress for care-takers is also psychosis of which is true. Another common delusion is the
(Aarsland et al., 1999). In addition, the appearance of accusation of spousal infidelity, which affects both men
psychosis markedly increases the risk of mortality and women and is even more difficult to manage.
(Goetz and Stebbins, 1995; Factor et al., 2003). In
one double-blind, placebo-controlled treatment trial 41.2.1.2. Psychosis with an impaired sensorium or
of psychosis in PD, 10% of the subjects died during dementia
or shortly after completing a 3-month trial (Parkinson Unlike psychosis in patients with a clear sensorium,
Study Group, 1999). In another, 5% of the subjects these patients are delirious or demented and, as a
died (Pollak et al., 2004). result, manifest psychotic features. Delirious and
The clinical scenario for psychosis in PD is fairly demented patients with psychosis do less well, in all
stereotypic and can be divided into two major cate- respects. They are less responsive to and less tolerant
gories: psychosis with a clear sensorium and psychosis of antipsychotic drugs. The common antipsychotics
with a clouded sensorium. In both conditions patients used in managing PD patients with psychosis,
typically develop hallucinations. The latter condition clozapine and quetiapine, are sedating. Whereas this
is often termed an encephalopathy by neurologists side-effect is beneficial in helping the patient sleep at
and a delirium by psychiatrists. night, it may increase the confusion that occurs when
the patient awakens during the night, as well as pro-
41.2.1.1. Psychosis with a clear sensorium duce a hangover state in the morning. In a demented
These patients are much more like the primary or delirious patient, this increased daytime somnolence
psychiatric patient in that they will have a normal contributes to the delirium. In some cases, we believe
attention span, memory and cognition. The psychosis this worsened sleepiness may also blunt the response
is manifest primarily by hallucinations and, to a lesser to the anti-PD medications, resulting in worsened
extent, by delusions, which are generally of a paranoid motor function. Since both drugs may also cause or
nature. The most common delusion in a descriptive worsen orthostatic hypotension, precautions to prevent
analysis of two double-blind, placebo-controlled anti- fainting, especially after lying supine at night, must be
psychotic trials (Table 41.1) was stealing (Friedman maintained.
et al., 2002). However, not all patients have delusions.
When the hallucinations are so severe that they are 41.2.1.3. Hallucinations
believed to be real, then the patient is considered Hallucinations are not part of the untreated state of
psychotic. At this point, food may be set out for the idiopathic PD but were described with postencephalitic
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 221
parkinsonism (de Ajuriaguerra, 1972) and constitute a psychotic conditions, they are perceived as real.
cardinal feature of untreated DLB (McKeith et al., Auditory hallucinations frequently make demeaning
1996). In terms of frequency, visual hallucinations comments in schizophrenia whereas the drug-induced
are most commonly reported (Fenelon et al., 2000; PD hallucinations are not bothersome and may even
Holroyd et al., 2001). We analyzed baseline data be entertaining.
regarding hallucinations (Table 41.2) on 160 subjects Visual hallucinations occur in 944% of drug-
from two identical antipsychotic studies performed treated patients, depending on the survey (Meco
simultaneously in Europe and the USA (Friedman et al., 1990; Haeske-Derwick, 1995; Sanchez-Ramos
et al., 2002). DLB patients were excluded, but 58 et al., 1996; Graham et al., 1997; McDowell and
patients were demented (defined as Mini-Mental State Harris, 1997; Fenelon et al., 2000; Barnes and David,
Examination (MMSE) < 24). Visual hallucinations were 2001; Holroyd et al., 2001). The hallucinations may
the most prevalent, followed by auditory, olfactory occur at any time and with any type of lighting.
and then tactile hallucinations. These data were from Fenelon and colleagues (2000) included the term
subjects enrolled in double-blind placebo-controlled presence hallucinations to denote the vivid sensation
trials and are therefore not representative of the general of the presence of somebody either somewhere in the
PD population, but are probably representative of mild room or . . . behind him. This presence was usually
to moderately psychotic PD patients with drug-induced that of a person, but occasionally was an animal
psychosis. and, in one case, the patient felt a guardian angel.
There are marked differences between the halluci- They also described passage hallucinations, which
nations encountered in treated PD and those reported were brief visions that were seen in the periphery of
with primary psychiatric disorders such as schizophre- the visual field. These so-called minor hallucinations
nia (Table 41.3). Primary psychiatric hallucinations occurred slightly more frequently than formed visual
are usually auditory and ego-syntonic, whereas drug- hallucinations, which were usually of non-threatening
induced hallucinations in PD are generally visual and people, animals or objects. The hallucinations usually
without emotional content. This is true even when last under 5 minutes and many last for seconds only.
the hallucinated image should have some emotional They usually occur a few times a week, but are rarely
content, such as a deceased spouse, a deceased pet or omnipresent. The hallucinations may look clear and
a long-unseen loved one. In PD, most patients recog- real or hazy and out of focus. Occasional patients will
nize that the hallucinations are not real, but in primary report using their spectacles to see the hallucinations
more clearly. They may be in color or black and white.
Sometimes the hallucinations are in the house, but
Table 41.2 frequently are outdoors, i.e. workers digging up the
Types of hallucinations in 160 Parkinsons disease garden, checking the telephone lines or playing in
patients with drug-induced psychosis the street. One of the most interesting aspects of the
hallucinations is their consistency. A patient who
Type of hallucination No. of patients hallucinates children wearing funny clothes tends to
see the same children wearing the same clothing each
Visual 155 time, often sitting in the same chairs in the house
Auditory 76
doing the same things. This type of stereotyped
Olfactory 25
hallucination is therefore quite different from a dream.
The hallucinated figures typically ignore the patient
but may talk or gesture among themselves without
Table 41.3 sound. The hallucinations may get up and walk out
Differences in the hallucinations of schizophrenia and of the door, but the closing door makes no noise. Less
Parkinsons disease frequently, the hallucination may be inanimate, like a
statue or plant.
Schizophrenia Drug-induced Parkinsons disease Visual hallucinations are more frequent in the
evening but may occur during the day as well. Some
Auditory Visual patients only see hallucinations in the light. The hal-
Poor insight Preserved insight
lucinations may be Lilliputian (i.e. small people
Demeaning/nasty Benign
Present for long periods Usually under 5 minutes
living in a houseplant) or of normal size. Giant peo-
Ego-syntonic Unrelated to emotional state ple are generally not part of the phenomenology,
Interact with patient Ignore patient although enlarged animals, like large rats or cats,
may be.
222 K. L. CHOU AND J. H. FRIEDMAN
Auditory hallucinations, on the other hand, are et al., 2000), are more likely to induce visual halluci-
usually quite different from the visual. Auditory nations than levodopa. Drug doses are also important
hallucinations occur primarily in people who already risk factors. The higher the dose, the higher the risk.
have visual hallucinations (Inzelberg et al., 1998). Yet the occurrence of hallucinations is only partly
In these cases, the visual hallucinations may talk to related to the serum or brain level of the drug. One
the patient and carry on interactive conversations. study infused high-dose levodopa intravenously into
When auditory hallucinations occur without visual hal- PD patients who suffered from visual hallucinations
lucinations, the sounds tend to be indistinct, such as at home (Goetz et al., 1998a). Despite receiving much
party noises coming from another room or voices talk- higher levels of drug than they did at home, none of
ing outside, but without individual words being heard. them hallucinated in the hospital research setting, indi-
The hallucinations almost always occur when the cating that there is a combined effect of the drug and
patient is otherwise unstimulated. The patient is the patients mental state at the time. It is believed,
usually alone, either reading or watching television, but without supportive data, that polypharmacy also
when he/she notices the hallucinated people watching increases risk. Some experts believe that there is more
him/her. risk from low levels of multiple psychoactive drugs
Major risk factors for visual hallucinations include than higher levels of a single or few numbers of other
impaired vision, disease duration, dementia, depres- drugs.
sion, sleep disorders and recent medication changes Tactile, olfactory and gustatory hallucinations tend
(Nausieda et al., 1982; Comella et al., 1993; Pappert to be considerably less common in PD patients with
et al., 1999; Arnulf et al., 2000; Barnes and David, drug-induced psychosis, but occur more than rarely
2001), but many patients have no risk factors. Most (Friedman et al., 2002). One report described tactile
hallucinating patients have no premorbid psychiatric hallucinations as affecting about 7% of hallucinators
history. A common misconception is that the halluci- (Goetz et al., 1998b). As with auditory hallucinations,
nations only arise when medications have been these non-visual hallucinations occur almost exclu-
increased. Although this is frequently true, it is not sively in people who already suffer from visual ones
always the case. Patients commonly develop hallucina- (Fenelon et al., 2002; Tousi and Frankel, 2004). The
tions on doses of drugs that have not been changed in tactile hallucinations tend to be animals, worms, bugs
several years. The association of diminished vision and other generally unpleasant things that are also seen
with visual hallucinations brings to mind Bonnets (Fenelon et al., 2002).
syndrome, in which elderly people with severe visual The recognition of hallucinations is usually quite
dysfunction but normal cognition develop visual hallu- simple. Nevertheless, when the patient is too demented
cinations. Since PD patients may have impaired vision to produce a reliable history or when the hallucinations
that is unrelated to their PD, they are subject to Bon- take place around sleep, it can be challenging. It is
nets hallucinations (Matsui et al., 2004). However, unknown whether PD patients experience an increase
the statistical association between visual dysfunction in hypnagogic or hypnopompic hallucinations. A var-
and hallucinations is loose, with hallucinators iant of these conditions, the rapid-eye movement
performing only slightly worse on eyechart testing (REM) intrusion, occurs when a patient may experi-
(mean visual acuity 20/44.6) than non-hallucinators ence a dream intruding into consciousness while
(20/32.8) (Holroyd et al., 2001). Dementia has been sleepy but not actually asleep. It has become clear in
clearly documented as a risk factor, which brings up recent years that patients frequently do not recognize
the possible alternative diagnosis of DLB. A promi- their sleepiness and may suffer from sleep attacks
nent expert has opined that most cases of drug-induced (Olanow et al., 2000), suggesting that a patient suffer-
hallucinations represent the premature unmasking of ing a REM intrusion may not recognize it as a dream,
DLB by use of dopaminergic drugs (McKeith, perso- but rather remember it later as a hallucination. How-
nal communication), but no data exist to support this ever, the most likely confusion arises in distinguishing
claim. nocturnal hallucinations from vivid dreams and REM
It is interesting that all of the PD medications sleep behavior disorder. In the former the patient
(anticholinergics, dopaminergics and amantadine) experiences realistic dreams whereas in the latter the
have been found to induce very similar hallucinations dream is acted out, typically of fighting or running
(Goetz et al., 1982b) despite their different mechan- away from a threat.
isms of action. Yet certain drugs are more likely than
others to induce these problems. For example, the 41.2.1.4. Treatment
dopaminomimetics, such as pramipexole, ropinirole The management of the psychotic PD patient is similar
and pergolide (Parkinson Study Group, 2000; Rascol in delirious and non-delirious patients. Most importantly,
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 223
the possibility of triggering factors should be considered. agonists in previously untreated PD patients have
The most likely triggers are infection or drug toxicity. demonstrated that the agonists are more likely than
Although infections generally produce fever and other levodopa to induce psychotic symptoms (Parkinson
systemic symptoms, this is not always the case. There- Study Group, 2000; Rascol et al., 2000). We would
fore, urinary tract infections and pneumonias should reduce COMT inhibitors and levodopa last. If psychosis
always be considered as possible underlying explanations persists despite the fact that anti-PD medications have
for the onset of psychosis. PD drug overdoses may been reduced to their lowest level consistent with toler-
produce psychotic symptoms with or without delirium. able motor function, then an antipsychotic medication
Of all the PD medications, only amantadine is cleared should be started.
by the kidney, so that a degree of renal failure which Other approaches to reducing the psychosis have
would produce no encephalopathic features in a typical included drug holidays (Friedman, 1991) and electro-
PD patient may produce a toxic delirium in one taking convulsive therapy (ECT) (Hurwitz et al., 1988; Factor
amantadine. Other common drugs taken by PD patients et al., 1995). The data on drug holidays are scanty
that may cause or exacerbate psychosis include pain or and were accumulated primarily as a treatment of
sleeping medications. However, digitalis toxicity as well clinical fluctuations, not psychosis or delirium (Goetz
as other drug toxicities may be contributory. Occasionally et al., 1982a). These data suggest that it is not helpful
other metabolic explanations are found, such as isolated and often harmful. The use of ECT is supported by
renal or hepatic impairment or hypoxia. isolated case reports and appears to work even in cases
Structural brain lesions are virtually never explana- not diagnosed with psychotic depressions. We consider
tions. Although a patient may have fallen and sustained ECT a treatment of last resort, although psychiatrists
a subdural hematoma or intraparenchymal hemorrhage, with an interest in ECT think this is a major and biased
these are exceedingly uncommon and, when found, are oversight.
usually the result, not the cause, of the psychiatric The atypical antipsychotic drugs (AA) represent the
decline. newest generation of antipsychotic drugs used primar-
When possible, the patient is best managed at ily in the treatment of schizophrenia but also the other
home, in a secure, comforting, familiar environment. psychotic disorders (primarily schizoaffective, bipolar
It is common for PD patients who had no psychotic and psychotic depressive disorders). Clozapine was
or delirious behavior at home to decompensate simply the first such drug, followed shortly thereafter by ris-
upon moving into the hospital environment. We peridone, olanzapine, quetiapine, ziprasidone and most
strongly recommend that, whenever possible, the neu- recently aripiprazole. Unfortunately there is no con-
rologist develop a working relationship with a limited sensus definition of atypicality. Although several
number of psychiatric inpatient units, so that their different suggestions have been entertained, the most
PD patients can be referred to these units. This allows commonly accepted definition is relative freedom
the clinical staff in such units to gain more experience from extrapyramidal side-effects at the usual antipsy-
with management of psychosis in this population, chotic doses. What relative means however has never
which ultimately results in better care for the patient. been addressed and for patients with PD or DLB, who
The approach to treatment in patients with PD is dif- are the most sensitive populations to the extrapyrami-
ferent than in other conditions where the drug causing dal effects of these drugs, the issue is of great
the side-effect is discontinued. First of all, all importance. Data now exist on the effects of each of
antiparkinsonian medications can produce mental dis- the AAs, with the exception of ziprasidone. Although
turbances, making it difficult to know which drug is ziprasidone is not the most recently released AA, it
responsible or contributing to the psychosis. Secondly, alone carries a warning about a prolonged QT interval
reductions in these PD medications may cause intoler- on the electrocardiogram, making it an unpopular drug
able motor decline. We therefore recommend the to try on the elderly frail. There are double-blind pla-
following approach. First, we suggest discontinuing cebo-controlled data on clozapine and olanzapine in
medications used to treat other conditions, such as pain the treatment of drug-induced psychosis in PD
and insomnia, before reducing the anti-PD medications. (Parkinson Study Group, 1999; Breier et al., 2002;
We would then discontinue, in the following order, Pollak et al., 2004). Prior to the first randomized trial
anticholinergics, selegiline and amantadine, because of clozapine there was a wealth of open-label data
these medications have such mild antiparkinsonian indicating that the drug, at very low doses in compar-
effects that they can usually be discontinued without ison to the doses used in schizophrenia, was extremely
difficulty. If the patient remains psychotic, the next step effective in treating the psychosis without worsening
is to reduce or stop the dopamine agonists, since studies of motor function (Friedman and Factor, 2000).
comparing the benefits of levodopa to dopamine In fact, several reports have demonstrated that clozapine
224 K. L. CHOU AND J. H. FRIEDMAN
has antitremor efficacy that rivals that of levodopa increasingly less likely thereafter but, in the USA,
and benztropine (Friedman and Lannon, 1990; Jansen, biweekly monitoring is required as long as the patient
1994; Friedman et al., 1997). In schizophrenia, takes the clozapine. The Food and Drug Administra-
clozapine was started at 25 mg/day and increased by tion, in order to secure the blood count monitoring,
25 mg each day on a t.i.d. regimen to a dose total of has required the blood count result before dispensing
300900 mg/day (Kane et al., 1988). For schizophrenics the clozapine. Only enough pills are dispensed to last
with fixed delusions, that is, delusions that had until the next blood draw, thus guaranteeing compli-
been present and unchanged for several months, the ance while on the clozapine. In Europe the monitoring
improvement was generally only partial and usually is reduced to monthly after the first 6 months. There
took several weeks or months. In PD patients, fixed have been no deaths attributed to clozapine among
delusions sometimes resolved overnight even when PD patients in the USA.
the delusions had been present for many months. The monitoring requirement has made alternatives
In addition, the dose required was often as low as to clozapine very attractive. The second AA, risperi-
6.25 mg/day. Before this was recognized, a double- done, unfortunately failed to preserve motor function.
blind placebo-controlled trial was instituted using Risperidone has all the side-effects of a low-potency
the schizophrenia titration schedule, resulting in the neuroleptic. It causes dose-dependent parkinsonism,
only negative study ever reported on the efficacy of acute dystonic reactions, akathisia and increases in
clozapine in PD (Wolters et al., 1990). In 1999 the prolactin secretion. It also induces tardive dyskinesia,
only two randomized, double-blind placebo-controlled but at a considerably lower rate than the historical
trials of low-dose clozapine were published (Parkinson controls to which it has been compared (Jeste, 2004).
Study Group, 1999; French Clozapine Parkinson Study The data on risperidone in PD are actually quite
Group, 1999). Both designs were almost identical and mixed, with some reports describing the complete
the results were very close. Both showed that low-dose absence of motor side-effects (Meco et al., 1994; Allen
clozapine, with doses held between 6.25 and 50 mg/ et al., 1995; Workman et al., 1997) whereas others
day, resulted in clinically and statistically significant reported severe motor worsening in each patient who
benefit in all measures of psychosis employed and took the drug (Ford et al., 1994; McKeith et al.,
that motor function did not decline. In fact there 1995). No double-blinded trial has been published
was a non-statistically significant motor benefit of and it is unlikely that any will be performed, given
clozapine due to its antitremor effect. (The tremor the mixed results.
benefit was statistically significant.) In the American Olanzapine was the subject of four double-blinded
study, the most commonly used dose, chosen in a trials, three of which were placebo-controlled and
double-blinded fashion, was only 6.25 mg/day and one actively controlled, comparing the risperidone to
the mean dose required was 25 mg/day (Parkinson clozapine (Goetz et al., 2000; Breier et al., 2002; Ondo
Study Group, 1999). In the French study, the mean et al., 2002). The clozapine trial was halted prema-
dose was 35 mg/day (Pollak et al., 2004). Interest- turely due to intervention by the Safety Monitoring
ingly, the schizophrenic dosing is t.i.d. whereas the Board on behalf of the olanzapine-treated patients.
PD dosing is generally limited to a single dose, given The Safety Monitoring Board declared that olanzapine
at bedtime. These different responses again under- was unsafe for use in PD because of worsened motor
score the major differences between schizophrenic function, despite mean dosages of about 5 mg/day.
psychosis and drug-induced psychosis in PD. The The three other trials used placebo controls and all
clozapine data from the double-blinded trials support were completed. All three had similar results. Olanza-
the way clozapine was used prior to the two trials. pine was ineffective as an antipsychotic and worsened
Generally clozapine is started at 6.25 mg at bedtime motor function. This was in direct contrast to the first
and increased, based on response and tolerance, by prospective study, in which PD patients had dramatic
6.25 mg to 12.5 mg each night, as needed. We improvement in their psychosis without any decline
recommend increasing the dose daily until the patient in motor function (Wolters et al., 1996). Two of these
sleeps through the night, at which point the dose negative studies were identical and performed in the
should be increased more slowly, based on whether USA and Europe simultaneously (Breier et al., 2002).
the patient is oversedated in the morning. Despite Each of the studies involved 80 subjects, the most ever
the low dosing, weekly white blood cell counts must in any study of a treatment for a behavioral abnormal-
be monitored as the agranulocytosis side-effect is not ity in PD. Thus 160 subjects were enrolled. Both stu-
dose-dependent and appears to be mildly more dies had very similar negative outcomes. Since
common in the elderly (Alvir et al., 1993), affecting olanzapine does not increase prolactin levels and does
between 1 and 2% within the first 3 months. It becomes not cause acute dystonic reactions, the sensitivity of
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 225
PD patients is surprising. It is also surprising that the reflecting a retrospective outcome analysis of 39 par-
drug did not improve the psychosis. The mean doses kinsonian, not necessarily PD, patients who had been
in these studies were about one-third to one-half of treated with clozapine for psychosis found that 15%
the doses used in primary psychoses, in contrast to had died by 5 years and 33% had been admitted to nur-
the relatively much smaller doses of clozapine and sing homes (Fernandez et al., 2004a). Few were off
risperidone, which were effective in drug-induced psy- antipsychotic treatment, although an Israeli group
chosis in PD. These discrepancies remain unexplained. (Klein et al., 2003) reported that 9 of 32 clozapine-
The third drug, quetiapine, has been subject only to treated patients no longer required the clozapine. In
a single double-blinded placebo-controlled trial (Ondo contrast, another report on only 6 patients who had
et al., 2005). This trial was powered to determine been on either quetiapine or clozapine for a mean
whether the drug interfered with motor function, not duration of 20 months had their antipsychotic slowly
to prove that it was an effective treatment. The study tapered and discontinued (Fernandez et al., 2004c).
confirmed the safety of the drug and did not worsen Five of the 6 suffered worsened psychosis and 3 of
motor function, but there was no statistically signifi- these had a more severe psychosis than at baseline,
cant improvement in psychosis. In addition, a double- suggesting the possibility of a rebound psychosis.
blinded comparison trial found no differences between Compared to the one historical report involving a
clozapine and quetiapine, but the number of subjects small number of patients (Goetz and Stebbins, 1995),
was small (Morgante et al., 2004). The open-label these data suggest a somber but marked improved
reports, involving over 400 patients, indicate that outcome with quetiapine and clozapine.
quetiapine, although somewhat less potent and less It is likely that most psychotic patients will require
effective than clozapine and without the benefit on their antipsychotic for life if the medications responsi-
tremor, is nevertheless an effective drug for treating ble are not dramatically altered. However, it is always
the psychosis. The mean dose of quetiapine for treat- appropriate to consider the possibility of tapering and
ing this condition is about 65 mg/day, in contrast to stopping an antipsychotic to determine if it is still
the 2535 mg/day of clozapine. Like clozapine, que- required. Since the recurrent psychosis can be treated
tiapine is sedating and may contribute to orthostatic very early, it is unlikely to become severe and hard
hypotension. to control, even if the concept of a rebound psychosis
The most recent AA is aripiprazole. This drug has turns out to be true.
partial dopamine agonist properties despite being an
effective antipsychotic. There was hope that aripipra-
zole may improve both the psychosis and the motor 41.2.2. Mood and anxiety fluctuations associated
dysfunction, but early reports suggest that this is not with motor fluctuations
the case (Fernandez et al., 2004b; Friedman et al.,
2005). At very low doses, even under 5 mg, aripipra- It is well known that some patients who exhibit motor
zole worsened motor function in some PD patients. fluctuations also experience associated mood fluctua-
Since the smallest commercially available dose is tions. In general, off periods are associated with
5 mg and since the lower doses were not effective in depressed mood and increased anxiety, whereas on
treating the psychosis, it is unlikely that this drug will periods are associated with normal or elevated moods
prove helpful. (Hardie et al., 1984; Cantello et al., 1986; Friedenberg
and Cummings, 1989). In addition, some patients
experience off states as relatively pure sensory or
41.2.1.5. Long-term outcome mood experiences and others report mood swings pre-
Little data exist on this topic. The first report ceding the changes in motor function. These observa-
described a small nursing-home cohort of PD patients tions imply that mood changes in fluctuating patients
with hallucinations, all of whom died within 2 years are not simply a reaction to the motor changes. It
(Goetz and Stebbins, 1995). This was in the era before may be that mood is directly affected by changing
treatment with AAs was initiated. After clozapine drug levels, with anxiety and depression having
found widespread use, a study of the long-term out- somewhat different temporal relationships to serum
come of patients who had participated in the American drug levels than motor responses (Maricle et al.,
double-blind trial of clozapine (all of whom partici- 1995b). In one study, two-thirds of patients with motor
pated in an open-label safety extension) found that fluctuations also suffered from mood fluctuations
25% of completers were dead, that 42% were in (Nissenbaum et al., 1987); however, these data were
nursing homes, 68% were demented and 69% were obtained by post hoc questioning of patients. Overall,
still psychotic (Factor et al., 2003). Another study, only a small number of PD patients have actually been
226 K. L. CHOU AND J. H. FRIEDMAN
studied during their motor fluctuations to determine Only one subject had positive correlations between
mood and anxiety changes (Menza et al., 1990; mood and motor function each day. The investigators
Maricle et al., 1995a, b). The results so far have been concluded that a consistent relationship between
difficult to interpret due to the small numbers of sub- anxiety and motor states is not the rule. Furthermore,
jects as well as different techniques for assessment the study failed to confirm a consistent temporal
and, perhaps most importantly, possibly different relationship between mood and motor states. Perhaps
patient populations. most surprising was this studys observation that mood
In a double-blind placebo-controlled trial of 2-hour was sometimes negatively correlated with motor
levodopa infusions in the off state in 8 PD patients function.
with clinical fluctuations, Maricle et al. (1995b) Another study reported that 22 out of 47 PD patients
demonstrated that mean depression and anxiety scores experienced non-motor symptoms associated with
improved in a dose-related manner to levodopa infu- their motor fluctuations whereas only 16 had purely
sions, although finger-tapping rate, the sole measure motor changes (Raudino, 2001). The non-motor
of motor function, did not differ much with the two symptoms reported in this study were autonomic symp-
different infusion rates. Six of the 8 subjects noted an toms, pain, mood changes and anxiety. Other reports
improvement in mood and anxiety with high-dose have described patients with mood or other non-motor
levodopa. Four had a response to low-dose infusion fluctuations during the day that are independent of
and 2 responded to placebo. Anxiety scores appeared motor changes (Nissenbaum et al., 1987).
to have a different time response to the infusions The fact that some patients become hypomanic in
than either the depression or the motor score, but the the on with dyskinesia state, whereas others are
numbers of subjects were too small for statistical euthymic, remains difficult to explain (Nissenbaum
significance. et al., 1987), but no more so than the wide phenomen-
Menza and colleagues (1990) reported that, in 10 ological spectrum of non-motor fluctuations that have
consecutive, non-demented, non-depressed PD patients been reported, which include pain, irritability,
with on, off and on with dyskinesia periods, mood cognitive slowing, racing thoughts, sweating, belching,
and anxiety fluctuated with the onoff status, but, constipation, respiratory dysfunction, akathisia,
interestingly, and in contrast to the report by Maricle restless legs, cough, hunger, limb edema, nausea and
et al. (1995b), worsened when the patients suffered temperature (Hillen and Sage, 1996; Quinn, 1998). It
from their dyskinesias. These authors described must be noted that mood and anxiety may fluctuate
improvement of mood and anxiety as the patient went in response to many of these very uncomfortable
from off to on, then worsening from on to on sensations, producing the usual conundrum of whether
with dyskinesias. This important observation suggests certain symptoms are causal, related or merely
that the mood change is not a function of dopamine coincidental.
brain release, since that hypothesis would predict Clearly, more research needs to be performed in
either stability of mood or further enhancement of order to understand better the phenomenology of mood
mood as dopamine responsiveness increased. It is and anxiety fluctuations in patients with PD. Although
commonly observed that PD patients do, in fact, some- the general impression is that in fluctuators mood and
times become hypomanic when they become dyski- motor function go hand in hand, some studies indicate
netic (Giovannoni et al., 2000); however that did not that this is frequently not the case. Simple explanations
occur in this report. The decline in mood and worsen- for mood and anxiety changes will probably never be
ing anxiety in this case may simply have been due to adequate.
the fact that dyskinesias were bothersome, which
would suggest that mood is a reflection of changes in 41.2.3. Repetitive or compulsive behaviors
motor responses rather than biochemical changes
in the brain or serum. Although there is a relationship between abnormal repe-
One study of 16 PD patients with motor fluctua- titive or obsessive-compulsive behaviors and PD, its
tions found that the correlation between motor state nature is far from clear (Evans et al., 2004; Kurlan,
and either mood or anxiety was not very strong over 2004; Voon, 2004). Even the terminology is somewhat
a 1-week period (Richard et al., 2001). In this study, confusing. Obsessive-compulsive disorder (OCD) is
7 patients experienced daily mood fluctuations and 7 described as intrusive cognitive events (obsessions),
patients reported daily anxiety fluctuations. However, which result in intentional repetitive behaviors (compul-
these were not necessarily the same patients. Some sions) designed to neutralize both the cognitive intrusions
patients had mood fluctuations without anxiety fluc- and associated anxiety (Voon, 2004). This is in contrast
tuations, whereas in others, the opposite was true. to the obsessive-compulsive spectrum disorders (OCSD),
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 227
which are intrusive events with associated repetitive ture as levodopa dependence or abuse, hedonistic
behaviors (Voon, 2004), the difference being that homeostatic dysregulation and, most commonly, the
cognitive intrusions are absent from OCSD. These dopamine dysregulation syndrome (Priebe, 1984; Nau-
two categories are thought to have different underlying sieda, 1985; Giovannoni et al., 2000; Lawrence et al.,
mechanisms, but overlapping phenomenologies, sow- 2003; Evans et al., 2004). The patients who develop this
ing confusion in our clinical understanding. Tics and syndrome tend to be male with a young onset of their
behaviors such as hair-pulling, nail-biting and foot- PD. Past drug abuse, heavy alcohol use or a history of
tapping fall into the OCSD category, whereas ritual mood disorders may be predisposing factors (Giovan-
behaviors and checking are considered OCD. In addi- noni et al., 2000; Lawrence et al., 2003). The rapid titra-
tion, OCD is classified as an anxiety disorder, which tion to high levels of levodopa use occurs relatively
the OCSD syndrome is not. There are also non-patho- early in the disease, often in excess of 2000 mg/day.
logical obsessive-compulsive personality traits that Consequently, these patients often have severe, violent
have sometimes been considered to be part of the dyskinesias and display aggressive and devious tenden-
PD character, such as inflexibility, neatness, punc- cies when physicians attempt to curb their levodopa
tuality, obeying rules, following commands and a use. The extra levodopa results in enhanced mood,
decrease in novelty-seeking behavior (Menza et al., vigor and productivity and patients often base their
1993). Studies of such character attributes have been increase in medications not on the control of motor
contradictory, leading to the conclusion that such symptoms, but on somatic cues (Evans et al., 2004).
associations are still highly speculative. Although Apomorphine, a short-acting dopamine agonist, may
the possibility of an obsessive-compulsive personal- exacerbate or even initiate the symptoms of this
ity being part of PD has been hypothesized for a disorder (Giovannoni et al., 2000).
long time, the occurrence of abnormal repetitive It may be debated whether dopamine dysregulation
behaviors in the setting of PD has been recognized syndrome represents a true addiction to levodopa, but
only recently (Friedman, 1994; Fernandez and Fried- many physicians acknowledge that patients become
man, 1999) and its relationship to the drug treatment psychologically dependent on levodopa. Such a defini-
of PD versus being intrinsic to the disease itself is tion requires a pattern of excessive and inappropriate
unclear (Evans et al., 2004; Kurlan, 2004; Voon, use that results in impairment of social functioning
2004; Friedman, 2005). and causes withdrawal when the drug is discontinued.
Although certain pathological non-psychotic In the PD population, it may be difficult to tell whether
behaviors have been attributed to levodopa, such as there is excessive use of levodopa, particularly
hypersexuality, other compulsive behaviors such as because the medication alleviates motor symptoms.
punding (see below), pathological gambling, com- Yet a case report of a PD patient who needed his
pulsive shopping and levodopa drug abuse (or the levodopa dose despite being quadriplegic from
dopamine dysregulation syndrome) also occur in GuillainBarre syndrome suggests that patients can
PD patients. Furthermore, the absence of studies develop a pattern of excessive and inappropriate use
looking at control populations makes interpretation (Merims et al., 2000). There is no doubt that patients
of these reports somewhat tenuous in arguing the with dopamine dysregulation syndrome display beha-
connection between either disease state or drug viors that are detrimental to normal social functioning
treatment. The main observations concerning these and, when limiting levodopa use, develop dysphoria
behaviors are that they developed while on dopami- and other symptoms consistent with withdrawal.
nergic therapy and thus likely result from alterations Additionally, central dopaminergic pathways, specifi-
of either dopamine or serotonin. However, the small cally the mesolimbic dopaminergic projections to the
number of patients involved and the variability of nucleus accumbens, have been implicated in models
responses to interventions make it quite difficult to of drug abuse and dependence (Koob and Le Moal,
infer much from these reports. 1997; Sanchez-Ramos, 2002), so it is no surprise that
patients have the potential to develop dependence on
levodopa.
41.2.3.1. Dopamine dysregulation syndrome Other abnormal behaviors can be seen with this syn-
It has become increasingly apparent that a small num- drome, including mania, psychosis, aggression, drug
ber of PD patients, mostly treated with levodopa, hoarding, punding (see below), hypersexuality, compul-
begin to take increasing amounts of their medication, sive shopping and pathological gambling. Management
despite the fact that their motor symptoms are well of this disorder can be difficult, but it is essential to
controlled with lower doses. This compulsive drug decrease and ration dopaminergic medications. Atypical
use pattern has been variably described in the litera- antipsychotics such as quetiapine or clozapine can
228 K. L. CHOU AND J. H. FRIEDMAN
be used to treat the behavioral manifestations (Giovan- years of PD and was unrelated to medications or his
noni et al., 2000). clinical condition. Another patient hummed, but only
as an off phenomenon (Friedman, 2005).
41.2.3.2. Punding
Punding refers to a repetitive behavior first described 41.2.3.3. Pathological gambling
in amphetamine addicts who developed an endless Although pathological gambling can be seen as part of
fascination with taking objects such as flashlights and the dopamine dysregulation syndrome or mania, it has
small electrical gadgets apart, then trying, usually been reported in patients with PD without other symp-
unsuccessfully, to put them back together (Rylander, toms of the above disorders. Pathological gambling is
1972). This type of repetitive behavior was calming characterized by a failure to resist the impulse to gam-
to the patient and considered pleasurable. Several PD ble despite severe consequences, whether personal,
patients have been described with similar behavior, financial or vocational. It is considered an impulse
all developing while under dopaminergic drug treat- control disorder in the Diagnostic and Statistical
ment, although in 2 patients, the punding appeared Manual of Mental Disorders, 4th edition (DSM-IV)
after the initiation of quetiapine and decreased or (American Psychiatric Association, 1994). This phe-
resolved when the quetiapine dose was reduced (Miwa nomenon was first reported in 12 patients with PD, 9
et al., 2004). The different punding behaviors that have of whom began gambling after initiating levodopa
been described include cataloging a small, inexpensive therapy (Molina et al., 2000). One patient, who
jewelry collection, repetitively adding tables of sums, gambled prior to levodopa therapy, reported that his
endlessly rearranging a collection of buttons, taking gambling behavior worsened after starting treatment.
apart and putting together flashlights, examining The authors of this paper noted that their patients were
television and other electrical cords and repeatedly relatively young and 5 of them had alcohol use or
reading the labels on cans in a supermarket (Friedman, dependence. Since this initial report, pathologic gam-
1994; Fernandez and Friedman, 1999; Evans et al., bling has been observed in other patients on levodopa
2004; Miwa et al., 2004). These behaviors are often therapy (Gschwandtner et al., 2001; Avanzi et al.,
connected to the patients occupation and the patients 2004) and in patients on dopamine agonist therapy
themselves realize that the time spent on their punding alone (Seedat et al., 2000; Driver-Dunckley et al.,
tasks is excessive (Friedman, 1994; Evans et al., 2003). The patients on dopamine agonists alone were
2004). In contrast to the punding of amphetamine on pramipexole or pergolide and, for many, their gam-
users, PD patients often feel irritable or anxious when bling behavior resolved when switched to ropinirole.
distracted from or stopping their behaviors, although Others noted improvement with reductions in total
most report no compulsion to pund (Fernandez and dopaminergic medication, the addition of antipsycho-
Friedman, 1999; Evans et al., 2004). tic therapy, antidepressant therapy, family control or
Although punding is hypothesized to be due to DBS (Molina et al., 2000; Seedat et al., 2000; Avanzi
excessive dopaminergic stimulation (Ridley and Baker, et al., 2004). Most did not respond to standard thera-
1982) and some patients have improved with decreased pies for pathological gambling, such as behavioral or
doses of dopaminergic medications (Fernandez and cognitive therapy (Molina et al., 2000; Avanzi et al.,
Friedman, 1999), one report describes similar stereo- 2004). Although these reports suggest that pathologic
typed behaviors in PD patients that were unresponsive gambling is linked to dopaminergic medications in
to reductions in antiparkinsonian therapy (Kurlan, PD, it should be noted that no report has specifically
2004). As mentioned before, 2 other PD patients have assessed the prevalence of pathologic gambling in the
been reported where punding began after the adminis- general population and compared it to the prevalence
tration of quetiapine (Miwa et al., 2004). These reports in patients with PD.
suggest that a simple hyperdopaminergic state is not
enough to account for these stereotyped behaviors.
41.3. Mental changes due to surgical
Hypomanic symptoms have occurred in conjunction
treatments for Parkinsons disease
with repetitive behaviors in some cases, implying that
the underlying pathophysiology is complex and may 41.3.1. Overview of surgical treatments for
involve a combination of dopaminergic, serotonergic Parkinsons disease
and other neurotransmitter systems (Kurlan, 2004).
Other repetitive behaviors have been reported, The surgical approach to PD has evolved dramatically
including 1 patient who made annoying sucking sounds since the pre-levodopa era. Early attempts at treating
which bothered his friends. This developed after many PD surgically were highly variable, partly due to an
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 229
incomplete understanding of basal ganglia pathophy- candidates for psychiatric and cognitive disorders, we
siology, but mostly because of poor techniques for tar- still have limited knowledge of who is subject to these
geting brain structures. Target localization improved adverse effects. Possible explanations include interrup-
dramatically with the advent of stereotactic techniques tion of fibers controlling mood and cognition, natural
(Spiegel et al., 1947), but unfortunately, surgical treat- progression of the neurobehavioral features of PD,
ments for PD were abandoned after the introduction of dopaminergic medication withdrawal or a combination
levodopa in the 1960s. It wasnt until the medical of the above. Hopefully, as the cognitive and psychia-
community realized that long-term levodopa therapy tric morbidities associated with ablation and DBS
severely restricted PD patients daily functioning due are studied more systematically, we will be able to
to the development of motor fluctuations and dyskine- understand the underlying etiologies better.
sias that there was a resurgence of interest in surgical
therapies for PD. Over the last two decades, major 41.3.2. Adverse psychiatric effects of surgical
progress has been made in the field of movement treatments for Parkinsons disease
disorders surgery, from the refinement of ablative
techniques to the development of new alternatives, Although cognitive side-effects have been reported
such as DBS (Benabid et al., 1991), and surgery is with thalamotomy in PD, this section will focus on
now an accepted form of therapy for advanced PD. adverse psychiatric and cognitive effects from the
The thalamus was originally the preferred target for most common surgical procedures currently performed
PD. However, because thalamic lesions improve only for advanced PD: pallidotomy, GPi DBS and STN
the tremor in PD and because bilateral lesions were DBS. Table 41.4 shows the common psychiatric
associated with a high incidence of speech problems changes seen with surgery. It is important to keep in
(Kelly and Gillingham, 1980), the thalamus has fallen mind that most surgical studies of PD patients have
out of favor as a target. The internal segment of the not used psychiatric scales to assess behavior. Thus,
globus pallidus (GPi) and the subthalamic nucleus most of the data on adverse psychiatric effects of PD
(STN) are much more commonly targeted now surgery consist of descriptive case reports or case ser-
because a lesion in either structure reduces all three ies. The cognitive consequences of PD surgery have
of the cardinal motor manifestations in PD, but been better studied, but are also limited because of
because no double-blind, randomized, head-to-head small sample sizes and short-term follow-up.
trials have been conducted between the two sites, it
is unclear which target is better. There are also 41.3.2.1. Psychosis
minimal data comparing the two different surgical Despite being the most common neuropsychiatric
techniques in use for PD surgery today: ablation and complication of pharmacologic therapy in PD, psycho-
DBS. Nevertheless, most surgical centers are now sis is a relatively rare occurrence after ablation
performing DBS. This is largely because DBS offers or DBS surgery and, in fact, has only been reported
several advantages over ablative therapy, including: after STN DBS. In one study, 4 out of 77 PD patients
(1) reversibility, meaning there is no permanent
destructive lesion made in the brain and likely will
not hamper future PD therapies; (2) DBS can be Table 41.4
performed bilaterally with relatively few adverse
effects; and (3) stimulation parameters can adjusted Adverse psychiatric effects reported with surgical
treatments in Parkinsons disease
to decrease adverse effects and increase clinical effect
as the disease progresses.
Pallidotomy
Despite the controversy regarding which surgical Depression
target or technique is clinically superior in PD, it is Hypomania
abundantly clear that both ablation and stimulation of Globus pallidus internal segment stimulation
either target can provide significant relief of many Mania
parkinsonian motor symptoms and reduce the severity Subthalamic nucleus stimulation
of long-term motor complications from levodopa ther- Apathy
apy. Unfortunately, as neurologists and neurosurgeons Anxiety
have slowly accumulated more experience with these Depression
procedures, especially DBS, it has been recognized Hypomania
Mania
that patients sometimes experience declines in mood
Mirthful laughter
and cognition. Despite the fact that most movement
Psychosis
disorders surgical centers actively screen potential
230 K. L. CHOU AND J. H. FRIEDMAN
undergoing STN DBS surgery and evaluated up to 3 cally target the posterior ventral pallidum in PD, which
years postoperatively suffered from psychosis (Funkie- contains purely sensorimotor fibers, making it the
wiez et al., 2004). One of these patients reportedly had most effective target for ameliorating parkinsonian
a transient psychosis that occurred 6 weeks after symptoms (Bakay et al., 1997). Lesions restricted to
electrode implantation. The other 3 had permanent this area do not result in mood disorders, whereas
psychoses, although 2 were demented. In another lesions outside this area may affect the non-motor
study examining 48 PD patients following bilateral basal ganglia circuits responsible for cognitive and
STN DBS surgery, 2 had psychotic syndromes that mood changes (Lombardi et al., 2000). Two patients
resolved without any specific therapy (Herzog et al., were recently reported to have transient hypomania
2003b). The Grenoble group (Krack et al., 2003) subsequent to pallidotomy (Okun et al., 2003). It was
reported 1 case of transient psychosis in a 5-year fol- discovered that both of these patients had lesions
low-up study of 49 STN DBS patients. Unfortunately involving the anteromedial (non-motor) portion of the
none of these studies reported the specifics of the GPi, which was believed to be responsible for their
psychotic episodes, so it is unknown whether they mood changes. Mania has also been reported as a
had hallucinations, delusions or both. Furthermore, it result of pallidal stimulation (Miyawaki et al., 2000).
is unclear if these patients had psychosis prior to In this case report, the patient had recurrent mania
surgery, a factor that could predispose them to develop which always occurred temporally after his stimulators
psychosis after implantation. In another study of 24 were turned on. When the stimulators were off, he
DBS patients (Houeto et al., 2002), 1 patient with never went into a manic state. His mood eventually
drug-induced psychosis prior to surgery developed a stabilized in the on stimulation state with the reduc-
florid psychosis with mystic delusions a few weeks tion of dopaminergic therapy, leading the authors to
postoperatively. However, another patient with drug- suspect that stimulation altered his sensitivity level
induced psychosis prior to surgery did not manifest to the mood-altering effects of levodopa.
psychotic symptoms afterwards. In contrast to pallidotomy or pallidal stimulation,
One patient has been reported as having developed mania has frequently been observed with STN stimula-
delusions subsequent to STN DBS surgery (Herzog tion. As mentioned above, 1 patient experienced mania
et al., 2003a). This patient had no psychiatric history. with delusions after STN DBS surgery (Herzog et al.,
Her delusions occurred concurrently with manic 2003a). Another study reported manic symptoms
symptoms, including euphoria, loosened associations, occurring in 3 out of 15 STN stimulation patients
flight of ideas and hyperactivity. The mania and the (Kulisevsky et al., 2002), including elation, inflated
delusions eventually needed to be controlled with a self-esteem, overactivity and sexual indiscretion. None
combination of carbamazepine and clozapine. had previous psychiatric episodes and in all 3 patients,
A few patients have experienced isolated visual hal- manic symptoms began shortly after stimulation was
lucinations after DBS surgery of the STN. One patient, turned on. Interestingly enough, stimulation was
off all dopaminergic medications after bilateral STN switched from the lowest contacts to higher ones in
DBS surgery, experienced formed visual hallucina- all 3 patients a week after surgery because of better
tions only when the stimulators were turned on motor responses and the symptoms resolved. In 1
(Diederich et al., 2000). Because the visual hallucina- patient, restimulating the lowest contact on the right
tions responded to clozapine therapy, the authors side caused the mania to recur. Because the lowest
concluded that electrical stimulation physiologically contacts were located caudally to the substantia nigra
mimicked drug-induced visual hallucinations and in all 3 patients, the authors speculated that stimulation
suggested that there may be a connection between may have affected limbic projections from the mid-
the STN and limbic pathways. However, not all cases brain. Romito and colleagues (2002), among their first
of visual hallucinations after surgery are induced by 30 STN DBS patients, described 2 who met DSM-IV
stimulation. Some are related to the development of diagnostic criteria for mania postoperatively. One
dementia (Krack et al., 2003; Rodriguez-Oroz et al., developed manic symptoms prior to the initial pro-
2004), whereas in others, the etiology is less clear gramming session, whereas the other developed
(Varma et al., 2003). symptoms a few hours after the stimulators were
turned on. The mania persisted even after discontinua-
41.3.2.2. Mania tion of all antiparkinsonian medications. Similar to the
Although manic symptoms have been known to occur patients reported by Kulisevsky et al. (2002), the lower
with pallidal lesions (Turecki et al., 1993), mania or contacts were stimulated in both of these patients.
hypomania hardly ever occurs after pallidotomy. However, in contrast, Romito and colleagues (2002)
This is largely because most neurosurgeons specifi- did not change the contacts in their patients and the
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 231
mania resolved spontaneously over the ensuing months It may be that targeting the STN itself predisposes
without therapy. patients to the development of postoperative depres-
In addition to mania, hypomania has also been sion. It is of particular interest that depressive sympto-
commonly described after placement of bilateral STN matology has been reported more often after bilateral
electrodes (Houeto et al., 2002; Herzog et al., 2003b; STN DBS surgery than after pallidotomy or GPi sti-
Krack et al., 2003; Funkiewiez et al., 2004). Nearly mulation. In fact, most studies report that severity of
all of the episodes reported occurred within the first depression is either unchanged (Vingerhoets et al.,
3 months of surgery and resolved spontaneously. 1999; Baron et al., 2000; Favre et al., 2000) or mildly
Moreover, 2 cases of mirthful laughter due to STN improved (Troster et al., 1997; Masterman et al., 1998;
stimulation have been described (Krack et al., 2001). Martinez-Martin et al., 2000; Straits-Troster et al.,
The laughter seen in these patients was acutely 2000) after pallidotomy or GPi stimulation, whereas
induced by stimulation increases in voltage or pulse only a few have reported otherwise. One group noted
width. It remains unclear, however, whether the STN that 12% of their pallidotomy patients developed
itself is involved in regulating affect and behavior or depression after surgery (Bezerra et al., 1999).
whether diffusion of the stimulation to limbic-related Interestingly, all of them had undergone left-sided
structures adjacent to the STN is responsible for these unilateral ablation. Another group conducted a rando-
changes in mood. mized trial of bilateral pallidotomy versus a combina-
tion of unilateral pallidotomy plus contralateral GPi
41.3.2.3. Depression stimulation, but had to discontinue the study early
Although the strong association between depression when the 3 patients undergoing bilateral pallidotomy
and PD had been recognized for some time, investiga- displayed a significant deterioration on the Hamilton
tors did not pay close attention to depression following depression and apathy scores (Merello et al., 2001).
surgical interventions in PD until 1999, when a case A third study reported that 2 out of 4 bilateral pallidot-
report of stimulation-induced depression following omy patients experienced postoperative depression
bilateral STN electrode implantation was published (Ghika et al., 1999).
(Bejjani et al., 1999). In this patient, electrical The reasons for a greater incidence of depression
stimulation through the lowest contact (0) of the left after STN DBS remain unclear. Several possibilities
STN electrode elicited symptoms of depression that exist, however. More attention has recently been
disappeared immediately when stimulation was turned focused on non-motor symptoms in PD, such as
off. Although this particular contact was estimated to depression and dementia, than in the past. It may be
lie in the left substantia nigra rather than the STN, this that this spotlight on cognitive and behavioral issues
observation shattered the concept that depression in PD has caused investigators to recognize psychiatric
was mainly a reaction to the parkinsonian symptoms side-effects more readily. The discrepancy between a
experienced by the patient and suggested that depres- patients expectations from surgery and the actual
sion could be hard-wired in the brain. A number of result may be a contributing factor. Furthermore, the
reports from different surgical centers have since STN is a small target, making it more likely for the
reported the development of depression following electrode to be misplaced. It is also probable that elec-
STN DBS surgery in patients with PD (Krack et al., trical stimulation in this area may activate limbic
1997; Volkmann et al., 2001; Berney et al., 2002; fibers traveling from the STN through the striatum to
Doshi et al., 2002; Thobois et al., 2002; Herzog et al., the prefrontal cortex, pathways that have been impli-
2003b; Kleiner-Fisman et al., 2003; Funkiewiez et al., cated in the neurobiology of depression (Groenewegen
2004), with a frequency ranging from 10% to 30%, and Berendse, 1990; Drevets, 2003). Finally, because
but none has been so clearly induced by stimulation. STN stimulation allows patients to reduce their medi-
Although PD patients undergoing STN DBS seem to cations, generally by approximately 50% (Thobois
show mild improvement overall when evaluated with et al., 2002; Kleiner-Fisman et al., 2003; Krack et al.,
depression rating scales at baseline and postoperatively 2003), patients may experience a depressed mood
(Ardouin et al., 1999; Daniele et al., 2003; Troster because of the withdrawal of levodopa or other PD
et al., 2003), certain individuals seem to be susceptible medication.
to severe mood changes after surgery, with some The existing literature suggests that those who
even attempting suicide (Krack et al., 1997; Berney develop a depressed mood postoperatively often have
et al., 2002; Doshi et al., 2002; Houeto et al., 2002; a prior psychiatric history. Of the 5 patients with
Kleiner-Fisman et al., 2003). It is still difficult at this depression after STN surgery in the group reported by
time, however, to predict which patients are at the Houeto and colleagues (2002), 4 had a previous
greatest risk. history of depression, whereas 2 of 6 patients with
232 K. L. CHOU AND J. H. FRIEDMAN
postoperative depression in a separate series had 41.3.3. Adverse cognitive effects of surgical
depression preoperatively (Berney et al., 2002). How- treatments for Parkinsons disease
ever, 15 other patients in these two studies had
depressive symptomatology prior to DBS, yet did not As with psychiatric symptoms, STN DBS procedures
experience a depressed mood afterwards. Therefore, are generally associated with more declines in
although a past psychiatric history may predispose cognitive function than with procedures involving
patients to postsurgical depression, it is clearly not the GPi, although long-term postoperative assessments in
only factor. Fortunately, it seems that most postopera- these patients remain limited. Although most patients
tive depressive episodes are transient. If treatment is remain cognitively stable after surgery overall, there
necessary, an increase in dopaminergic medication or are reports of PD patients with dementia or borderline
the addition of an antidepressant, such as a serotonin cognitive function who worsen irreversibly postopera-
uptake inhibitor, is usually sufficient (Doshi et al., tively (Limousin et al., 1998; Hariz et al., 2000).
2002; Thobois et al., 2002; Krack et al., 2003; Furthermore, patients of advanced age and increased
Funkiewiez et al., 2004). PD severity may have worse cognitive outcomes after
surgery for PD (Vingerhoets et al., 1999; Baron et al.,
41.3.2.4. Apathy 2000; Kubu et al., 2000; Saint-Cyr et al., 2000). As a
Apathy as a sequela of surgical procedures in PD has result, it is essential to screen potential surgical candi-
been reported specifically with bilateral STN DBS dates for the presence of a dementing illness with
(Trepanier et al., 2000; Daniele et al., 2003; Krack preoperative neuropsychological testing. Such testing
et al., 2003; Funkiewiez et al., 2004). These patients ensures that only the most optimal candidates are
are generally not depressed or demented and typically selected and also provides a baseline for comparison
sit in a chair for most of the day (Funkiewiez et al., of postsurgical changes in cognition.
2004). The pathophysiology is unknown. Although
an increase in dopaminergic agents or antidepressant 41.3.3.1. Neuropsychological assessment
therapy may help apathy in PD, most of the patients Preoperative neuropsychological testing involves the
with post-DBS apathy did not respond to dopaminer- assessment of multiple cognitive domains. These are
gic therapy. Nevertheless, signs of apathy should be generally grouped into five categories: (1) general cog-
sought in patients following STN stimulation surgery nitive or intellectual ability; (2) language; (3) learning
and treatment should be attempted. and memory; (4) executive function; and (5) visuospa-
tial function. Common neuropsychological tests used
41.3.2.5. Anxiety to evaluate these different domains are listed in
Anxiety disorders are commonly seen in patients with Table 41.5.
PD. However, they do not seem to develop after surgi- Loss of general intellectual ability is one of the
cal procedures for PD. In fact, symptoms of anxiety hallmarks of a dementing illness (Saint-Cyr and
generally improve in PD patients after pallidotomy, Trepanier, 2000). Because the main purpose of neu-
pallidal stimulation and STN stimulation (Fields et al., ropsychological testing prior to surgery is to rule out
1999; Straits-Troster et al., 2000; Higginson et al., dementia, evaluation of intellectual ability is essential.
2001; Martinez-Martin et al., 2002; Daniele et al., The common neuropsychological tests that are utilized
2003). Only one study reported a decompensation of (Table 41.5) can estimate current intellectual ability
anxiety symptoms after surgery in PD patients (Houeto and premorbid intellectual level and provide a global
et al., 2002). In this study, 24 PD patients undergoing cognitive screen. PD patients with evidence of border-
STN DBS were evaluated for behavioral disorders line or impaired cognitive function based on these tests
before and after surgery. Seventeen (71%) were diag- are not considered good surgical candidates. Evalua-
nosed with generalized anxiety and 4 (17%) were diag- tion of the domain of language involves examining
nosed with agoraphobia before the DBS procedure. the ability to comprehend and produce verbal language
Unfortunately, in all 17 of the anxiety patients and 2 easily and fluently. Future long-term follow-up studies
of the agoraphobic patients, symptoms worsened after assessing language ability after surgery for PD will be
surgery. One patient without psychiatric symptoms prior important because decreased verbal fluency has been
to surgery was discovered to have anxiety postopera- the most consistent change reported postoperatively
tively, but the reasons for this decompensation remain (see section 41.3.3.3 below). Memory disturbance
unclear. Given such sparse data, it is obvious that more can be another sign of dementia. In PD, free recall is
studies need to be conducted in order to determine how typically impaired, whereas cued recall is preserved
anxiety symptoms respond to surgery in PD. (Pillon et al., 1994). This is in contrast to other
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 233
Table 41.5 cognitive decline (Laitinen, 2000) and among the early
pioneers of pallidotomy, only Leksells group reported
Common neuropsychological tests used in the evaluation
general postoperative cognitive decline (Svennilson
of Parkinsons disease patients before and after surgery
et al., 1960). This decline was a permanent deficit
General cognitive/intellectual ability that occurred in 4 of 81 patients. More recently, Baron
 Wechsler Adult Intelligence Scale-III (WAIS-III) and colleagues reported that, of 9 patients undergoing
 American New Adult Reading Test (ANART) pallidotomy, 3 showed significant declines in DRS
 Mattis Dementia Rating Scale (DRS) scores over 4 years (Baron et al., 2000). However,
 Mini-Mental State Examination (MMSE) these 3 patients were older at the time of surgery and
Language it is unclear if the cognitive decline was due to the pal-
 Subsets of the WAIS-III lidotomy or because their older age put them at greater
 Boston Naming Test risk of developing dementia. In contrast, a study of
 Category Fluency (Animals trial)
43 patients undergoing unilateral pallidotomy showed
 Controlled Oral Word Association Test
no significant changes in the DRS scores at 3 and
Learning and memory
 Wechsler Memory Scale-III (WMS-III) 12 months, respectively (Obwegeser et al., 2000).
 Hopkins Verbal Learning Test In general, DRS scales show no significant dete-
 California Verbal Learning Test rioration with either GPi (Troster et al., 1997; Ardouin
Executive function et al., 1999; Fields et al., 1999) or STN DBS (Ardouin
 Digit span and digit span backwards et al., 1999; Pillon et al., 2000; Funkiewiez et al.,
 Wisconsin Card Sorting Test (WCST) 2004). However, one small study found a small but
 Trail Making Test statistically significant improvement (6.7%) in the
 Stroop Color-Word Test MMSE after 12 months of STN stimulation (Daniele
Visuospatial function et al., 2003).
 Construction subscore of the DRS
 Hooper Visual Organization Test 41.3.3.3. Language
 Benton Judgment of Line Orientation
No studies have reported an improvement in language
function postoperatively. On the contrary, declines in
language function have been consistently reported after
dementing illnesses such as Alzheimers disease where
surgical procedures for PD. Specifically, reductions in
both free and cued recall is disrupted. Thus, a deficit in
verbal fluency have been observed after pallidotomy
cued recall in a PD patient prior to surgery may imply
(Uitti et al., 1997; Ghika et al., 1999; Kubu et al.,
a memory disorder more severe than what would
2000; Lacritz et al., 2000; Obwegeser et al., 2000;
be expected. The domain of executive function
Trepanier et al., 2000), pallidal stimulation (Troster
allows patients to formulate, plan and execute actions.
et al., 1997; Ghika et al., 1998) and STN stimulation
Problems in this domain generally suggest frontal lobe
(Ardouin et al., 1999; Pillon et al., 2000; Saint-Cyr
dysfunction (Lezak, 1995), a finding commonly seen
et al., 2000; Trepanier et al., 2000; Alegret et al.,
in PD and thus important to evaluate. The presence
2001; Gironell et al., 2003; Funkiewiez et al., 2004).
and extent of visuospatial deficits in PD remain deba-
Although there are studies that have reported no
table (Pillon et al., 2001). Because of this, visuospatial
change in verbal fluency after pallidotomy (Soukup
function has not been consistently assessed pre- and
et al., 1997; Turner et al., 2002; Gironell et al.,
postsurgery in PD patients.
2003), these are clearly in the minority. Even more
interesting is the fact that this decrease in fluency
41.3.3.2. Global cognitive and intellectual ability seems to occur with left-sided pallidotomy, but not
It is important to make sure that PD patients are not right pallidotomy (Uitti et al., 1997; Kubu et al.,
demented prior to surgery. Therefore, testing for intel- 2000; Lacritz et al., 2000; Obwegeser et al., 2000;
lectual function and global cognitive ability is always Trepanier et al., 2000). These differential findings are
performed preoperatively. However, most centers do not found with unilateral GPi stimulation (Troster
not routinely test intellectual function after surgery. et al., 1997; Vingerhoets et al., 1999), although the
The Mattis Dementia Rating Scale (DRS) has emerged number of patients studied so far has been small.
as a popular test of global cognitive ability, but only a Ghika and colleagues (1998) reported that 2 of 6
few studies have examined changes in the DRS before patients undergoing bilateral GPi DBS had decreased
and after surgery. verbal fluency postoperatively, but most others have
Most of the early pallidotomy literature did not reported no significant change (Ardouin et al., 1999;
routinely employ neuropsychological tests to assess Fields et al., 1999; Trepanier et al., 2000).
234 K. L. CHOU AND J. H. FRIEDMAN
A recent review of neuropsychological conse- the 3- and 6-month postoperative visits (Trepanier
quences following STN DBS found that 69% of the et al., 2000).
studies reported significant declines in measures of
phonemic fluency, category fluency or both (Woods 41.3.3.5. Executive function
et al., 2002). These deficits continue to be present in Executive function is generally unchanged after
long-term studies of STN DBS that have followed pallidotomy and GPi stimulation (Troster et al., 1997;
patients anywhere from 12 to 36 months postopera- Ardouin et al., 1999; Burchiel et al., 1999; Fields
tively (Saint-Cyr et al., 2000; Daniele et al., 2003; et al., 1999; Ghika et al., 1999; Vingerhoets et al.,
Funkiewiez et al., 2004). It is unclear why these reduc- 1999; Trepanier et al., 2000; Gironell et al., 2003).
tions in verbal fluency tests occur. It does not seem to However, one group demonstrated greater persevera-
be related to the stimulation itself, given that, in one tion on the Wisconsin Card Sorting Test in PD patients
study, verbal fluency deficits were present at 3 and tested 3 months after a right pallidotomy (Lacritz
12 months after STN surgery, regardless of stimulation et al., 2000), whereas another found that performance
status (Pillon et al., 2000). declined after pallidotomy on the Tower of London
Test, a finding that suggests reduced planning ability
41.3.3.4. Learning and memory (Turner et al., 2002).
Changes in learning and memory have been variable Similarly, most groups have found no significant
following pallidotomy. Whereas many studies showed changes in attention or executive function after STN
either no change or an improvement on memory testing DBS surgery (Ardouin et al., 1999; Burchiel et al.,
postpallidotomy (Uitti et al., 1997; Lacritz et al., 2000; 1999; Pillon et al., 2000; Trepanier et al., 2000;
Obwegeser et al., 2000; Turner et al., 2002; Gironell Perozzo et al., 2001; Daniele et al., 2003; Gironell
et al., 2003), others have shown a decline in memory et al., 2003), with only a couple of studies showing
(Ghika et al., 1999; Trepanier et al., 2000). In one of improvement (Alegret et al., 2001; Daniele et al.,
these studies, free recall worsened in patients with a 2003). Although the surgery itself does not seem to
left-sided pallidotomy as opposed to patients who have much of an effect on this domain overall, some
underwent right-sided pallidotomy (Trepanier et al., recent studies have suggested that stimulation itself
2000), a pattern similar to that observed with verbal may have a positive effect on frontal lobe function.
fluency. Interestingly enough, another study observed One study reported the results of both STN and GPi
an improvement in memory in patients who had a stimulation on neuropsychological measures in the sti-
pallidotomy on the right side (Obwegeser et al., mulator on and off states and discovered that, when
2000). However, the data remain too limited to draw stimulation was on, both STN and GPi groups
any conclusions. improved on various measures of executive function,
Thus far pallidal DBS has not been shown to including the Trail Making Test (parts A and B) and
worsen performance on learning and memory testing. the interference task on the Stroop test (Jahanshahi
A few studies examining both unilateral (Troster et al., 2000). Pillon and colleagues (2000) reported
et al., 1997; Vingerhoets et al., 1999) and bilateral similar findings on the Stroop and Trail Making Tests
GPi stimulation (Ardouin et al., 1999; Burchiel et al., with stimulators turned on and off in a group of 48
1999) reported no change in learning or memory, STN DBS patients.
whereas a separate study showed that bilateral GPi
stimulation actually improved free recall, as assessed 41.3.3.6. Visuospatial function
by the California Verbal Learning Test in 6 patients Only a few studies have looked at measures of visuos-
(Fields et al., 1999). patial function. One pallidotomy study showed no
Most studies following STN electrode implantation significant changes in this domain, as assessed by the
have demonstrated no change on measures of learning Wechsler Adult Intelligence Scale-Revised Block
and memory (Ardouin et al., 1999; Burchiel et al., Design and the Benton Judgment of Line Orientation
1999; Moro et al., 1999; Alegret et al., 2001; Perozzo (Obwegeser et al., 2000). Another pallidotomy study
et al., 2001; Gironell et al., 2003). One study observed showed a transient decrease in visuoconstructional abil-
a decline in immediate and delayed recall on Reys ity, as assessed by copying the Rey-Osterrieth Complex
auditory verbal learning test 3 months postoperatively, Figure, although the effects were transient and had dis-
but this decline returned to baseline at both the 6- and appeared by 1 year (Trepanier et al., 1998). It was also
12-month follow-up visit (Daniele et al., 2003). In con- unclear whether the difficulty in copying the figure was
trast, another study found significant declines in long- due solely to a visuospatial deficit, as reduced planning
delay free recall and long-delay cued recall at both ability may also cause patients to have problems with
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 235
this test. Pallidal DBS overall seems to have no effect on
Ardouin C, Pillon B, Peiffer E et al. (1999). Bilateral
visuospatial function (Troster et al., 1997; Ardouin et al., subthalamic or pallidal stimulation for Parkinsons
1999; Vingerhoets et al., 1999), but one study reported disease affects neither memory nor executive functions:
that bilateral GPi DBS caused a decline in the construc- A consecutive series of 62 patients. Ann Neurol 46:
tion subscore on the DRS (Fields et al., 1999). Likewise, 217223.
STN stimulation appears not to affect significantly cog- Arnulf I, Bonnet AM, Damier P et al. (2000). Hallucinations,
nitive function in this domain, with only one study REM sleep, and Parkinsons disease: A medical hypoth-
reporting a negative effect (assessed with the Judgment esis. Neurology 55: 281288.
of Line Orientation test) (Alegret et al., 2001). Avanzi M, Uber E, Bonfa F (2004). Pathological gambling
in two patients on dopamine replacement therapy for
Parkinsons disease. Neurol Sci 25: 98101.
41.4. Conclusions Bakay RA, Starr PA, Vitek JL et al. (1997). Posterior ventral
pallidotomy: Techniques and theoretical considerations.
In order to provide quality care to the PD patient, we Clin Neurosurg 44: 197210.
can no longer be content with just treating the motor Barnes J, David AS (2001). Visual hallucinations in Parkin-
sons disease: A review and phenomenological survey.
symptoms because the cognitive and behavioral mani-
J Neurol Neurosurg Psychiatry 70: 727733.
festations that result from PD treatment are just as dis-
Baron MS, Vitek JL, Bakay RA et al. (2000). Treatment of
abling, if not more so. This is true not only of the advanced Parkinsons disease by unilateral posterior GPi
pharmacologic treatments currently available, but also pallidotomy: 4-year results of a pilot study. Mov Disord
of the various surgical options. Future research focus- 15: 230237.
ing on understanding the underlying mechanisms of Bejjani BP, Damier P, Arnulf I et al. (1999). Transient
drug-induced psychosis and its relation to PD and acute depression induced by high-frequency deep-brain
DLB should enable us to develop better treatments stimulation. N Engl J Med 340: 14761480.
and perhaps alter the long-term outcome of PD Benabid AL, Pollak P, Gervason C et al. (1991). Long-term
patients with psychosis. Elucidating why dopaminer- suppression of tremor by chronic stimulation of the ventral
gic treatment causes abnormal behaviors in certain intermediate thalamic nucleus. Lancet 337: 403406.
Berney A, Vingerhoets F, Perrin A et al. (2002). Effect on
patients and not others should help us in that regard,
mood of subthalamic DBS for Parkinsons disease: A con-
but discovering how DBS can induce or exacerbate
secutive series of 24 patients. Neurology 59: 14271429.
these symptoms may be the key to unlocking how Bezerra ML, Martinez JV, Nasser JA (1999). Transient acute
cognition and behavior are hard-wired in the brain. depression induced by high-frequency deep-brain stimula-
tion. N Engl J Med 341: 1003.
References Breier A, Sutton VK, Feldman PD et al. (2002). Olanzapine
in the treatment of dopamimetic-induced psychosis in
Aarsland D, Larsen JP, Karlsen K et al. (1999). Mental patients with Parkinsons disease. Biol Psychiatry 52:
symptoms in Parkinsons disease are important contribu- 438445.
tors to caregiver distress. Int J Geriatr Psychiatry 14: Burchiel KJ, Anderson VC, Favre J et al. (1999). Comparison
866874. of pallidal and subthalamic nucleus deep brain stimulation
Aarsland D, Larsen JP, Tandberg E et al. (2000). Predictors for advanced Parkinsons disease: Results of a rando-
of nursing home placement in Parkinsons disease: A mized, blinded pilot study. Neurosurgery 45: 13751382.
population-based, prospective study. J Am Geriatr Soc Cahn-Weiner DA, Grace J, Ott BR et al. (2002). Cognitive
48: 938942. and behavioral features discriminate between Alzheimers
Alegret M, Junque C, Valldeoriola F et al. (2001). Effects of and Parkinsons disease. Neuropsychiatry Neuropsychol
bilateral subthalamic stimulation on cognitive function in Behav Neurol 15: 7987.
Parkinson disease. Arch Neurol 58: 12231227. Cantello R, Gilli M, Riccio A et al. (1986). Mood changes asso-
Allen RL, Walker Z, DAth PJ et al. (1995). Risperidone for ciated with end-of-dose deterioration in Parkinsons dis-
psychotic and behavioural symptoms in Lewy body ease: A controlled study. J Neurol Neurosurg Psychiatry
dementia. Lancet 346: 185. 49: 11821190.
Alvir JM, Lieberman JA, Safferman AZ et al. (1993). Cloza- Comella CL, Tanner CM, Ristanovic RK (1993). Polysomno-
pine-induced agranulocytosis. Incidence and risk factors in graphic sleep measures in Parkinsons disease patients
the United States. N Engl J Med 329: 162167. with treatment-induced hallucinations. Ann Neurol 34:
American Psychiatric Association (1994). Diagnostic and Sta- 710714.
tistical Manual of Mental Disorders, 4th edn. (DSM-IV). Cummings JL (1997). The Neuropsychiatric Inventory:
American Psychiatric Association, Washington, DC. Assessing psychopathology in dementia patients.
Apaydin H, Ahlskog JE, Parisi JE et al. (2002). Parkinson Neurology 48: S10S16.
disease neuropathology: Later-developing dementia and Daniele A, Albanese A, Contarino MF et al. (2003). Cogni-
loss of the levodopa response. Arch Neurol 59: 102112. tive and behavioural effects of chronic stimulation of the
236 K. L. CHOU AND J. H. FRIEDMAN

subthalamic nucleus in patients with Parkinsons disease. J French Clozapine Parkinson Study Group (1999). Clozapine
Neurol Neurosurg Psychiatry 74: 175182. drug-induced psychosis in Parkinsons disease. Lancet
de Ajuriaguerra J (1972). Etude psychopathologique des par- 353: 20412042.
kinsoniens. In: J de Ajuriaguerra, G Gauthier (Eds.), Friedenberg DL, Cummings JL (1989). Parkinsons disease,
Monoamines, Noyaux Gris Centraux et Syndrome de depression, and the on-off phenomenon. Psychosomatics
Parkinson. Masson, Geneve, pp. 327355. 30: 9499.
Dewey RB Jr, OSuilleabhain PE (2000). Treatment of Friedman JH (1985). Drug holidays in the treatment of Par-
drug-induced psychosis with quetiapine and clozapine in kinsons disease. A brief review. Arch Intern Med 145:
Parkinsons disease. Neurology 55: 17531754. 913915.
Diederich NJ, Alesch F, Goetz CG (2000). Visual hallucina- Friedman JH (1991). The management of the levodopa
tions induced by deep brain stimulation in Parkinsons psychoses. Clin Neuropharmacol 14: 283295.
disease. Clin Neuropharmacol 23: 287289. Friedman JH (1994). Punding on levodopa. Biol Psychiatry
Doshi PK, Chhaya N, Bhatt MH (2002). Depression leading 36: 350351.
to attempted suicide after bilateral subthalamic nucleus Friedman JH (2005). More on repetitive behaviors in Parkin-
stimulation for Parkinsons disease. Mov Disord 17: sons disease. Mov Disord 20, 509510.
10841085. Friedman JH, Factor SA (2000). Atypical antipsychotics in
Drevets WC (2003). Neuroimaging abnormalities in the the treatment of drug-induced psychosis in Parkinsons
amygdala in mood disorders. Ann NY Acad Sci 985: disease. Mov Disord 15: 201211.
420444. Friedman JH, Lannon MC (1990). Clozapine-responsive tre-
Driver-Dunckley E, Samanta J, Stacy M (2003). Pathological mor in Parkinsons disease. Mov Disord 5: 225229.
gambling associated with dopamine agonist therapy in Friedman JH, Koller WC, Lannon MC et al. (1997). Benztro-
Parkinsons disease. Neurology 61: 422423. pine versus clozapine for the treatment of tremor in
Evans AH, Katzenschlager R, Paviour D et al. (2004). Pund- Parkinsons disease. Neurology 48: 10771081.
ing in Parkinsons disease: Its relation to the dopamine Friedman JH, Messing S, Oakes D et al. (2002). Parkinson
dysregulation syndrome. Mov Disord 19: 397405. Study Group. A descriptive and comparative analysis of
Factor SA, Molho ES, Brown DL (1995). Combined psychotic symptoms in three placebo-controlled, double-
clozapine and electroconvulsive therapy for the treatment blinded trials of atypical antipsychotic drugs in the treat-
of drug-induced psychosis in Parkinsons disease. ment of drug-induced psychosis in Parkinsons disease.
J Neuropsychiatry Clin Neurosci 7: 304307. Mov Disord 17: 1105.
Factor SA, Feustel PJ, Friedman JH et al. (2003). Longitudi- Friedman JH, Berman R, Carson W et al. (2005). Low dose
nal outcome of Parkinsons disease patients with psychosis. aripiprazole for the treatment of drug induced psychosis
Neurology 60: 17561761. in Parkinsons disease. Ninth International Congress of
Favre J, Burchiel KJ, Taha JM et al. (2000). Outcome of uni- Parkinsons Disease and Movement Disorders, New
lateral and bilateral pallidotomy for Parkinsons disease: Orleans, MS.
Patient assessment. Neurosurgery 46: 344353. Funkiewiez A, Ardouin C, Caputo E et al. (2004). Long term
Fenelon G, Mahieux F, Huon R et al. (2000). Hallucinations effects of bilateral subthalamic nucleus stimulation on
in Parkinsons disease: Prevalence, phenomenology and cognitive function, mood, and behaviour in Parkinsons
risk factors. Brain 123 (Pt 4), 733745. disease. J Neurol Neurosurg Psychiatry 75: 834839.
Fenelon G, Thobois S, Bonnet AM et al. (2002). Tactile hallu- Ghika J, Villemure JG, Fankhauser H et al. (1998). Efficiency
cinations in Parkinsons disease. J Neurol 249: 16991703. and safety of bilateral contemporaneous pallidal stimu-
Fernandez HH, Friedman JH (1999). Punding on L-dopa. lation (deep brain stimulation) in levodopa-responsive
Mov Disord 14: 836838. patients with Parkinsons disease with severe motor
Fernandez HH, Donnelly EM, Friedman JH (2004a). Long- fluctuations: A 2-year follow-up review. J Neurosurg 89:
term outcome of clozapine use for psychosis in 713718.
parkinsonian patients. Mov Disord 19: 831833. Ghika J, Ghika-Schmid F, Fankhauser H et al. (1999). Bilat-
Fernandez HH, Trieschmann ME, Friedman JH (2004b). Ari- eral contemporaneous posteroventral pallidotomy for the
piprazole for drug-induced psychosis in Parkinson disease: treatment of Parkinsons disease: Neuropsychological
Preliminary experience. Clin Neuropharmacol 27: 45. and neurological side effects. Report of four cases and
Fernandez HH, Trieschmann ME, Okun MS (2004c). Rebound review of the literature. J Neurosurg 91: 313321.
psychosis: Effect of discontinuation of antipsychotics in Giovannoni G, OSullivan JD, Turner K et al. (2000). Hedonis-
Parkinsons disease. Mov Disord 20 (1), 104105. tic homeostatic dysregulation in patients with Parkinsons
Fields JA, Troster AI, Wilkinson SB et al. (1999). Cognitive disease on dopamine replacement therapies. J Neurol
outcome following staged bilateral pallidal stimulation for Neurosurg Psychiatry 68: 423428.
the treatment of Parkinsons disease. Clin Neurol Neuro- Gironell A, Kulisevsky J, Rami L et al. (2003). Effects of
surg 101: 182188. pallidotomy and bilateral subthalamic stimulation on
Ford B, Lynch T, Greene P (1994). Risperidone in Parkinsons cognitive function in Parkinson disease. A controlled
disease. Lancet 344: 681. comparative study. J Neurol 250: 917923.
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 237

Goetz CG, Stebbins GT (1993). Risk factors for nursing Houeto JL, Mesnage V, Mallet L et al. (2002). Behavioural
home placement in advanced Parkinsons disease. Neurol- disorders, Parkinsons disease and subthalamic stimula-
ogy 43: 22272229. tion. J Neurol Neurosurg Psychiatry 72: 701707.
Goetz CG, Stebbins GT (1995). Mortality and hallucinations Hurwitz TA, Calne DB, Waterman K (1988). Treatment of
in nursing home patients with advanced Parkinsons dis- dopaminomimetic psychosis in Parkinsons disease with
ease. Neurology 45: 669671. electroconvulsive therapy. Can J Neurol Sci 15: 3234.
Goetz CG, Tanner CM, Klawans HL (1982a). Drug holiday Inzelberg R, Kipervasser S, Korczyn AD (1998). Auditory
in the management of Parkinson disease. Clin Neurophar- hallucinations in Parkinsons disease. J Neurol Neurosurg
macol 5: 351364. Psychiatry 64: 533535.
Goetz CG, Tanner CM, Klawans HL (1982b). Pharmacology Jahanshahi M, Ardouin CM, Brown RG et al. (2000). The
of hallucinations induced by long-term drug therapy. Am J impact of deep brain stimulation on executive function
Psychiatry 139: 494497. in Parkinsons disease. Brain 123 (Pt 6), 11421154.
Goetz CG, Pappert EJ, Blasucci LM et al. (1998a). Intrave- Jansen EN (1994). Clozapine in the treatment of tremor in
nous levodopa in hallucinating Parkinsons disease Parkinsons disease. Acta Neurol Scand 89: 262265.
patients: High-dose challenge does not precipitate halluci- Jeste DV (2004). Tardive dyskinesia rates with atypical anti-
nations. Neurology 50: 515517. psychotics in older adults. J Clin Psychiatry 65 (Suppl 9),
Goetz CG, Vogel C, Tanner CM et al. (1998b). Early dopami- 2124.
nergic drug-induced hallucinations in parkinsonian patients. Kane J, Honigfeld G, Singer J et al. (1988). Clozapine for the
Neurology 51: 811814. treatment-resistant schizophrenic. A double-blind compar-
Goetz CG, Blasucci LM, Leurgans S et al. (2000). Olanzapine ison with chlorpromazine. Arch Gen Psychiatry 45:
and clozapine: Comparative effects on motor function in 789796.
hallucinating PD patients. Neurology 55: 789794. Kelly PJ, Gillingham FJ (1980). The long-term results of
Graham JM, Grunewald RA, Sagar HJ (1997). Hallucinosis stereotaxic surgery and L-dopa therapy in patients with
in idiopathic Parkinsons disease. J Neurol Neurosurg Parkinsons disease. A 10-year follow-up study. J Neuro-
Psychiatry 63: 434440. surg 53: 332337.
Groenewegen HJ, Berendse HW (1990). Connections of the Klein C, Gordon J, Pollak L et al. (2003). Clozapine in
subthalamic nucleus with ventral striatopallidal parts of Parkinsons disease psychosis: 5-year follow-up review.
the basal ganglia in the rat. J Comp Neurol 294: 607622. Clin Neuropharmacol 26: 811.
Gschwandtner U, Aston J, Renaud S et al. (2001). Pathologic Kleiner-Fisman G, Fisman DN, Sime E et al. (2003). Long-
gambling in patients with Parkinsons disease. Clin term follow up of bilateral deep brain stimulation of the
Neuropharmacol 24: 170172. subthalamic nucleus in patients with advanced Parkinson
Haeske-Derwick HC (1995). Hallucinations in Parkinsons disease. J Neurosurg 99: 489495.
disease: Characteristics and associated clinical features. Kofman OS (1984). Are levodopa drug holidays justified?
Int J Geriatr Psychiatry 10: 487495. Can J Neurol Sci 11: 206209.
Hardie RJ, Lees AJ, Stern GM (1984). On-off fluctuations in Koob GF, Le Moal M (1997). Drug abuse: Hedonic homeo-
Parkinsons disease. A clinical and neuropharmacological static dysregulation. Science 278: 5258.
study. Brain 107 (Pt 2), 487506. Krack P, Limousin P, Benabid AL et al. (1997). Chronic stimu-
Hariz MI, Johansson F, Shamsgovara P et al. (2000). Bilateral lation of subthalamic nucleus improves levodopa-induced
subthalamic nucleus stimulation in a parkinsonian patient dyskinesias in Parkinsons disease. Lancet 350: 1676.
with preoperative deficits in speech and cognition: Persis- Krack P, Kumar R, Ardouin C et al. (2001). Mirthful laugh-
tent improvement in mobility but increased dependency: ter induced by subthalamic nucleus stimulation. Mov Dis-
A case study. Mov Disord 15: 136139. ord 16: 867875.
Herzog J, Reiff J, Krack P et al. (2003a). Manic episode with Krack P, Batir A, Van Blercom N et al. (2003). Five-year
psychotic symptoms induced by subthalamic nucleus follow-up of bilateral stimulation of the subthalamic
stimulation in a patient with Parkinsons disease. Mov nucleus in advanced Parkinsons disease. N Engl J Med
Disord 18: 13821384. 349: 19251934.
Herzog J, Volkmann J, Krack P et al. (2003b). Two-year Kubu CS, Grace GM, Parrent AG (2000). Cognitive outcome
follow-up of subthalamic deep brain stimulation in following pallidotomy: The influence of side of surgery and
Parkinsons disease. Mov Disord 18: 13321337. age of patient at disease onset. J Neurosurg 92: 384389.
Higginson CI, Fields JA, Troster AI (2001). Which symp- Kulisevsky J, Berthier ML, Gironell A et al. (2002). Mania
toms of anxiety diminish after surgical interventions for following deep brain stimulation for Parkinsons disease.
Parkinson disease? Neuropsychiatry Neuropsychol Behav Neurology 59: 14211424.
Neurol 14: 117121. Kurlan R (2004). Disabling repetitive behaviors in Parkin-
Hillen ME, Sage JI (1996). Nonmotor fluctuations in patients sons disease. Mov Disord 19: 433437.
with Parkinsons disease. Neurology 47: 11801183. Lacritz LH, Cullum CM, Frol AB et al. (2000). Neuropsycho-
Holroyd S, Currie L, Wooten GF (2001). Prospective study logical outcome following unilateral stereotactic pallidot-
of hallucinations and delusions in Parkinsons disease. omy in intractable Parkinsons disease. Brain Cogn 42:
J Neurol Neurosurg Psychiatry 70: 734738. 364378.
238 K. L. CHOU AND J. H. FRIEDMAN

Laitinen LV (2000). Behavioral complications of early Menza MA, Golbe LI, Cody RA et al. (1993). Dopamine-
pallidotomy. Brain Cogn 42: 313323. related personality traits in Parkinsons disease. Neurology
Lawrence AD, Evans AH, Lees AJ (2003). Compulsive use 43: 505508.
of dopamine replacement therapy in Parkinsons disease: Merello M, Starkstein S, Nouzeilles MI et al. (2001). Bilat-
Reward systems gone awry? Lancet Neurol 2: 595604. eral pallidotomy for treatment of Parkinsons disease
Lezak MD (1995). Executive function. In: MD Lezak, (Ed.), induced corticobulbar syndrome and psychic akinesia
Neuropsychological Assessment. Oxford University Press, avoidable by globus pallidus lesion combined with con-
Oxford. tralateral stimulation. J Neurol Neurosurg Psychiatry 71:
Limousin P, Krack P, Pollak P et al. (1998). Electrical stimu- 611614.
lation of the subthalamic nucleus in advanced Parkinsons Merims D, Galili-Mosberg R, Melamed E (2000). Is there
disease. N Engl J Med 339: 11051111. addiction to levodopa in patients with Parkinsons disease?
Lombardi WJ, Gross RE, Trepanier LL et al. (2000). Rela- Mov Disord 15: 10141016.
tionship of lesion location to cognitive outcome following Miwa H, Morita S, Nakanishi I et al. (2004). Stereotyped
microelectrode-guided pallidotomy for Parkinsons dis- behaviors or punding after quetiapine administration in
ease: Support for the existence of cognitive circuits in Parkinsons disease. Parkinsonism Relat Disord 10:
the human pallidum. Brain 123 (Pt 4), 746758. 177180.
Maricle RA, Nutt JG, Carter JH (1995a). Mood and anxiety Miyawaki E, Perlmutter JS, Troster AI et al. (2000). The
fluctuation in Parkinsons disease associated with levo- behavioral complications of pallidal stimulation: A case
dopa infusion: Preliminary findings. Mov Disord 10: report. Brain Cogn 42: 417434.
329332. Molina JA, Sainz-Artiga MJ, Fraile A et al. (2000). Patholo-
Maricle RA, Nutt JG, Valentine RJ et al. (1995b). Dose- gic gambling in Parkinsons disease: A behavioral mani-
response relationship of levodopa with mood and anxiety festation of pharmacologic treatment? Mov Disord 15:
in fluctuating Parkinsons disease: A double-blind, 869872.
placebo-controlled study. Neurology 45: 17571760. Morgante L, Epifanio A, Spina E et al. (2004). Quetiapine
Martinez-Martin P, Valldeoriola F, Molinuevo JL et al. and clozapine in parkinsonian patients with dopaminergic
(2000). Pallidotomy and quality of life in patients with Par- psychosis. Clin Neuropharmacol 27: 153156.
kinsons disease: An early study. Mov Disord 15: 6570.
Moro E, Scerrati M, Romito LM et al. (1999). Chronic
Martinez-Martin P, Valldeoriola F, Tolosa E et al. (2002).
subthalamic nucleus stimulation reduces medication
Bilateral subthalamic nucleus stimulation and quality of
requirements in Parkinsons disease. Neurology 53: 8590.
life in advanced Parkinsons disease. Mov Disord 17:
Nausieda PA (1985). Sinemet abusers. Clin Neuropharma-
372377.
col 8: 318327.
Masterman D, DeSalles A, Baloh RW et al. (1998). Motor,
Nausieda PA, Weiner WJ, Kaplan LR et al. (1982). Sleep
cognitive, and behavioral performance following unilat-
disruption in the course of chronic levodopa therapy: An
eral ventroposterior pallidotomy for Parkinson disease.
early feature of the levodopa psychosis. Clin Neurophar-
Arch Neurol 55: 12011208.
macol 5: 183194.
Matsui H, Udaka F, Oda M et al. (2004). [Two cases of Par-
kinsons disease in which visual hallucinations disappeared Nissenbaum H, Quinn NP, Brown RG et al. (1987). Mood
after cataract surgery.] No To Shinkei 56: 351354. swings associated with the on-off phenomenon in
McDowell SA, Harris JP (1997). Visual problems in Parkin- Parkinsons disease. Psychol Med 17: 899904.
sons disease: A questionnaire survey. Behav Neurol 10: Obwegeser AA, Uitti RJ, Lucas JA et al. (2000). Predictors
7781. of neuropsychological outcome in patients following
McKeith IG, Ballard CG, Harrison RW (1995). Neuroleptic microelectrode-guided pallidotomy for Parkinsons dis-
sensitivity to risperidone in Lewy body dementia. Lancet ease. J Neurosurg 93: 410420.
346: 699. Okun MS, Bakay RA, DeLong MR et al. (2003). Transient
McKeith IG, Galasko D, Kosaka K et al. (1996). Consensus manic behavior after pallidotomy. Brain Cogn 52: 281283.
guidelines for the clinical and pathologic diagnosis of Olanow CW, Schapira AH, Roth T (2000). Waking up to
dementia with Lewy bodies (DLB): Report of the sleep episodes in Parkinsons disease. Mov Disord 15:
consortium on DLB international workshop. Neurology 212215.
47: 11131124. Ondo WG, Levy JK, Vuong KD et al. (2002). Olanzapine
Meco G, Bonifati V, Cusimano G et al. (1990). Hallucina- treatment for dopaminergic-induced hallucinations. Mov
tions in Parkinson disease: Neuropsychological study. Ital Disord 17: 10311035.
J Neurol Sci 11: 373379. Ondo WG, Tintner R, Vuong KD et al. (2005). Double-blind,
Meco G, Alessandria A, Bonifati V et al. (1994). Risperidone placebo-controlled, unforced titration parallel trial of
for hallucinations in levodopa-treated Parkinsons disease quetiapine for dopaminergic-induced hallucinations in
patients. Lancet 343: 13701371. Parkinsons disease. Mov Disord 20, 958963.
Menza MA, Sage J, Marshall E et al. (1990). Mood changes Pappert EJ, Goetz CG, Niederman FG et al. (1999). Hallucina-
and on-off phenomena in Parkinsons disease. Mov tions, sleep fragmentation, and altered dream phenomena in
Disord 5: 148151. Parkinsons disease. Mov Disord 14: 117121.
TREATMENT-INDUCED MENTAL CHANGES IN PARKINSONS DISEASE 239

Parkinson Study Group (1999). Low-dose clozapine for the Saint-Cyr JA, Trepanier LL, Kumar R et al. (2000). Neurop-
treatment of drug-induced psychosis in Parkinsons dis- sychological consequences of chronic bilateral stimulation
ease. N Engl J Med 340: 757763. of the subthalamic nucleus in Parkinsons disease. Brain
Parkinson Study Group (2000). Pramipexole vs levodopa as 123 (Pt 10), 20912108.
initial treatment for Parkinson disease: A randomized con- Sanchez-Ramos J (2002). The straight dope on addiction to
trolled trial. JAMA 284: 19311938. dopamimetic drugs. Mov Disord 17: 223225.
Perozzo P, Rizzone M, Bergamasco B et al. (2001). Deep Sanchez-Ramos JR, Ortoll R, Paulson GW (1996). Visual
brain stimulation of the subthalamic nucleus in Parkinsons hallucinations associated with Parkinson disease. Arch
disease: Comparison of pre- and postoperative neuropsy- Neurol 53: 12651268.
chological evaluation. J Neurol Sci 192: 915. Seedat S, Kesler S, Niehaus DJ et al. (2000). Pathological
Pillon B, Deweer B, Michon A et al. (1994). Are explicit gambling behaviour: Emergence secondary to treatment
memory disorders of progressive supranuclear palsy of Parkinsons disease with dopaminergic agents. Depress
related to damage to striatofrontal circuits? Comparison Anxiety 11: 185186.
with Alzheimers, Parkinsons, and Huntingtons diseases. Soukup VM, Ingram F, Schiess MC et al. (1997). Cognitive
Neurology 44: 12641270. sequelae of unilateral posteroventral pallidotomy. Arch
Pillon B, Ardouin C, Damier P et al. (2000). Neuropsycholo- Neurol 54: 947950.
gical changes between off and on STN or GPi stimu- Spiegel EA, Wycis HT, Marks M et al. (1947). Stereotaxic
lation in Parkinsons disease. Neurology 55: 411418. apparatus for operations on the human brain. Science
Pillon B, Boller F, Levy R et al. (2001). Cognitive deficits 106: 349350.
and dementia in Parkinsons disease. In: F Boller, S Straits-Troster K, Fields JA, Wilkinson SB et al. (2000).
Cappa (Eds.), Vol. 6, Elsevier Science BV, Amsterdam, Health-related quality of life in Parkinsons disease after
pp. 311371. pallidotomy and deep brain stimulation. Brain Cogn 42:
Pollak P, Tison F, Rascol O et al. (2004). Clozapine in drug 399416.
induced psychosis in Parkinsons disease: A randomised, Svennilson E, Torvik A, Lowe R et al. (1960). Treatment of
placebo controlled study with open follow up. J Neurol parkinsonism by stereotatic thermolesions in the pallidal
Neurosurg Psychiatry 75: 689695. region. A clinical evaluation of 81 cases. Acta Psychiatr
Priebe S (1984). Levodopa dependence: A case report. Phar- Scand 35: 358377.
macopsychiatry 17: 109110. Thobois S, Mertens P, Guenot M et al. (2002). Subthalamic
Quinn NP (1998). Classification of fluctuations in patients nucleus stimulation in Parkinsons disease: Clinical
with Parkinsons disease. Neurology 51: S25S29. evaluation of 18 patients. J Neurol 249: 529534.
Rascol O, Brooks DJ, Korczyn AD et al. (2000). A five-year Tousi B, Frankel M (2004). Olfactory and visual hallucina-
study of the incidence of dyskinesia in patients with early tions in Parkinsons disease. Parkinsonism Relat Disord
Parkinsons disease who were treated with ropinirole 10: 253254.
or levodopa. 056 Study Group. N Engl J Med 342: Trepanier LL, Saint-Cyr JA, Lozano AM et al. (1998). Neu-
14841491. ropsychological consequences of posteroventral pallidot-
Raudino F (2001). Non motor off in Parkinsons disease. omy for the treatment of Parkinsons disease. Neurology
Acta Neurol Scand 104: 312315. 51: 207215.
Richard IH, Justus AW, Kurlan R (2001). Relationship Trepanier LL, Kumar R, Lozano AM et al. (2000). Neurop-
between mood and motor fluctuations in Parkinsons sychological outcome of GPi pallidotomy and GPi or
disease. J Neuropsychiatry Clin Neurosci 13: 3541. STN deep brain stimulation in Parkinsons disease. Brain
Ridley RM, Baker HF (1982). Stereotypy in monkeys and Cogn 42: 324347.
humans. Psychol Med 12: 6172. Troster AI, Fields JA, Wilkinson SB et al. (1997). Unilateral
Rodriguez-Oroz MC, Zamarbide I, Guridi J et al. (2004). pallidal stimulation for Parkinsons disease: Neurobeha-
Efficacy of deep brain stimulation of the subthalamic vioral functioning before and 3 months after electrode
nucleus in Parkinsons disease 4 years after surgery: Dou- implantation. Neurology 49: 10781083.
ble blind and open label evaluation. J Neurol Neurosurg Troster AI, Fields JA, Wilkinson S et al. (2003). Effect of
Psychiatry 75: 13821385. motor improvement on quality of life following subthala-
Romito LM, Raja M, Daniele A et al. (2002). Transient mic stimulation is mediated by changes in depressive
mania with hypersexuality after surgery for high frequency symptomatology. Stereotact Funct Neurosurg 80: 4347.
stimulation of the subthalamic nucleus in Parkinsons Turecki G, Mari Jde J, Del Porto JA (1993). Bipolar disorder
disease. Mov Disord 17: 13711374. following a left basal-ganglia stroke. Br J Psychiatry 163: 690.
Rylander G (1972). Psychoses and the punding and chorei- Turner KR, Reid WG, Homewood J et al. (2002). Neurop-
form syndromes in addiction to central stimulant drugs. sychological sequelae of bilateral posteroventral pallidot-
Psychiatr Neurol Neurochir 75: 203212. omy. J Neurol Neurosurg Psychiatry 73: 444446.
Saint-Cyr JA, Trepanier LL (2000). Neuropsychologic Uitti RJ, Wharen REJr, Turk MF et al. (1997). Unilateral pal-
assessment of patients for movement disorder surgery. lidotomy for Parkinsons disease: Comparison of outcome in
Mov Disord 15: 771783. younger versus elderly patients. Neurology 49: 10721077.
240 K. L. CHOU AND J. H. FRIEDMAN

Varma TR, Fox SH, Eldridge PR et al. (2003). Deep brain sti- Wolters EC, Hurwitz TA, Mak E et al. (1990). Clozapine in
mulation of the subthalamic nucleus: Effectiveness in the treatment of parkinsonian patients w ith dopaminomi-
advanced Parkinsons disease patients previously reliant on metic psychosis. Neurology 40: 832834.
apomorphine. J Neurol Neurosurg Psychiatry 74: 170174. Wolters EC, Jansen EN, Tuynman-Qua HG et al. (1996). Olan-
Vingerhoets G, van der Linden C, Lannoo E et al. (1999). zapine in the treatment of dopaminomimetic psychosis in
Cognitive outcome after unilateral pallidal stimulation in patients with Parkinsons disease. Neurology 47: 10851087.
Parkinsons disease. J Neurol Neurosurg Psychiatry 66: Woods SP, Fields JA, Troster AI (2002). Neuropsychological
297304. sequelae of subthalamic nucleus deep brain stimulation in
Volkmann J, Allert N, Voges J et al. (2001). Safety and effi- Parkinsons disease: A critical review. Neuropsychol Rev
cacy of pallidal or subthalamic nucleus stimulation in 12: 111126.
advanced PD. Neurology 56: 548551. Workman RH Jr, Orengo CA, Bakey AA et al. (1997). The
Voon V (2004). Repetition, repetition, and repetition: Compul- use of risperidone for psychosis and agitation in demented
sive and punding behaviors in Parkinsons disease. Mov patients with Parkinsons disease. J Neuropsychiatry Clin
Disord 19: 367370. Neurosci 9: 594597.
Section 7

Surgical treatment
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 42

Ablative surgery for the treatment of Parkinsons disease

FREDERICK A. LENZ*

Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA

42.1. Introduction 42.2. Mechanisms of parkinsonian


motor symptoms
Spiegel and Wycis (1954; Cooper, 1954; Spiegel et al.,
1958) pioneered stereotactic surgery for the treatment PD affects approximately 1% of the population over 65
of Parkinsons disease (PD) by introducing coagula- years (Adams et al., 1996). The three cardinal signs of
tion of the globus pallidus stereotactic pallidotomy. PD are resting tremor, cogwheel rigidity and bradykine-
(Hassler et al., 1979), introduced ablation of the ventral sia (Paulson and Stern, 1997). After years of treatment
oral nucleus of thalamus (Vo, consisting of Voa and with levodopa, patients with PD often develop intract-
Vop, anterior and posterior subnuclei), the terminus able complications of this therapy, including dyskinesias
for pallidal afferents to the thalamus. Microelectrode and fluctuations (Marsden and Parkes, 1977; Marsden
recordings demonstrated that the area posterior to and Fahn, 1987). The forebrain mechanisms of some
Vop, i.e. the terminus of cerebellar afferents (ventral of these symptoms have recently been clarified, lead-
intermediate: Vim), was later found to have rhythmic ing to a rationale for effective targets for treatment
bursting activity close to the frequency of tremor (Guiot of PD.
et al., 1962). Vim then became the target of choice for
tremor of all types. 42.2.1. Bradykinesia
The introduction of levodopa in the early 1970s led
to a dramatic decrease in the number of stereotactic The motor circuit of the basal ganglia includes a
surgeries for movement disorders (Levy, 1992). Two direct inhibitory pathway from the putamen to the
decades later, drug-related complications of medical internal segment of the GPi and an indirect, excita-
treatment, including dyskinesias and fluctuations, had tory pathway from the putamen to GPi via the
proven intractable (Marsden and Parkes, 1977; Marsden STN (see Ch. 1). In monkeys, GPi-inhibitory
and Fahn, 1987). Neurophysiologic studies in the (GABAergic) efferents in the motor loop project to
realistic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurons of the monkey thalamic nucleus ventral lat-
(MPTP) model of PD had demonstrated increased activ- eral pars oralis (VLo) (Penney and Young, 1981;
ity in the subthalamic nucleus (STN) and the internal Young and Penney, 1984), corresponding to human
segment of the globus pallidus (GPi) (Bergmam et al., Vop. Cortical projections activate these pathways,
1990). Finally, the demonstration that posteroventral with a resultant reduction in inhibitory GPi activity
GPi pallidotomy was effective for advanced PD via the direct pathway and an increase in inhibitory
(Laitinen et al., 1992) led to a resurgence in ablative GPi activity via the indirect pathway.
approaches for the treatment of PD. The introduction The motor manifestations of parkinsonism are
of stimulation techniques led to decreased frequency seen with dysfunction of the nigrostriatal dopaminergic
of ablative approaches to the movement disorders pathway (Hornykiewicz, 1974). Dopaminergic projections
(Siegfried and Lippitz, 1994; Benabid et al., 1996; from the substantia nigra tend to activate the direct path-
Clatterbuck et al., 2000). way (D1 dopamine receptor-mediated) and decrease the

*Correspondence to: Professor F. A. Lenz, Department of Neurosurgery, Meyer Building 8-181, Johns Hopkins Hospital,
600 North Wolfe Street, Baltimore, MD 21287-7713, USA. E-mail: flenz1@jhmi.edu, Tel: 1-410-955-2257, Fax:1- 410-
614-9877.
244 F. A. LENZ
activity of the indirect pathway (D2-receptor-mediated). et al., 1994). Therefore sensory cells participating in a
Therefore, in PD there is decreased inhibition of reflex or feedback loop might cause tremor. Finally, a
GPi through the direct pathway and increased excita- transfer function analysis has demonstrated a feedback
tion though the indirect pathway. As a result of these loop in > 90% of cells in the Vim and Vop (Schnider
effects of dopamine deficiency there is increased GPi et al., 1989).
activity leading to inhibition of Vop (Vitek et al.,
1993). Thus thalamocortical and corticothalamic 42.2.3. Dyskinesias
motor circuits might be inhibited, leading to slowing
of movement initiation and of movement itself, i.e. The current model of hyperkinetic disorders (DeLong,
bradykinesia (Penney and Young, 1981; Young and 1990) suggests that a decrease in pallidal activity dis-
Penney, 1984; DeLong, 1990). inhibits the pallidal relays, VLo/Vop, by decreased
inhibitory input to the thalamus from the pallidum
42.2.2. Tremor (Penney and Young, 1981). In hemiballism the activity
of neurons in GPi is characterized by firing rates
In the case of Parkinsons tremor the most current which are lower and more irregular than parkinonsian
hypothesis is a thalamic oscillator activated by hyperpo- patients off or on medications (intravenous apo-
larization of the thalamocortical cells in VLo/Vop due morphine) (Suarez et al., 1997; Vitek et al., 1999),
to increased inhibitory input from the pallidum in PD consistent with studies in monkeys (Nini et al., 1995).
(Bergman et al., 1990; Pare et al., 1990; see also The pauses and grouped discharges of GPi neurons
Lamarre and Joffroy, 1979; Marsden, 1984; Elble and may be related to the random bursts of electromyo-
Koller, 1990). When inhibited, these cells generate a gram (EMG) activity hemiballistic movements (Vitek
somatic calcium spike which produces an action poten- et al., 1999). The importance of patterned GPi activity
tial burst (LTS bursts, low-threshold spike bursts) with in hemiballismus is consistent with the efficacy of pal-
a following inhibition, leading to recurrent oscillations lidotomy since pallidotomy will interrupt activity but
(Filion et al., 1988; Buzsaki et al., 1990; Pare et al., not decreased pallidal firing (DeLong, 1990). A simi-
1990). However such bursts are rarely found in recordings lar mechanism seems to be involved in drug (apomor-
from Vim and Vop of parkinsonian patients undergoing phine)-related dyskinesias (Hutchison et al., 1997a;
thalamotomy (Zirh et al., 1998; Magnin et al., 2000). Merello et al., 1999).
Another proposed central generator is GPi, specifi-
cally neurons that project to and inhibit Vop neurons. 42.2.4. Dystonia
This hypothesis is inconsistent with higher rates of tre-
mor-related activity in a cerebellar relay (Vim: 25%) Dystonia is a common feature of PD. During dystonia
than a pallidal relay of patients with PD (Vop: 21%) of multiple etiologies, firing rates of cells in GPi are
(Lenz et al., 1994; Hua et al., 1998). Furthermore, patterned and are intermediate between those in hemi-
GPi activity at tremor frequency is rarely correlated ballismus and normal monkeys (Vitek et al., 1995,
with tremor in PD (Hutchison et al., 1997b; Lemstra 1998b; Suarez et al., 1997). However, normal firing
et al., 1999) or in monkey models of parkinsonism rates can occur with mild dystonia whereas decreased
(Raz et al., 2000). Therefore, the activity of cells in firing rates develop with repeated active movements
thalamus and pallidum is not consistent with a pallidal which also exacerbate dystonia (Lenz et al., 1998). Thus
generator for parkinsonian tremor. the inverse relationship between GPi cellular firing and
Another proposed mechanism of parkinsonian dystonia may be the result of cortically mediated, activ-
tremor is the peripheral-feedback hypothesis. This ity-dependent changes in striatal activity transmitted to
hypothesis proposes that tremor is the oscillation of GPi and then to Vop through the direct pathway (Albin
unstable stretch reflex arcs (long-loop reflex arc) et al., 1989; Carlson and Carlson, 1990).
which may traverse motor cortex in much the same way Decreased patterned firing of GPi neurons in
tendon tap reflexes traverse the spinal cord (Desmedt, patients with dystonia should produce an increased
1978; Lenz et al., 1983a, b). The increased gain of these patterned firing in the monkey/human thalamic palli-
reflexes may cause parkinsonian rigidity and tremor, in dal relay VLo/Vop. However, decreased, patterned
much the same way as increased spinal reflexes cause firing was reported in Vop more than Vim during dys-
spasticity and clonus (Stein and Lee, 1981). This tonia (Lenz et al., 1999), perhaps related to the
hypothesis has been supported by the finding that thala- dystonia-related plasticity of firing in GPi (Lenz
mic neuronal activity precedes tremor in the case of et al., 1998). Thalamic dystonia frequency activity
thalamic neurons with sensory inputs, but not those correlated with and leading dystonia is significantly
without (Strick, 1976; Lenz et al., 1990, 1994; Vitek more common in Vop than Vim. Lesions of GPi
ABLATIVE SURGERY FOR THE TREATMENT OF PARKINSONS DISEASE 245
(Vitek and Lenz, 1998) and presumed Vop (Andrew Attempts to decrease errors in MRI scan due to
et al., 1983; Tasker et al., 1988; Cardoso et al., 1995; these artifacts include software modifications and
Lenz et al., 1999) can relieve dystonia, which suggests overlapping (fusion) of the three-dimensional MRI
that, for neurons in Vop, patterned activity is more database with the CT database which is not prone to
important than decreased firing rates in the mechanism these types of artifacts (Alexander et al., 1995). These
of dystonia (Lenz et al., 1999). Therefore, there is databases can then be merged with atlas maps of anat-
reasonable rationale for lesions of the pallidum and omy to estimate the location of nuclei relative to the
Vop as treatment for the dystonia of PD. radiologic anatomy. These atlas maps may be trans-
formed either to match the ACPC line in isolation or
to match the ACPC line and other structures, such as
42.3. Stereotactic surgical technique
the margins of the third ventricle or the internal
capsule (Kelly et al., 1987; Dostrovsky, 1998).
Radiologic and physiologic landmarks are usually used
Another approach is to estimate the target directly
to localize accurately the target for ablative procedures
from the ACPC line as determined radiologically.
(Bakay et al., 1992; Lozano et al., 1996; Garonzik et al.,
The target for Vim is 3 mm anterior and 14 mm lateral
2002; Patel et al., 2003). Many surgeons employ radi-
to PC and 2 mm above the ACPC line. Alternatively,
ologic localization of the anterior (AC) and posterior
the target in Vim can be estimated in the lateral plane
commissure (PC) and of the border between the capsule
midway between the internal capsule and the lateral
and thalamus using computed tomography (CT) or
edge of the T2 intense medial dorsal nucleus of the
magnetic resonance imaging (MRI) scans. The radiolo-
thalamus (Hua et al., 2004).
gic estimate of location is refined physiologically
GPi can be estimated 3 mm anterior, 3 mm inferior
by microelectrode recording before radiofrequency
and 2022 mm lateral to the mid commissural point.
lesioning (Mandir et al., 1997; Garonzik et al., 2002;
Direct targeting can be carried out on axial and coro-
Hua et al., 2004). Alternative approaches are to loca-
nal inversion recovery series. On an axial cut through
lize radiologically using ventriculography or CT/MR
the ACPC line at the anterior posterior coordinate is
fusion techniques (Carlson and Iacono, 1999) and to
the mid commissural point and the medial lateral
localize physiologically using semimicroelectrode
coordinate is 4 mm medial to the external lamina
recording or macrostimulation (Burchiel, 1995) and
separating the internal from the external pallidal seg-
to lesion using radiosurgery (Friedman et al., 1996;
ment. The vertical axis was targeted from the location
Kondziolka, 2002). The relative efficacy and safety
of the optic tract and GPi on the coronal cut through
of these different techniques have not been examined
the mid commissural point (Starr et al., 1999).
systematically.
The location of STN can be estimated at 4 mm
posterior, 4 mm inferior and 12 mm lateral to the
42.4. Radiologic localization midpoint of ACPC (Guridi et al., 1999; Patel et al.,
2003). STN can be directly localized using 2 mm,
Ventriculography can still be used as a means of T2-weighted spin-echo sequence axial and coronal
locating the ACPC line (Diederich et al., 1992; scans through the thalamus and midbrain. On such a
Schuurman et al., 2000), although it has largely been series STN can also be localized 7 mm lateral and
replaced by CT and MRI scanning. Computerized 4 mm anterior to the center of the red nucleus (Starr
imaging is as accurate as ventriculography (Tasker et al., 2002). Fusion of other MR series, such as gradi-
et al., 1991) and does not carry the risks of ventricular ent echo T2 and fast spin-echo series or CT can be used
puncture and instillation of contrast media into the to localize STN and target the dorsal part of the poster-
ventricles. MRI scanning is slightly less accurate than ior third of STN (Patel et al., 2003). Physiological
CT scanning, with errors of approximately 2 mm on confirmation of the radiologic estimate is carried out
average and 4 mm at maximum (Kondziolka et al., by stimulation or recording at and around the target.
1992; Gerdes et al., 1994; Holtzheimer et al., 1999).
This error is due to artifacts related to inhomogeneities 42.5. Physiologic localization
and non-linearities in the gradient field the position-
dependent strength in the magnetic field (Hardy and Radiologic targeting should be verified by identifying
Barnett, 1998). Artifacts can be induced by metal or the different nuclei (GPi, STN and Vim) on the basis
magnetic suseptibility artifacts, produced at the of properties such as spontaneous activity and neuro-
interface between materials (e.g. air and bone) which nal firing related to the movement disorders and to
have different tendencies to affect the magnetic field passive and active movements. Nuclear properties are
in a region. also defined by microstimulation (< 100 mA)-evoked
246 F. A. LENZ
sensations and motor effects such as alteration in the stimulation determines the amount of local current
movement disorder, e.g. decreased tone or tremor. spread (Ranck, 1975).
Physiologic localization can be carried out by stimula- Semimicroelectode recordings are carried out using
tion using a macroelectrode (impedance < 1000 ohms) low-impedance microelectrodes with an impedance of
or by stimulation and recording using a semimicroelec- less than 100 kohms. The semimicroelectrode signal is
trode (impedance < 100 kohms) or a microelectrode often amplified against a concentric ring electrode
(impedance > 500 kohms). which is located on a radius of 0.4 mm around the
In our institution a map of physiologic results is microelectrode (Taren et al., 1968; Burchiel, 1995;
made to the same scale as a set of transparent atlas Ohye, 1998). Recordings through a semimicroelec-
maps from the sagittal sections of the Schaltenbrand trode will yield recordings of local field potentials
and Bailey atlas (1959) parasagittal sections for (slow waves) or multiunit activity. Bipolar stimulation
different targets as follows: 13.5 mm lateral atlas through a concentric ring electrode can be used alone or
map for Vim, 21 mm for GPi and 11 mm for the in combination with recording through a semimicroelec-
STN. The fusion and fitting of the imaging studies to trode (Ojemann and Ward, 1982; Tasker et al., 1982;
the physiologic and atlas maps can also be accom- Laitinen and Hariz, 1996).
plished through a number of commercially available
surgical navigation systems. 42.6. Stereotactic lesioning technique
42.5.1. Microelectrode localization 42.6.1. Radiofrequency

Microelectrodes for physiologic monitoring and One or two lesions are then made at the target defined
recording are designed to isolate single action poten- by the above techniques. Lesions are made by the
tials (Hubel, 1957; Jasper and Bertrand, 1966; Lenz technique of radiofrequency coagulation using an
et al., 1988a) and to withstand microstimulation which electrode with a 1.1 mm outer diameter and a 3 mm
degrades the electrode. Typically these characteristics are exposed tip and a thermister at the tip of the electrode
achieved by constructing electrodes from a platinum-iri- (TM electrode: Radionics, Burlington, MA). Tempera-
dium alloy or from tungsten, producing a tapered tip and ture is held constant at 60 C over a 1-minute interval.
insulating with glass (Hubel, 1957; Wolbarsht et al., The temperature is increased in 510 C steps during
1960; Umbach and Ehrhardt, 1965; Albe-Fessard, 1973; subsequent 1-minute intervals to a level of approxi-
Ohye, 1982; Lenz et al., 1988a). The electrode impedance mately 80 C. Since temperature is increased in steps
is usually (Lozano et al., 1996) greater than 500 kohms of about 10 C, the average length of time to make
(Jasper and Bertrand, 1966; Lenz et al., 1988a). A high- the lesion is 4 minutes. Neurologic examination stres-
impedance microelectrode is required to isolate single ses the function of structures adjacent to the target so
units (Lenz et al., 1988a). that during GPi coagulation motor strength and visual
The assembled electrode is attached to a hydraulic fields are stressed. In the case of Vim, the examination
or piezoelectric microdrive and mounted on the stresses cutaneous sensory, pyramidal and cerebellar
stereotaxic frame. Some microdrive systems incorpo- function, plus speech. These abbreviated exams should
rate a coarse drive so that overlying structures can be be carried out before, during and after each stage of
traversed quickly. The tip is then retracted into a lesion-making. The coagulum of each such separate
protective cylindrical housing while the whole assem- lesion made by this technique is approximated to a
bly is advanced to a new depth (Vitek et al., 1998a). cylinder with a diameter of 3 mm and a length of
The microdrive may then be employed from this new 5 mm (Cosman and Cosman, 1985; Lenz et al., 1995).
depth for detailed exploration of deeper structures.
Another option is to use the microdrive throughout 42.6.2. Localization of Vim
the trajectory by advancing it each time it reaches
the end of its traverse (Lenz et al., 1988a). Sensory cells responding to sensory stimulation in small,
The signal from the microelectrode is amplified and well-defined, receptive fields are found in the ventral
filtered. Multiple neuronal discharges of varying sizes caudal thalamic nucleus (Vc), posterior to Vim (Lenz
may be seen on an oscilloscope and heard by use of an et al., 1988c). There is a well-described mediolateral
audio monitor. In addition to recording, microstimula- somatotopy within Vc (Jasper and Bertrand, 1966; Lenz
tion of subcortical structures may be delivered. et al., 1988c) proceeding from representation of oral
We employ biphasic, square-wave pulse trains of structures medially to leg laterally. In anterior Vc and
0.10.3 ms pulses for up to 10 seconds at a frequency further anterior in Vim, neuronal firing is related to pas-
of 300 Hz (Lenz et al., 1993). The current used in sive joint movement (deep sensory cells) or to active
ABLATIVE SURGERY FOR THE TREATMENT OF PARKINSONS DISEASE 247
movement (voluntary cells) (Raeva, 1986; Lenz et al., Cells in GPe have narrower action potentials and fire
1990). Some neurons respond to both active movement either at high frequency (approximately 50 Hz) with
and to sensory stimulation such as joint movement intermittent pauses (pause cells) or at lower fre-
(combined cells) (Lenz et al., 1990, 1994). Deep quency (approximately 20 Hz) with intermittent
sensory, combined and voluntary cells have activity bursts (burst cells). Cells in GPi fire tonically at high
correlated with EMG activity during tremor (Lenz rates (6080 Hz). Cellular activity is also recorded
et al., 1985, 1988b; Hua et al., 1998a) and have a soma- during passive movements of upper and lower extre-
totopy parallel to that in Vc. Stimulation in Vc will mities both ipsilateral and contralateral to the record-
evoke somatic sensations (Lenz et al., 1993). Stimula- ing site to identify the cellular receptive field. Cells
tion in Vim may produce brief movements or alter in GPi of PD patients often respond to movement
ongoing tremor or dystonia (Lenz et al., 1999). of multiple joints.
Thalamic semimicroelectrode recordings (Ohye, At the target the optic tract is about 1 mm below
1982, 1998; Burchiel, 1995) reveal patterns of neuronal the lower border of GPi. The optic tract can be iden-
activity parallel to those of microelectrode recordings. tified by the hissing sound resulting from the firing of
Macrostimulation through a low-impedance electrode multiple myelinated axons which increases in
(impedance often less than 1000 kohms) can reliably response to a flashing light (visual-evoked poten-
identify Vim by alterations in the movement disorder tials). Microstimulation in the optic tract evokes mul-
(Tasker et al., 1982) and capsule by stimulation-evoked tiple colored sparkles of light. The internal capsule
tetanic contraction of skeletal muscle at low threshold behind GPi can be identified by the same hissing
(Ojemann and Ward, 1982; Tasker et al., 1982). sound plus microstimulation-evoked muscle twitches.
Stimulation of intralaminar nuclei medial to Vc or These data are used to generate a functional map of the
Vim may evoke the recruiting response long-latency, target zone in sagittal section. Two lesions are made
high-voltage, negative waves occurring over much of within the part of GPi where cells respond to move-
the cortex at the frequency of stimulation (usually less ments of the extremities (Lozano et al., 1996; Mandir
than 10 Hz) (Jones, 1985; Fisher et al., 1996). et al., 1997; Vitek et al., 1998a). Each lesion is made
An analysis of the locations of tremor cells suggests to 70 with the tip of the electrode 2 mm above the
that the optimal target for thalamotomy is located 2 optic tract and to 80 with the tip 34 mm above
mm anterior to Vc and 3 mm above the ACPC line the optic tract and in front of the internal capsule.
(Lenz et al., 1995). Alternatively, targets in thalamot- Alternately, the lateral extend to the target may be
omy have been placed anterior to the site at which estimated by lateral microelectrode trajectories and
evoked potentials can be recorded in response to cuta- multiple small lesions (13 per trajectory) can be used
neous stimulation of the fingers (Guiot et al., 1973; to fit the lesion to the contours of GPi (Bakay et al.,
Kelly et al., 1978; Fox et al., 1991). Lesions have been 1992; Vitek et al., 1998a).
made in the region where electrical stimulation The value of microelectrode-guided lesion location in
produces effects on tremor and anterior to the region pallidotomy has often been debated. In one report micro-
where electrical stimulation evokes sensations (Tasker electrode recording led to a change in final location for
et al., 1982). Lesions have also been made in the lesion placement in 14/20 (70%) cases (Hiner et al.,
region where cells respond to somatosensory stimula- 1997) with an average change of 3.5 mm. In another,
tion of muscle, joint and tendon and where electrical 75% of cases were within 3  1 mm of the final lesion
stimulation affects tremor (Ohye and Narabayashi, site, whereas 25% (4/10) were > 5 mm away (Azizi
1979). et al., 1996). Another study reported an incidence of
correction of the radiologic target by microelectrode
42.6.3. Localization of target within the internal recording which was too high to justify pallidotomy
segment of the globus pallidus without microelectrode recording (Taha et al., 1996).
Although these results suggest that microelectrode
During pallidotomy, microelectrode penetrations recording increases the accuracy of pallidotomy, this
toward the radiologic target are made from a coronal position has not been proven (Hariz, 2002).
burrhole about 25 mm from the midline. Striatum,
globus pallidus external segment (GPe) and GPi each 42.6.4. Localization of the subthalamic
have a characteristic pattern of neural activity (DeLong, nucleus lesions
1971; Lozano et al., 1996; Mandir et al., 1997; Vitek
et al., 1998a). Striatal cells are characterized by During the subthalamic explorations, trajectories are
broad action potentials and a slow firing rate made from a coronal burrhole about 25 mm from
(approximately 1 Hz), often with long silent periods. the midline. Striatum and Voa thalamus each have a
248 F. A. LENZ
characteristic pattern of neural activity (Hutchison For each procedure this review considered the
et al., 1998). As the electrode approaches the STN following subjects: (1) indications prior to the TTA-
from anterior and dorsal directions, striatal cells are AAN/MDS position papers; (2) randomized controlled
often encountered and are characterized by broad studies of the procedures; (3) studies included in the
action potentials and a slow firing rate (approximately position papers and then the final ATT-AAN/MDS
1 Hz), often with long silent periods. When the thala- recommendation.
mus is entered the action potentials become narrower
and often occur in bursts of the LTS type (see above).
42.7.1. General comments on all procedures
LTS bursts are preceded by a silent period of 20100
ms and consist of a interspike interval of less than
Across all procedures the TTA-AAN/MDS position
6 ms followed by interspike intervals of less than
papers made a number of general comments, as follows.
16 ms (Zirh et al., 1998; Ramcharan et al., 2000).
The publications included in these position papers were
A relatively acellular gap of 16 mm is observed
carried out by groups having neurosurgeons and neuro-
below the thalamus and above the STN, depending
physiologists with particular expertise in functional
upon the anteriorposterior location of the trajectory.
stereotactic procedures, as well as neurologists with
The STN is characterized by multiple spike trains
expertise in movement disorders. Inexperienced centers
recorded from closely packed neurons, each with a
had less success and more complications in all surgical
mean rate of about 20 ms. Microstimulation evokes
procedures. Cognitive impairment was a predictor of
paresthesias posterior to STN, muscle contractions
poor outcome and patients with advanced age derived
lateral to STN and decreases in tremor or tone within
less benefit. Contraindications were medical or psychia-
STN. The single lesion may be made dorsolateral to
tric conditions and abnormal imaging studies, including
STN (Patel et al., 2003) or two lesions may be made
focal lesions or atrophy greater than expected for age
within STN but located 3 mm apart in the medial
(Hallett and Litvan, 1999).
lateral plane (Lopez-Flores et al., 2003), with much
Many of the complications of ablative stereotactic
different results (see below).
surgery are common to all procedures. Complications
from lesion-making can arise from infection or intracra-
42.7. Results
nial hemorrhage. Hemorrhages comprise a significant
percentage of operative mortalities in stereotactic sur-
In this section we will consider the indications,
gery, occurring in 16% of procedures (Louw and
efficacy, complications/safety for pallidotomy, thala-
Burchiel, 1998). Hemorrhages may occur at the lesion
motomy and subthalamotomy. The analysis of efficacy
site or at cortical sites, resulting in intracerebral or sub-
and safety was based on the comprehensive assess-
dural hematomas. The risk of radiologically defined
ment of lesioning procedures for the treatment of PD.
hemorrhage during functional stereotactic procedures
This assessment by the Therapeutics and Technology
employing coagulation is 9/57 overall (16%; Vim 5/
Assessment Committee of the American Academy of
23: 22%; GPi 4/34: 12%) and the risk of symptomatic
Neurology (TTA-AAN) led to a position paper on
bleeding is 4/57 (7%) (Terao et al., 2003). This is
surgery for PD based on a systematic evaluation of
greater than the risk of deep brain stimulation (DBS)
the literature (Hallett and Litvan, 1999). This paper
surgery (3%: 2/59). Following stereotactic biopsy, radi-
was subsequently updated and endorsed in a scientific
ologically defined bleeds occurred in 40/500 (8%);
position paper by the Movement Disorder Society
delayed hemorrhages as defined by CT and symptoms
(MDS) (Hallett and Litvan, 2000).
occurred rarely, but always occurred within 48 hours
The literature searches were based on papers identi-
of the procedure (0.4%, 2/500) (Kondziolka et al.,
fied in four databases for the medical literature which
1998). Overall 12 patients were symptomatic (1.2%)
were searched for terms related to surgical treatment
and 1 died (0.2%). In that study platelet counts of less
of PD. All papers included in the analysis had to meet
than 150 000 were identified as a risk of bleeding in
the minimum standard of class III evidence as follows:
comparison with higher counts.
non-randomized, retrospective studies with historical
controls which were peer-reviewed, used validated
methods of assessment and provided consistent, clini- 42.7.2. Pallidotomy for Parkinsons disease
cal (rather than technical) data. Although most studies
considered were prospective, a few included a concur- At the time of rebirth of surgical approaches to the
rent control group, a requirement necessary to meet treatment of PD, pallidotomy was considered an
class II evidence, and fewer still were randomized, option for treatment of patients with advanced PD who
controlled studies. have motor drug-related complications, particularly
ABLATIVE SURGERY FOR THE TREATMENT OF PARKINSONS DISEASE 249
dyskinesias and fluctuations (Laitinen et al., 1992; Krack unchanged in the medical group. The UPDRS-2
et al., 2000). These symptoms should be disabling and activities of daily living subscale improved by 23%
unresponsive to optimal medical therapy if they are to in the surgical group and diminished by 16% in the
be considered as indications for pallidotomy. Patients medical group. All but 2 of the patients in the medical
with relatively less advanced disease but who are dis- arm crossed over but results were not reported. In
abled by tremor or dystonia are also candidates for these the surgical group of 19 there were 6 patients (32%)
procedures. with persistent complications, including: psychosis
and dysarthria/imbalance (one each) and 4 with minor
42.7.2.1. Randomized, controlled studies complications including dysarthria, facial weakness,
of pallidotomy urinary incontinence and mental status change.
There are two randomized, controlled studies of Therefore, controlled randomized trials have shown
pallidotomy for the treatment of PD (de Bie et al., consistent benefit for motor function as measured
1999; Vitek et al., 2003). In the most recent study 18 by the UPDRS motor scale. Complications in pallidot-
patients were randomized each to best medical therapy omy ranged from 17% to 57%, suggesting technical
and to unilateral pallidotomy (Vitek et al., 2003). differences between centers.
The change in the Unified Parkinsons Disease Rating
Scale (UPDRS) scale at 6 months demonstrated that 42.7.2.2. Prospective uncontrolled studies
the pallidotomy group had significant improvement A review combining results of unilateral pallidotomy
whereas the medical arm was slightly worse (overall examined all original studies of surgical outcome with
35% versus 5%, off motor 32% versus 3%). In the or without validated measures of clinical PD status or
surgical patients improvements were most significant a control group (Alkhani and Lozano, 2001). Those
in the symptoms of contralateral tremor, rigidity and studied using the validated UPDRS scores at 1 year
dyskinesia. Improvements in postural stability, gait and reported 45% improvement in off UPDRS motor sub-
bradykinesia were less significant (Vitek et al., 2003). scale and 35% improvement in the combined motor and
Improvements during the off periods were always activities of daily living subscales. In addition, there
larger than those in the on periods, which were often was a 41% improvement in the Schwab and England
insignificant. activities of daily living scale (Alkhani and Lozano,
Patients in the medical arm were given the option 2001). Contralateral on-period dyskinesias improved
of surgery at 6-month follow-up and all patients opted by 86%.
for surgery. Follow-up in 20 of the 36 patients was The beneficial effect on ipsilateral dyskinesias was
obtained at 2 years postoperative and showed highly lost by 1 year whereas the effect on gait and balance
significant (P < 0.00001) improvements in the UPDRS was lost by 36 months. Greater than 50% improve-
overall (21%), in the off motor subscale (26%) and in ments at 1 year were observed in tremor and rigidity
the drug-related complications subscale (fluctuations scales. This is consistent with another review of
and dyskinesias, 58%). Sustained improvements in tre- studies of unilateral pallidotomy which showed a
mor, rigidity, bradykinesia, percentage on time and 30% improvement in the UPDRS motor subscale and
drug-related dyskinesias were also observed. There in the UPDRS overall (Okun and Vitek, 2004).
was a highly significant relationship between UPDRS Levodopa-equivalent dose was not decreased post-
score at 2 years and age. Patients who were aged 50 operatively (Alkhani and Lozano, 2001; Okun and
or less had twice the reduction in the UPDRS score Vitek, 2004). Continued benefit has been documented
of those greater than 60 years of age. There were some up to 2 years postoperatively (Lang et al., 1997).
significant complications (6/36: 17%). Two patients Complications were common. Overall there was a
had seizures intraoperatively, delaying the completion hemorrhage rate of 1.7%, resulting in a mortality rate
of the procedure by weeks. Four patients had hemor- of 0.4% (Alkhani and Lozano, 2001). The incidence
rhages, 1 of whom had worsening of speech, which of visual field defects was variable, as a function of
resolved by 6 months postoperatively. institution, with permanent deficits in 1.5%. Persistent
Similar results were obtained in an earlier facial and limb weakness was observed in 1.3% and
randomized, controlled, single-blind, multicenter trial 0.9%, respectively. Persistent dysarthria, hypophonia
of pallidotomy versus best medical therapy (de Bie and dysphagia were reported in 1.6%, 1.3% and 0.5%,
et al., 1999). At 6 months the off condition of the respectively. Postoperative confusion, which was
UPDRS motor subscale improved (44%) significantly usually transient, occurred in about 10% of patients.
in the surgical group whereas it decreased in medical Overall, the morbidity rate was 23%, of which 14% were
controls (7%). The on-phase motor UPDRS permanent, as assessed by summing all complications
improved by 50% in the surgical group but was across and within patients and dividing by the total
250 F. A. LENZ
number of patients. Older patients were more likely to et al., 1998; Scott et al., 1998; Trepanier et al., 1998).
have cognitive problems and left-sided lesions were Unilateral pallidotomy was recommended as effective
associated with persistent verbal deficits (Saint-Cyr and safe based on class III evidence. Unilateral pallidot-
et al., 1996). omy was thought to be indicated for patients with PD
with fluctuations, bradykinesia, tremor and rigidity and
42.7.2.3. Bilateral pallidotomy particularly for those with dyskinesias. The benefit for
gait and postural disturbances was judged to be less pro-
A small, multicenter prospective study of staged
nounced. Based on the unreliable, contradictory data
bilateral pallidotomy controlled the second side
and reports of speech complications, bilateral pallidot-
against the first and found significant differences
omy was given a type D negative recommendation.
between the two pallidotomies (Parkin et al., 2002).
Three months after the first pallidotomy there was a
42.7.3. Thalamotomy for Parkinsons disease
significant decrease in the off UPDRS motor score
(mean 27%, P < 0.05) and dyskinesias were comple-
Studies published prior to the mid-1980s established
tely abolished in 40%. After the second side these
thalamotomy as an option for patients with PD who
figures were 31% and 63%. At 1 year after the second
are disabled by tremor and who have not responded
side the improvement in the motor scales diminished to medication, including levodopa-carbidopa and antic-
but the effect on activities of daily living scales and
holinergics (Narabayashi, 1982; Nagaseki et al., 1986).
dyskinesias was maintained. Persistent complications
Contraindications to thalamotomy include patients
were reported in 4%, much less than the average for
who have dementia, significant medical illness or Par-
unilateral pallidotomies (Saint-Cyr et al., 1996).
kinsons plus syndromes (e.g. ShyDrager, multisystem
An earlier retrospective study of 25 patients reported
atrophy).
efficacy for simultaneous, bilateral pallidotomy in the
treatment of bilateral symptoms and dyskinesias 42.7.3.1. Randomized, controlled trial of Vim
without surgical complications (Iacono et al., 1997). thalamotomy versus Vim-DBS
A prospective study compared the efficacy of the
A recent trial compared Vim thalamotomy versus DBS
second staged side of a bilateral pallidotomy with
in 68 patients with PT (n 45), as well as patients
the first side (de Bie et al., 2002). Clinical ratings after
with essential tremor (n 13) and intention tremor
the first surgery were significantly improved for the
(n 10) (Schuurman et al., 2000). Among patients
off UPDRS motor and activities of daily living scales
with parkinsonian tremor, abolition or slight residual
and the Schwab and England scales. There was a trend
tremor was seen post-Vim-DBS 21/21 (100%) and in
toward improvement in the first two of these scales
21/23 (93%) of patients postthalamotomy. Overall,
for the second as compared to the first procedure.
types of tremor functional scales (Schuling et al.,
Three out of 10 patients had persistent complications
1993) demonstrated a significantly greater increase in
following the first pallidotomy whereas 8/10 had such
functional ability for Vim-DBS (16%) versus thala-
complications after the second, including dysarthria
motomy (2%). Functional status was improved in sig-
(n 7), hypothonia (n 1), hemiplegia (n 1) and
nificantly more patients following Vim-DBS (54%,
visual field defect (n 1). Thus, the pallidotomy is
18/33) than thalamotomy (24%, 8/34). Overall, tremor
associated with an incremental improvement of
was abolished or minimal residual in 30/33 (90%) of
more than the improvement after the first side with a
patients treated with Vim-DBS and 27/34 (79%)
variable rate of complications.
of patients treated with thalamotomy (Schuurman
et al., 2000).
42.7.2.4. TTA-AAN/MDS recommendation The only death, secondary to intracerebral hema-
regarding pallidotomy toma, occurred in the Vim-DBS group. Nevertheless,
Forty studies of pallidotomy were found in the position complications overall were more common in the thala-
papers, of which approximately one-half met the cri- motomy group. In PT patients at 6 months postopera-
teria listed above (Laitinen et al., 1992; Dogali et al., tively cognitive deterioration was found in 1 patient
1995; Lozano et al., 1995; Baron et al., 1996; Fazzini postthalamotomy versus none post-Vim-DBS, mild
et al., 1997; Hariz and De Salles, 1997; Kazumata dysarthria in 3 versus 1, severe dysarthria in 3 versus
et al., 1997; Kishore et al., 1997; Krauss et al., 1997; 1, gait and balance disturbance was mild in 3 versus 0
Lang et al., 1997; Soukup et al., 1997; Uitti et al., and severe in 4 versus 0 and there was arm ataxia in
1997; Biousse et al., 1998; Cahn et al., 1998; Giller 1 versus 0. Overall, significantly more patients (16/
et al., 1998; Lozano et al., 1998; Masterman et al., 34) had complications postthalamotomy than patients
1998; Ondo et al., 1998; Shannon et al., 1998; Samuel following Vim-DBS stimulation (6/33).
ABLATIVE SURGERY FOR THE TREATMENT OF PARKINSONS DISEASE 251
42.7.3.2. Prospective, uncontrolled studies was demonstrated between blinded raters and tremor
of thalamotomy for Parkinsons disease severity scores were consistently better contralateral
Vim thalamotomies have been performed for PT over to surgery. This study suggests that thalamotomy can
five decades and advances in technology during this abolish tremor.
period have improved the accuracy and safety of Tasker (1998) has reported that tremor which was
this operation. CT or MR imaging as well as micro- completely abolished contralateral to a thalamic lesion
electrode recording has advanced the technical for 3 months never recurred. Such results have
standards of thalamotomy. Tasker (1990) reported that prompted some to propose that thalamotomy may
good results from thalamotomy increased from 45% actually alter the course of PD (Matsumoto et al.,
for surgeries performed before 1967 to 8696% for 1984). An investigation into this issue has revealed
surgeries carried out during the 1970s. Reports of three subgroups of PD in which the disease progresses
thalamotomies performed from 1980 to 1990 reveal so slowly that elimination of the major symptoms may
good results in 8694% of cases (Fox et al., 1991; appear to have altered the progression of the disease.
Jankovic et al., 1995). PD of the young-onset, postencephalitis type and a
One such study reported the results of Vim subset of patients with idiopathic parkinsonism all
thalamotomy performed between 1982 and 1994 in seem to have very slow progression of their parkinso-
42 patients with severe, asymmetric, medically intract- nian symptoms (Tasker, 1998). Thus elimination of
able parkinsonian tremor (Jankovic et al., 1995). Thala- tremor in these patients might give the appearance
motomies were guided by CT or ventriculographic of a long-term cure.
localization and microelectrode recording. Treating
physicians performed outcome assessments using the 42.7.3.3. Thalamotomy for dyskinesias
UPDRS and a global tremor-rating scale. Tremor was in Parkinsons disease
completely abolished in 72% (31/42) of cases, whereas
In addition to the reduction of tremor symptoms,
an additional 14% (6/42) showed significant improve-
several studies have shown that patients require less
ment in tremor and functional ability. None of the
or no levodopa after thalamotomy (Fox et al., 1991;
patients who were tremor-free after surgery experienced Jankovic et al., 1995). This decreased requirement
a recurrence of the tremor at long-term follow-up
for levodopa helps to reduce the drug-related compli-
(60158 months).
cations of PD (e.g. dyskinesias). Independent of this
Similar efficacy rates were reported in another study,
reduction of levodopa, thalamotomy has been shown
in which 37 Vim thalamotomies were performed between
to reduce levodopa-induced dyskinesias (Jankovic
1984 and 1989 for medically refractory parkinsonian
et al., 1995). Narabayashi et al. (1984) reported that,
tremor (Fox et al., 1991). CT localization and micro-
although levodopa dyskinesias are ameliorated by
electrode guidance were used. At the time of discharge,
Vim thalamotomy, the optimal site for a lesion to
complete abolition of contralateral tremor was seen relieve dyskinesias may be Vop, just anterior to Vim.
in 94% (34/36), which was maintained in 81% (13/16)
Such reduction in dyskinesia and increased tolerance
at 3 years. All recurrences occurred within 3 months
of levodopa is important for patients who have other
of surgery.
parkinsonian symptoms in addition to tremor, such as
In a third study ventriculography and macrostimula-
bradykinesia and rigidity (Poewe and Granata, 1997).
tion were used to localize the lesion site, which was
thought to be in Vop, a thalamic pallidal relay nucleus
(Wester and Hauglie-Hanssen, 1990), unlike the Vim 42.7.3.4. Complications of thalamotomy
target chosen in other studies reviewed here. Good to Neurological complications specific to thalamotomy
moderate improvement was achieved in 82% (27/33) can be explained by lesion-induced disruption of the
of patients with Parkinsons tremor, as assessed by cerebellothalamocortical pathway or damage to thala-
the referring neurologists. mic and nearby structures. In a series of 60 patients
One blinded, long-term assessment of the efficacy with essential, parkinsonian or cerebellar tremor,
of thalamotomy compared tremor in the arm contralat- functional deficits in the immediate postoperative
eral to thalamotomy versus the ipsilateral arm period were reported in 58% of patients (Jankovic
(Diederich et al., 1992). The potentially confounding et al., 1995). These transient deficits included weak-
effect of asymmetry in tremor was not considered. ness (34%), dysarthria (29%), ataxia (8%), dystonia
Thalamotomies were performed between 1976 and (5%) and sensory deficits (3%). Cognitive deficits
1985 and patients were followed for a mean of 10 were seen, including disorientation and somnolence,
years. A significant concordance for UPDRS score as well as speech and language deficits, including
252 F. A. LENZ
hypophonia. Functional deficits persisted in 23% but of subthalmotomy included patients with relatively
were generally mild and did not increase disability early PD characterized by hemiparkinsonism (off
(Jankovic et al., 1995). Hoehn and Yahr scale 3.1) (Patel et al., 2003). In this
In a series of 34 patients operated on for parkinso- study many patients were disabled by tremor but not
nian tremor, there were permanent complications in other parkinsonian symptoms, i.e. the tremor variant
14% (5), including apraxia (1), dysarthria (2), dyspha- of PD. In the other study subthalamotomy was carried
sia (1) and abulia (1) (Fox et al., 1991). Transient out in patients (n 18) with bilateral involvement
complications in 61% included cognitive decline (5), indicating more advanced disease, as reflected in the
central facial (10) or hand weakness (7) and hand off Hoehn and Yahr grade of III in 22% and of IV
numbness (2). Half of the transient deficits resolved in 78% (Alvarez et al., 2005).
by 1 week and most deficits, including transient and These two studies of subthalamotomy for PD were
permanent, had resolved at the 3-month follow-up also much different with respect to techniques and
visit. results. In the first, unilateral single lesions of the dor-
Parkinsonian patients in the study of Wester and solateral STN were made to 80 C for 60 seconds with
Hauglie-Hassen (1990) had mild complications, not an electrode having a 2 mm exposed tip. Postoperative
yielding invalidity, in 36% (12/33), including: mild MRIs demonstrated extension of the lesions dorsal to
mental status changes in 4, mild contralateral weak- STN, i.e. zona incerta, in 19 of 21 cases.
ness in 3, dysphasia in 4 and dysarthia in 1. Significant At 6 months there was an improvement in total
complications occurred in 18% (6/33), including: men- UPDRS of 22% off and 24% on, both of which
tal status change in 3, dysphasia in 1 and dysarthia in were maintained at 24 months, 18% and 11% respec-
2. Thus, significant or permanent side-effects were tively. There was a significant improvement in off
often observed in patients with PD, as in essential UPDRS activities of daily living scale at 6 (19%) but
and intention tremor. Therefore, the best available not 24 months. UPDRS III (motor) off was signifi-
evidence supports a rate of transient complications in cantly improved at 6 months contralateral to the lesion
approximately 60% of patients and persistent compli- (36%) and overall (19%) and at 24 months (16% con-
cations in the range of 1520%. tralateral and 38% overall). There were also significant
improvements in tremor, rigidity and bradykinesia in
42.7.3.5 TTA-AAN/MDS recommendation all patients followed for 12 and 24 months. Levodopa-
regarding thalamotomy equivalent doses were approximately halved post-
For thalamotomy, 18 articles were found in the TTA- operatively. Neuropsychological tests revealed mild
AAN study, but only the four studies cited above met cognitive deficits of verbal function observed on tests
the TTAS-AAN criteria (Wester and Hauglie-Hanssen, such as the Rey auditory verbal learning test. Intract-
1990; Fox et al., 1991; Diederich et al., 1992; Jankovic able dyskinesias occurred in 1 patient but responded
et al., 1995). Thalamotomy was recommended as to implantation of a DBS electrode into the zona
effective and safe for asymmetric, severe, medically incerta.
intractable tremor, particularly for the tremor variant The technique and results of the second study of
of PD. This was a positive recommendation based on subthalamotomy for PD (Alvarez et al., 2005) were
results of prospective studies with historical controls. much different from the first. Lesions were made to
It was judged to be possibly effective for the treatment a temperature of 70 C for 60 seconds with an electrode
of dyskinesias and rigidity. Thalamotomy was not having a 2 mm exposed tip. Two of these lesions were
thought to be effective for micrographia, bradykinesia made centered in the STN and located 3 mm apart in
or difficulties of gait or speech. Thalamotomy on the the mediallateral plane. UPDRS motor scores were
second side was felt to be effective for the treatment improved by 50% in the off state and 38% in the
of tremor but to be associated with a high incidence on state, measured at the time of last assessment, a
of speech and swallowing difficulty. Therefore a class minimum of 3 years postoperatively. Tremor, rigidity,
D negative recommendation was made for bilateral bradykinesia, activities of daily living and dyskinesias
thalamotomy; Vim-DBS was recommended on the were all significantly improved at the time of the last
second side. assessment.
Three patients had severe postoperative dyskinesias
42.7.4. Subthalamotomy which resolved over 36 months. Two of these
patients developed severe persistent gait instability.
There are no historical studies of subthalamotomy for Two of these and 1 other patient had severe persistent
the treatment of PD so that indications for surgery postoperative dysharthria. These 4 patients had lesion
were based upon general principles. The first study temperatures of 80 C for 1 minute and had large
ABLATIVE SURGERY FOR THE TREATMENT OF PARKINSONS DISEASE 253
lesions that were 3040% larger than the average for 110135 Gy and 140165 Gy groups. A good to
the rest of the population. Two of the patients with excellent result (increase of 23 UPDRS grades at three
severe dyskinesias and 1 other developed severe ataxia evaluations) was significantly more common in the
and broad-based gait which improved substantially high- (78%) than the low-dose group (56%), which
by 1 year. Severe ataxia occurred in a third patient was less than their success with standard stereotactic
who also had a larger than expected lesion. Thus there procedures. No complications were reported at mini-
were significant, persistent complications in 26% of mum follow-up of 5 months (median 28 months).
patients in this study. Another series of MRI-guided gamma-knife radiosur-
Therefore, both studies demonstrated significant gery reported complete to nearly complete relief of
long-term improvements at 2 years postoperatively. tremor in 88% of cases (n 27) (Young et al., 1998).
The first had relatively limited morbidity, whereas An older series of patients with intention, parkinsonian
the second had significant, persistent morbidity, per- and undefined tremor reported good outcome of MRI-
haps related to inclusion of patients with more guided gamma-knife thalamotomy in 66% of cases
advanced PD or to the bilateral, large lesions. Subtha- (n 9) (Rand et al., 1992). No complications were
lamotomy was not included in the TTC-AAC/MDS reported to occur in these latter two series.
review. The results reviewed here suggest that both Linear accelerator thalamotomy targeting the cen-
unilateral and bilateral subthalamotomy may be tral median and parafascicular nuclei for the treatment
effective procedures. The safety of these procedures of neuropathic and central pain has been reported in
is in doubt because of inconsistent reports of signifi- 3 cases (Frighetto et al., 2004). The lesion (75100 Gy)
cant persistent complications, possibly related to was made 4 mm anterior to PC and 10 mm lateral
surgical technique and the extent of the lesions. Since to the ACPC line using a 5 mm collimator along 58
there is only one report each for unilateral and bilateral non-coplanar arcs separated by 20 . Postoperatively
subthalamotomy, it is not possible at present to assess immediate pain relief was reported in all 3 cases based
the consistency of these results or to make a recom- on the assessment of the treating physician. Post-MRIs
mendation. showed lesions 7  8 mm in the posterior ventral medial
thalamus and 3.5  5 mm in the posterior ventral thala-
42.7.5. Radiosurgery for the treatment mus. No complications were reported.
of movement disorders In contrast to these series is a report of 9 complica-
tions encountered at Emory University Altanta, GA,
The use of MRI to provide radiologic localization has from a series of gamma-knife ablations for movement
led to the development of stereotactic radiosurgical disorders carried out at a nearby medical center (Okun
ablation. The majority of stereotactic gamma-knife et al., 2001, 2002). These complications occurred
procedures are carried out with an 50% isodose plan among an estimated total of 118 patients, including
(Kondziolka, 2002) using 4 mm collimators, some- PD in 96: 7 had the tremor-predominant variant and
times located on a secondary collimator helmet there was essential tremor in 22 (Okun et al., 2001,
(Young et al., 1998) and sometimes on the standard 201 2002). In this series, patients were found to have onset
cobalt source helmet. Maximal doses of 120160 Gy of benefit from the radiation never to 6 months and
are usually used (Niranjan et al., 1999), although doses onset of complications from 5 to 10 months. Compli-
of approximately 180 Gy (Friedman et al., 1996; Pan cations included weakness or paresis (n 3), visual
et al., 1996) or 200 Gy have also been reported (Friehs loss, speech/bulbar symptoms (n 3), dysphagia/
et al., 1997). Lesions with a volume of approximately aspiration pneumonia/death (n 1).
250 mm3 are created (Duma et al., 1998; Young et al., This report resulted in a debate centered on techni-
1998). The lesion placement is estimated from the que (Kondziolka, 2002), interpretation of the accuracy
usual location of Vim in relation to the AC and PC and of these lesions (De Salles et al., 2003) and the com-
the internal capsule (Duma et al., 1998; Young et al., plications of gamma-knife versus open procedures
1998). (Kondziolka, 2002). The conservative interpretation
In the largest of these series, MRI-guided gamma- of this debate is that gamma-knife (4 mm collimator,
knife procedures (4 mm collimator, 110165 Gy, 50% 12040 Gy, targeting with a 50% isodose line) is
isodose line at the medial edge of the internal capsule) indicated for thalamotomy, but not pallidotomy, in
were carried out in 34 patients with Parkinsons patients whose high surgical risk precludes a microelec-
tremor at high risk for standard stereotactic procedures trode-guided, radiofrequency procedure (Kondziolka,
(Duma et al., 1998). Patients and their physicians rated 2002). The need for a consensus and for prospective
the outcome using the UPDRS tremor scale. Retrospec- or controlled studies of gamma-knife movement
tively, the groups were stratified by radiation dose into disorder surgery is clear (Okun et al., 2002).
254 F. A. LENZ
42.7.6. Lesions versus stimulation for treatment of dyskinesias was significantly better in the subthala-
of Parkinsons disease mic stimulation group. Pallidotomy patients had
persistent complications in 9/14 (64%) whereas the
42.7.6.1. Pallidotomy
stimulation patients had complications in 8/20 (40%).
As a way of evaluating ablative versus stimulation pro- This study demonstrated significantly greater benefit
cedures we now compare the best data for ablative for bilateral subthalamic stimulation than unilateral
procedures with that for stimulation of the same ana- pallidotomy with numerous significant complications
tomic locus. In the two randomized, controlled trials of both procedures.
of pallidotomy for the treatment of PD there was a
significant improvement in off motor UPDRS score 42.7.6.2. Thalamotomy
of 3544% versus slight deterioration in the medical
The primary alternative to Vim thalamotomy for tremor
group (5% and 7%) at 6 months (de Bie et al.,
has been implantation of DBS electrodes into the VIM.
1999; Vitek et al., 2003). Results of the surgical group
The two have recently been compared in a recent trial
at 6 months were preserved at 2 years in the whole
of surgery for parkinsonian, essential and intention
cohort following cross-over (Vitek et al., 2003). Two
patients had seizures intraoperatively which delayed tremor (Schuurman et al., 2000). Among patients with
parkinsonian tremor (PT), abolition or slight residual
the pallidotomy. There were 4 hemorrhages, 1 with
tremor was seen post-Vim-DBS in 21/21 (100%) and
temporary worsening of speech, which resolved by 6
21/23 (93%) of patients postthalamotomy. Functional
months postoperatively (17%).
status was reported to be improved in significantly
Similar results were obtained in an earlier rando-
more patients following Vim-DBS (18/33, 54%) than
mized, controlled, single-blind, multicenter trial of
in patients with thalamotomy (24%, 8/34). Although
pallidotomy versus best medical therapy (de Bie
the only death occurred in the DBS group, there were
et al., 1999). At 6 months the off condition of the
UPDRS motor subscale improved (44%) significantly significantly more complications in the group under-
going thalamotomy (16/34, P < 0.05) than in those
in the surgical group whereas it decreased in medical
undergoing Vim-DBS (6/33). Thus Vim-DBS was safer
controls (7%). The on motor UPDRS and activities
and more effective than thalamotomy in PD.
of daily living subscale were both improved in com-
parison with the medical group. In the surgical group
of 19 there were 2 patients with major complications 42.7.6.3. Subthalamotomy
and 4 patients with minor but persistent side-effects The subthalamotomy study with bilateral surgery in
(32%). patients with more advanced disease is more compar-
Pallidal stimulation in patients with advanced PD able to a study of STN-DBS (Alvarez et al., 2005).
(n 38) in a prospective trial with a non-randomized UPDRS motor scores were improved by 50% in the
control group of patients with subthalamic stimulators off state at a minimum of 3 years postoperatively.
showed a UPDRS motor score improvement of 49% Tremor, rigidity, bradykinesia activities of daily living
and an increase in on time without dyskinesias from and dyskinesias were all significantly decreased at the
24 to 64% of the day at 3 months. In the total study time of the last assessment. Severe persistent complica-
(n 140) there were 7 hemorrhages (5%), 6 with tions, including imbalance, dyskinesias and ataxia,
neurologic deficits, of which 4 were persistent (3%). occurred in 28% of these patients. On the contrary, no
Thus the results of a controlled prospective study for severe persistent complications were encountered in a
GPi-DBS are similar to those of the randomized study of subthalamotomy in patients with less advanced
controlled study of pallidotomy with higher rates of PD, with unilateral lesions (Patel et al., 2003).
surgical complications in the pallidotomy group. A prospective, uncontrolled study of bilateral
A small randomized, controlled, multicenter trial of STN-DBS in patients with advanced PD with 5-year
unilateral pallidotomy versus bilateral STN stimula- follow-up (n 42) had UPDRS motor scale (54%)
tion has been reported; the latter is often considered and activities of daily living (49%) improvements in
to be the best surgical treatment for advanced PD. comparison with preoperative baseline (Krack et al.,
Patients had advanced PD (n 34, Hoehn and Yahr 2003). All subscales except speech were improved
> 4 in 20/34) (Esselink et al., 2004) and both surgical with respect to baseline. Levodopa equivalents were
groups were improved by off UPDRS motor scale; the decreased in comparison with baseline, as was the
primary outcome was variable. The improvement amount of drug-related dyskinesia. Surgical complica-
was less in the pallidotomy group (15%) than in the tions were limited to death secondary to an intracereb-
subthalamic stimulation group (37%). Dyskinesias ral hematoma and suicide (1 each). Thus the efficacy
improved in both groups; the duration but not severity is similar for STN-DBS and subthalamotomy.
ABLATIVE SURGERY FOR THE TREATMENT OF PARKINSONS DISEASE 255
However, the risks are higher for bilateral, but not uni-
Biousse V, Newman NJ, Carroll C et al. (1998). Visual fields
lateral, subthalamotomy. in patients with posterior GPi pallidotomy. Neurology 50:
Therefore, the efficacy is similar for ablative and 258265.
stimulation procedures in the subthalamus and palli- Burchiel KJ (1995). Thalamotomy for movement disorders.
dum. The efficacy of Vim-thalamotomy is less than In: PL Gildenberg, (Ed.), Neurosurgery Clinics of North
that for Vim-DBS. The rate of complications, both America. WB Saunders Company, Philadelphia, pp. 5571.
transient and permanent, is much higher for ablative Buzsaki G, Smith A, Berger S et al. (1990). Petit mal epilepsy
procedures. Thus, lesions of the subthalamus, thalamus and parkinsonian tremor: hypothesis of a common pace-
and pallidum are indicated when stimulation is maker. Neuroscience 36: 114.
contraindicated by the particular circumstances of an Cahn DA, Sullivan EV, Shear PK et al. (1998). Neuropsycho-
logical and motor functioning after unilateral anatomically
individual patient.
guided posterior ventral pallidotomy. Preoperative
performance and three-month follow-up. Neuropsychiatry
Acknowledgments Neuropsychol Behav Neurol 11: 136145.
Cardoso F, Jankovic J, Grossman RG et al. (1995). Outcome
Some of the studies described in this chapter were after stereotactic thalamotomy for dystonia and hemibal-
supported by grants to FAL from the National lismus. Neurosurgery 36: 501507.
Institutes of Health (RO1:NS38493, RO1:NS40059). Carlson JD, Iacono RP (1999). Electrophysiological versus
image-based targeting in the posteroventral pallidotomy.
Comput Aided Surg 4: 93100.
References
Carlson M, Carlson A (1990). Interactions between glutami-
nergic and monoaminergic systems within the basal
Adams RD, Victor M, Ropper AH (1996). Principles of Neu- ganglia. Trends Neurosci 13: 272276.
rology. McGraw-Hill, New York. Clatterbuck R, Lee J-I, Lenz FA (2000). Movement disorder
Albe-Fessard D (1973). Electrophysiological methods for the surgery: lesions or stimulation. Prog Neurol Surg 15:
identification of thalamic nuclei. Z Neurol 205: 1528. 227235.
Albin RL, Young AB, Penney JB (1989). The functional Cooper IS (1954). Intracerebral injection of procaine into the
anatomy of basal ganglia disorders. Trends Neurosci 12: globus pallidus in hyperkinetic disorders. Science 119:
366375. 417418.
Alexander E 3rd, Kooy HM, van Herk M et al. (1995). Mag- Cosman ER, Cosman BJ (1985). Methods of making nervous
netic resonance image-directed stereotactic neurosurgery: system lesions. In: RH Wilkins, SS Rengachary (Eds.),
use of image fusion with computerized tomography to Neurosurgery. McGraw-Hill, New York, pp. 24902499.
enhance spatial accuracy. J Neurosurg 83: 271276. de Bie RM, de Haan RJ, Nijssen PC et al. (1999). Unilateral
Alkhani A, Lozano AM (2001). Pallidotomy for Parkinson pallidotomy in Parkinsons disease: a randomised, single-
disease: a review of contemporary literature. J Neurosurg blind, multicentre trial. Lancet 354: 16651669.
94: 4349. de Bie RM, Schuurman PR, Esselink RA et al. (2002). Bilat-
Alvarez L, Macias R, Lopez G et al. (2005). Bilateral subtha- eral pallidotomy in Parkinsons disease: a retrospective
lamotomy in Parkinsons disease: initial and long-term study. Mov Disord 17: 533538.
response. Brain 128: 570583. De Salles AA, Frighetto L, Lacan G et al. (2003). Radiosur-
Andrew J, Fowler CJ, Harrison MJ (1983). Stereotaxic thala- gery can achieve precision needed for functional neurosur-
motomy in 55 cases of dystonia. Brain 106: 9811000. gery. Arch Neurol 60: 14941496.
Azizi A, Goldman WH, Moreledge D (1996). Posteroventral DeLong MR (1971). Activity of pallidal neurons during
pallidotomy: comparison of the accuracy of anatomical movement. J Neurophysiol 34: 414427.
targets defined by MR and CT imaging with physiological DeLong MR (1990). Primate models of movement disorders
targets defined by microelectrode recording. Neurology of basal ganglia origin. Trends Neurosci 13: 281285.
46 (Suppl): A199. Desmedt JE (1978). Progress in Clinical Neurophysiology.
Bakay RA, DeLong MR, Vitek JL (1992). Posteroventral palli- Cerebral Motor Control in Man: Long Loop Mechanisms
dotomy for Parkinsons disease. J Neurosurg 77: 487488. Karger, Basel.
Baron MS, Vitek JL, Bakay RA et al. (1996). Treatment of Diederich N, Goetz CG, Stebbins GT et al. (1992). Blinded
advanced Parkinsons disease by posterior GPi pallidotomy: evaluation confirms long-term assymmetric effect of
1-year results of a pilot study. Ann Neurol 40: 355366. unilateral thalamotomy or subthalamotomy on tremor in
Benabid AL, Pollak P, Gao D et al. (1996). Chronic electrical Parkinsons disease. Neurology 42: 13111314.
stimulation of the ventralis intermedius nucleus of the tha- Dogali M, Fazzini E, Kolodny E et al. (1995). Stereotactic
lamus as a treatment of movement disorders. J Neurosurg ventral pallidotomy for Parkinsons disease. Neurology
84: 203214. 45: 753761.
Bergman H, Wichmann T, DeLong MR (1990). Reversal of Dostrovsky JO (1998). The use of inexpensive personal
experimental parkinsonism by lesions of the subthalamic computers for map generation and data analysis. In: PL
nucleus. Science 249: 14361438. Gildenberg, RR Tasker (Eds.), Textbook of Stereotactic
256 F. A. LENZ

and Functional Neurosurgery. McGraw-Hill, New York, Hallett M, Litvan I (1999). Evaluation of surgery for Parkin-
pp. 20312036. sons disease: a report of the Therapeutics and Technology
Duma CM, Jacques DB, Kopyov OV et al. (1998). Gamma Assessment Subcommittee of the American Academy of
knife radiosurgery for thalamotomy in parkinsonian tre- Neurology. The Task Force on Surgery for Parkinsons
mor: a five year experience. J Neurosurg 88: 10441049. Disease. Neurology 53: 19101921.
Elble RJ, Koller W (1990). Tremor Johns Hopkins Univer- Hallett M, Litvan I (2000). Scientific position paper of the
sity Press, Baltimore. Movement Disorder Society evaluation of surgery for
Esselink RA, de Bie RM, de Haan RJ et al. (2004). Unilateral Parkinsons disease. Task Force on Surgery for Parkin-
pallidotomy versus bilateral subthalamic nucleus stimula- sons Disease of the American Academy of Neurology
tion in PD: a randomized trial. Neurology 62: 201207. Therapeutic and Technology Assessment Committee.
Fazzini E, Dogali M, Sterio D et al. (1997). Stereotactic pal- Mov Disord 15: 436438.
lidotomy for Parkinsons disease: a long-term follow-up of Hardy PA, Barnett GH (1998). Spatial distortion in magnetic
unilateral pallidotomy. Neurology 48: 12731277. resonance imaging: impact on stereotactic localization.
Filion M, Tremblay L, Bedard PJ (1988). Abnormal influences In: PL Gildenberg, RR Tasker (Eds.), Textbook of Stereo-
of passive limb movement on the activity of globus pallidus tactic and Functional Neurosurgery. McGraw-Hill, New
neurons in parkinsonian monkeys. Brain Res 444: 165176. York, pp. 271280.
Fisher RS, Uematsu S, Krauss GL et al. (1996). Placebo- Hariz MI (2002). Safety and risk of microelectrode recording
controlled pilot study of centromedian thalamic stimula- in surgery for movement disorders. Stereotact Funct
tion in treatment of intractable seizures. Epilepsia 33 (5): Neurosurg 78: 146157.
841851. Hariz MI, De Salles AA (1997). The side-effects and compli-
Fox MW, Ahlskog EJ, Kelly PJ (1991). Stereotactic ventro- cations of posteroventral pallidotomy. Acta Neurochir
lateralis thalamotomy for medically refactory tremor in Suppl (Wien) 68: 4248.
post-levodopa era Parkinsons disease patients. J Neuro- Hassler R, Mundinger F, Reichert T (1979). Stereotaxis in
surg 75: 723730. the Parkinson Syndrome. Springer Verlag, Berlin.
Friedman JH, Epstein M, Sanes JN et al. (1996). Gamma Hiner B, Madden K, Neal J (1997). Effect of microelectrode
knife pallidotomy in advanced Parkinsons disease. Ann recording on final lesion placement in pallidotomy. Neu-
Neurol 39: 535538. rology 48 (Suppl 2): A251.
Friehs GM, Ojakangas CL, Schrottner O et al. (1997). Holtzheimer PE 3rd, Roberts DW, Darcey TM (1999). Mag-
Radiosurgical lesioning of the caudate nucleus as a treat- netic resonance imaging versus computed tomography for
ment for parkinsonism: a preliminary report. Neurol Res target localization in functional stereotactic neurosurgery.
19: 97103. Neurosurgery 45: 290297.
Frighetto L, De Salles A, Wallace R et al. (2004). Linear Hornykiewicz D (1974). The mechanisms of action of
accelerator thalamotomy. Surg Neurol 62: 106113. L-dopa in Parkinsons disease. Life Sci 15 (7): 12491259.
Garonzik IM, Hua SE, Ohara S et al. (2002). Intraoperative Hua SE, Lenz FA, Zirh TA et al. (1998a). Thalamic neuronal
microelectrode and semi-microelectrode recording during activity correlated with essential tremor. J Neurol Neuro-
the physiological localization of the thalamic nucleus ven- surg Psychiatry 64: 273276.
tral intermediate. Mov Disord 17 (Suppl 3): S135S144. Hua S, Reich SG, Zirh AT et al. (1998b). The role of the tha-
Gerdes JS, Hitchon PW, Neerangun W et al. (1994). Com- lamus and basal ganglia in parkinsonian tremor. Mov
puted tomography versus magnetic resonance imaging in Disord 13 (Suppl 3): 4042.
stereotactic localization. Stereotact Funct Neurosurg 63: Hua SE, Garonzik IM, Lee J-I et al. (2004). Thalamotomy
124129. for tremor. In: RH Winn (Ed.), Youmans Neurological
Giller CA, Dewey RB, Ginsburg MI et al. (1998). Stereotactic Surgery. Saunders, Philadelphia, pp. 27692784.
pallidotomy and thalamotomy using individual variations of Hubel DH (1957). Tungsten microelectrode for recording
anatomic landmarks for localization. Neurosurgery 42: from single units. Science 125: 549550.
5662. Hutchison WD, Levy R, Dostrovsky JO et al. (1997a). Effects
Guiot G, Hardy J, Albe-Fessard D (1962). Delimitation pre- of apomorphine on globus pallidus neurons in parkinsonian
cis des structures sous-corticales et identification de patients. Ann Neurol 42: 767775.
noyaux thalamiques chez lhomme par lelectrophyiologie Hutchison WD, Lozano AM, Tasker RR et al. (1997b).
stereotactic. Neurochirurgia 5: 118. Identification and characterization of neurons with
Guiot G, Derome P, Arfel G et al. (1973). Electrophysiological tremor-frequency activity in human globus pallidus. Exp
recordings in stereotaxic thalamotomy for parkinsonism. Brain Res 113: 557563.
In: H Krayenbuhl, PE Maspes, WH Sweet (Eds.), Progress Hutchison WD, Allan RJ, Opitz H et al. (1998). Neuro-
in Neurological Surgery. Karger, Basel, pp. 189221. physiological identification of the subthalamic nucleus
Guridi J, Gorospe A, Ramos E et al. (1999). Stereotactic in surgery for Parkinsons disease. Ann Neurol 44:
targeting of the globus pallidus internus in Parkinsons 622628.
disease: imaging versus electrophysiological mapping. Iacono RP, Carlson JD, Kuniyoshi S et al. (1997). Contem-
Neurosurgery 45: 278287. poraneous bilateral pallidotomy. Neurosurg Focus 2: e5.
ABLATIVE SURGERY FOR THE TREATMENT OF PARKINSONS DISEASE 257

Jankovic J, Cardoso F, Grossman RG et al. (1995). Outcome Lemstra AW, Verhagen ML, Lee JI et al. (1999). Tremor-
after stereotactic thalamotomy for parkinsonian, essential frequency (36 Hz) activity in the sensorimotor arm repre-
and other types of tremor. Neurosurgery 37: 680686. sentation of the internal segment of the globus pallidus in
Jasper HH, Bertrand G (1966). Thalamic units involved in patients with Parkinsons disease. Neurosci Lett 267:
somatic sensation and voluntary and involuntary move- 129132.
ments in man. In: DP Purpura, MD Yahr (Eds.), The Thala- Lenz FA, Tatton WG, Tasker RR (1983a). Electromyographic
mus. Columbia University Press, New York, pp. 365390. response to displacement of different forelimb joints in the
Jones EG (1985). The Thalamus. Plenum, New York. squirrel monkey. J Neurosci 3: 783794.
Kazumata K, Antonini A, Dhawan V et al. (1997). Preopera- Lenz FA, Tatton WG, Tasker RR (1983b). The effect of cor-
tive indicators of clinical outcome following stereotaxic tical lesions on the electromyographic response to joint
pallidotomy. Neurology 49: 10831090. displacement in the squirrel monkey forelimb. J Neurosci
Kelly PJ, Derome P, Guiot G (1978). Thalamic spatial varia- 3: 795805.
bility and the surgical results of lesions placed with neuro- Lenz FA, Tasker RR, Kwan HC et al. (1985). Cross-correla-
physiologic control. Surg Neurol 9: 307315. tion analysis of thalamic neurons and EMG activity in
Kelly PJ, Ahlskog JE, Goerss SJ et al. (1987). Computer- parkinsonian tremor. Appl Neurophysiol 48: 305308.
assisted stereotactic ventralis lateralis thalamotomy with Lenz FA, Dostrovsky JO, Kwan HC et al. (1988a). Methods
microelectrode recording control in patients with Parkin- for microstimulation and recording of single neurons and
sons disease. Mayo Clin Proc 62: 655664. evoked potentials in the human central nervous system.
Kishore A, Turnbull IM, Snow BJ et al. (1997). Efficacy, J Neurosurg 68: 630634.
stability and predictors of outcome of pallidotomy for Lenz FA, Tasker RR, Kwan HC et al. (1988b). Single unit
Parkinsons disease. Six-month follow-up with additional analysis of the human ventral thalamic nuclear group: corre-
1-year observations. Brain 120 (Pt 5): 729737. lation of thalamic tremor cells with the 36 Hz component
Kondziolka D (2002). Gamma knife thalamotomy for of parkinsonian tremor. J Neurosci 8: 754764.
disabling tremor. Arch Neurol 59: 16601664. Lenz FA, Dostrovsky JO, Tasker RR et al. (1988c). Single-
Kondziolka D, Dempsey PK, Lunsford LD et al. (1992). unit analysis of the human ventral thalamic nuclear group:
A comparison between magnetic resonance imaging and somatosensory responses. J Neurophysiol 59: 299316.
computed tomography for stereotactic coordinate determi- Lenz FA, Kwan H, Dostrovsky JO et al. (1990). Single unit
nation. Neurosurgery 30: 402407. analysis of the human ventral thalamic nuclear group.
Kondziolka D, Firlik AD, Lunsford LD (1998). Complica- Activity correlated with movement. Brain 113: 17951821.
tions of stereotactic brain surgery. Neurol Clin 16: 3554. Lenz FA, Seike M, Richardson RT et al. (1993). Thermal
Krack P, Poepping M, Weinert D et al. (2000). Thalamic, and pain sensations evoked by microstimulation in the
pallidal, or subthalamic surgery for Parkinsons disease? area of human ventrocaudal nucleus. J Neurophysiol 70:
J Neurol 247 (Suppl 2): II122II134. 200212.
Krack P, Batir A, Van Blercom N et al. (2003). Five-year Lenz FA, Kwan HC, Martin RL et al. (1994). Single unit
follow-up of bilateral stimulation of the subthalamic analysis of the human ventral thalamic nuclear group. Tre-
nucleus in advanced Parkinsons disease. N Engl J Med mor-related activity in functionally identified cells. Brain
349: 19251934. 117: 531543.
Krauss JK, Desaloms JM, Lai EC et al. (1997). Microelec- Lenz FA, Normand SL, Kwan HC et al. (1995). Statistical
trode-guided posteroventral pallidotomy for treatment of prediction of the optimal site for thalamotomy in parkinso-
Parkinsons disease: postoperative magnetic resonance nian tremor. Mov Disord 10: 318328.
imaging analysis. J Neurosurg 87: 358367. Lenz FA, Suarez JL, Verhagen Metman L et al. (1998). Palli-
Laitinen LV, Hariz MI (1996). Movement disorders. In: dal activity during dystonia: somatosensory reorganization
JR Youmans (Ed.), Neurological Surgery. WB Saunders and changes with severity. J Neurol Neurosurg Psychiatry
Company, Philadelphia, pp. 35753609. 65: 767770.
Laitinen LV, Bergenheim AT, Hariz MI (1992). Leksells Lenz FA, Jaeger CJ, Seike MS et al. (1999). Thalamic single
posteroventral pallidotomy in the treatment of Parkinsons neuron activity in patients with dystonia: dystonia-related
disease. J Neurosurg 76: 5361. activity and somatic sensory reorganization. J Neurophy-
Lamarre Y, Joffroy A (1970). Thalamic unit activity in monkey siol 82: 23722392.
with experimental tremor. In: A Barbeau, FH McDowell Levy R (1992). A Short History of Stereotactic Neurosurgery.
(Eds.), L-Dopa and Parkinsonism. Davis, Philadelphia, pp. American Association of Neurological Surgeons. Park
163170. Ridge, IL. Ref Type: Serial (Book, Monograph).
Lamarre Y, Joffroy AJ (1979). Experimental tremor in mon- Lopez-Flores G, Miguel-Morales J, Teijeiro-Amador J et al.
key: activity of thalamic and precentral cortical neurons in (2003). Anatomic and neurophysiological methods for
the absence of peripheral feedback. Adv Neurol 24: the targeting and lesioning of the subthalamic nucleus:
109122. cuban experience and review. Neurosurgery 52: 817830.
Lang AE, Lozano AM, Montgomery E et al. (1997). Postero- Louw DF, Burchiel KJ (1998). Ablative therapy for movement
ventral medial pallidotomy in advanced Parkinsons disorders. Complications in the treatment of movement dis-
disease. N Engl J Med 337: 10361042. orders. Neurosurg Clin N Am 9: 367373.
258 F. A. LENZ

Lozano AM, Lang AE, Galvez-Jimenez N et al. (1995). Ohye C (1998). Neural noise recording in functional neurosur-
Effect of GPi pallidotomy on motor function in Parkin- gery. In: PL Gildenberg, RR Tasker (Eds.), Textbook of
sons disease. Lancet 346: 13831387. Stereotactic and Functional Neurosurgery. McGraw-Hill,
Lozano AM, Hutchison W, Kiss Z et al. (1996). Methods for New York, pp. 941948.
microelectrode guided posteroventral pallidotomy. J Neuro- Ohye C, Narabayashi H (1979). Physiological study of
surg 84: 194202. presumed ventralis intermedius neurons in the human
Lozano AM, Lang AE, Hutchison WD (1998). Pallidotomy thalamus. J Neurosurg 50: 290297.
for tremor. Mov Disord 13 (13 Suppl 3): 107110. Ojemann GA, Ward AA Jr (1982). Abnormal movement
Magnin M, Morel A, Jeanmonod D (2000). Single-unit ana- disorders. In: JR Youmans, (Ed.), Neurological Surgery.
lysis of the pallidum, thalamus and subthalamic nucleus in WB Saunders Company, Philadelphia, pp. 38213857.
parkinsonian patients. Neuroscience 96: 549564. Okun MS, Vitek JL (2004). Lesion therapy for Parkinsons
Mandir AS, Rowland LH, Dougherty PM et al. (1997). disease and other movement disorders: update and contro-
Microelectrode recording and stimulation techniques during versies. Mov Disord 19: 375389.
stereotactic procedures in the thalamus and pallidum. Adv Okun MS, Stover NP, Subramanian T et al. (2001). Compli-
Neurol 74: 159165. cations of gamma knife surgery for Parkinson disease.
Marsden CD (1984). Origins of normal and pathological tre- Arch Neurol 58: 19952002.
mor. In: LJ Findley, R Capildeo (Eds.), Movement Disor- Okun MS, Vitek JL, DeLong MR (2002). Not our knife:
ders: Tremor. Macmillan Press Ltd., London, pp. 3785. gamma knife surgery for Parkinson disease. Arch Neurol
Marsden CD, Fahn S (1987). Problems in the Dyskinesias. 59: 13341335.
In: CD Marsden, S Fahn (Eds.), Movement Disorders II. Ondo WG, Jankovic J, Lai EC et al. (1998). Assessment of
Butterworths, London. motor function after stereotactic pallidotomy. Neurology
Marsden CD, Parkes JD (1977). Success and problems of 50: 266270.
long-term levodopa therapy in Parkinsons disease. Lancet Pan L, Dai JZ, Wang BJ et al. (1996). Stereotactic Gamma
1: 345349. thalamotomy for the treatment of parkinsonism. Stereotact
Masterman D, DeSalles A, Baloh RW et al. (1998). Motor, Funct Neurosurg 66 (Suppl 1): 329332.
cognitive, and behavioral performance following unilat- Pare D, Curro Dossi R, Steriade M (1990). Neuronal basis
eral ventroposterior pallidotomy for Parkinson disease. of the parkinsonian resting tremor: a hypothesis and its
Arch Neurol 55: 12011208. implications for treatment. Neuroscience 35: 217226.
Matsumoto K, Shichijo F, Fukami T (1984). Long-term Parkin SG, Gregory RP, Scott R et al. (2002). Unilateral and
follow-up review of cases of Parkinsons disease after uni- bilateral pallidotomy for idiopathic Parkinsons disease: a
lateral or bilateral thalamotomy. J Neurosurg 53: 332337. case series of 115 patients. Mov Disord 17: 682692.
Merello M, Balej J, Delfino M et al. (1999). Apomorphine Patel NK, Heywood P, OSullivan K et al. (2003). Unilateral
induces changes in GPi spontaneous outflow in patients subthalamotomy in the treatment of Parkinsons disease.
with Parkinsons disease. Mov Disord 14: 4549. Brain 126: 11361145.
Nagaseki Y, Shibazaki T, Hirai T et al. (1986). Long-term Paulson HL, Stern MB (1997). Clinical manifestations of Par-
follow-up results of selective VIM-thalamotomy. J Neuro- kinsons disease. In: RL Watts, WC Koller (Eds.), Move-
surg 65: 296302. ment Disorders. McGraw Hill, New York, pp. 183199.
Narabayashi H (1982). Surgical approach to tremor. In: Penney JB Jr, Young AB (1981). GABA as the Pallidothala-
CD Marsden, S Fahn (Eds.), Movement Disorders. Butter- mic neurotransmitter: implications for basal ganglia func-
worth Scientific, London, Boston, Sydney, Wellington, tion. Brain Res 207: 195199.
Durban, Toronto, pp. 292299. Poewe W, Granata R (1997). Pharmacologic treatment of Par-
Narabayashi H, Yokochi F, Nakajima Y (1984). Levodopa- kinsons disease. In: RL Watts, WC Koller (Eds.), Move-
induced dyskinesia and thalamotomy. J Neurol Neurosurg ment Disorders. McGraw-Hill, New York, pp. 201220.
Psychiatry 47: 831839. Raeva S (1986). Localization in human thalamus of units
Nini A, Feingold A, Slovin H et al. (1995). Neurons in the glo- triggered during verbal commands, voluntary move-
bus pallidus do not show correlated activity in the normal ments and tremor. Electroencephalogr Clin Neurophysiol
monkey, but phase-locked oscillations appear in the MPTP 63: 160173.
model of parkinsonism. J Neurophysiol 74: 18001805. Ramcharan EJ, Gnadt JW, Sherman SM (2000). Burst and
Niranjan A, Jawahar A, Kondziolka D et al. (1999). A compar- tonic firing in thalamic cells of unanesthetized, behaving
ison of surgical approaches for the management of tremor: monkeys. Vis Neurosci 17: 5562.
radiofrequency thalamotomy, gamma knife thalamotomy Ranck JB (1975). Which elements are excited in electrical
and thalamic stimulation. Stereotact Funct Neurosurg 72: stimulation of mammalian central nervous system: a
178184. review. Brain Res 98: 417440.
Ohye C (1982). Depth microelectrode studies. In: G Schal- Rand RW, Jacques DB, Melbye RW et al. (1992). Gamma
tenbrand, AE Walker (Eds.), Stereotaxy of the Human knife thalamotomy and pallidotomy in patients with move-
Brain. Anatomical, Physiological and Clinical Applica- ment disorders: preliminary results. Stereotact Funct Neu-
tions. Georg Thieme, Stuttgart, pp. 372389. rosurg 61 (Suppl 1): 6592.
ABLATIVE SURGERY FOR THE TREATMENT OF PARKINSONS DISEASE 259

Raz A, Vaadia E, Bergaman H (2000). Firing patterns and Strick PL (1976). Activity of ventrolateral thalamic neurons
correlations of spontaneous discharge of pallidal neurons during arm movement. J Neurophysiol 39: 10321044.
in the normal and the tremulous 1-methyl-4-phenyl- Suarez JL, Verhagen Metman L, Reich SG et al. (1997). Pal-
1,2,3,6-tetrahydropyridine vervet model of parkinsonism. lidotomy for hemiballismus: efficacy and characteristics of
J Neurosci 20: 85598571. neuronal activity. Ann Neurol 42: 807811.
Saint-Cyr J, Trepanier L, Lang A (1996). Neuropsychological Taha JM, Favre J, Burchiel KJ (1996). The value of macro-
outcome of posteroventral pallidotomy in parkinsonian stimulation in patients who underwent microrecording
patients. Mov Disord 11: 161. during pallidotomy. Congress of Neurological Surgeons
Samuel M, Caputo E, Brooks DJ et al. (1998). A study of Abstract at 46th Annual meeting, Chicago, IL, October
medial pallidotomy for Parkinsons disease: clinical out- 5, 1994, pp. 264265. Ref Type: Abstract.
come, MRI location and complications. Brain 121 (Pt 1): Taren J, Guiot G, Derome P et al. (1968). Hazards of stereo-
5975. taxic thalamectomy. Added safety factor in corroborating
Schaltenbrand G, Bailey P (1959). Introduction to Stereotaxis X-ray target localization with neurophysiological methods.
with an Atlas of the Human Brain. Thieme, Stuttgart. J Neurosurg 29: 173182.
Schnider SM, Kwong RH, Lenz FA et al. (1989). Detection Tasker RR (1990). Thalamotomy. Neurosurg Clin N Am 1:
of feedback in the central nervous system using system 841864.
identification techniques. Biol Cybern 60: 203212. Tasker RR (1998). Ablative therapy for movement disorders.
Schuling J, de Haan R, Limburg M et al. (1993). The Does thalamotomy alter the course of Parkinsons disease.
Frenchay Activities Index. Assessment of functional status Neurosurg Clin N Am 9: 375380.
in stroke patients. Stroke 24: 11731177. Tasker RR, Organ LW, Hawrylyshyn P (1982). The Thala-
Schuurman PR, Bosch DA, Bossuyt PM et al. (2000). mus and Midbrain in Man: a Physiologic Atlas Using
A comparison of continuous thalamic stimulation and Electrical Stimulation. Thomas, Springfield, Illinous.
thalamotomy for suppression of severe tremor. N Engl J Tasker RR, Doorly T, Yamashiro K (1988). Thalamotomy in
Med 342: 461468. generalized dystonia. Adv Neurol 50: 615631.
Scott R, Gregory R, Hines N et al. (1998). Neuropsychological, Tasker RR, Dostovsky JO, Dolan EJ (1991). Computerized
neurological and functional outcome following pallidotomy tomography (CT) is just as accurate as ventriculography
for Parkinsons disease. A consecutive series of eight simul- for functional stereotactic thalamotomy. Stereotact Funct
taneous bilateral and twelve unilateral procedures. Brain Neurosurg 57: 157166.
121 (Pt 4): 659675. Terao T, Takahashi H, Yokochi F et al. (2003). Hemorrhagic
Shannon KM, Penn RD, Kroin JS et al. (1998). Stereotactic complication of stereotactic surgery in patients with
pallidotomy for the treatment of Parkinsons disease. movement disorders. J Neurosurg 98: 12411246.
Efficacy and adverse effects at 6 months in 26 patients. Trepanier LL, Saint-Cyr JA, Lozano AM et al. (1998).
Neurology 50: 434438. Neuropsychological consequences of posteroventral
Siegfried J, Lippitz B (1994). Chronic electrical stimulation pallidotomy for the treatment of Parkinsons disease.
of the VL-VPL complex and of the pallidum in the treat- Neurology 51: 207215.
ment of movement disorders: personal experience since Uitti RJ, Wharen RE Jr, Turk MF et al. (1997). Unilateral
1982. Stereotact Funct Neurosurg 62: 7175. pallidotomy for Parkinsons disease: comparison of outcome
Soukup VM, Ingram F, Schiess MC et al. (1997). Cognitive in younger versus elderly patients. Neurology 49: 10721077.
sequelae of unilateral posteroventral pallidotomy. Arch Umbach W, Ehrhardt KJ (1965). Micro-electrode recording
Neurol 54: 947950. in the basal ganglia during stereotaxic operations. Confin
Spiegel EA, Wycis HT (1954). Ansotomy in paralysis agi- Neurol 26: 315317.
tans. AMA Arch Neurol Psychiatry 71: 598614. Vitek JL, Lenz FA (1998). Contemporary pallidotomy for
Spiegel EA, Wycis HT, Baird HW 3rd (1958). Long-range treatment of dystonia and other movement disorders. In:
effects of electropallidoansotomy in extrapyramidal and J Jankovic, RG Grossman, G Krauss (Eds.), Pallidotomy
convulsive disorders. Neurology 8: 734740. for Treatment of Parkinsons Disease and Other Movement
Starr PA, Vitek JL, DeLong M et al. (1999). Magnetic reso- Disorders. Lippincott-Raven, New York, pp. 267274.
nance imaging-based stereotactic localization of the glo- Vitek JL, Kaneoke Y, Turner R et al. (1993). Neuronal activ-
bus pallidus and subthalamic nucleus. Neurosurgery 44: ity in the internal (GPi) and external (GPe) segments of
303313. the globus pallidus (GP) of parkinsonian patients is similar
Starr PA, Christine CW, Theodosopoulos PV et al. (2002). to that in the MPTP-treated primate model of parkinson-
Implantation of deep brain stimulators into the sub- ism. Society for Neuroscience Abstract 19: 1584. Ref
thalamic nucleus: technical approach and magnetic reso- Type: Abstract.
nance imaging-verified lead locations. J Neurosurg 97: Vitek JL, Ashe J, DeLong MR et al. (1994). Physiologic
370387. properties and somatotopic organization of the primate
Stein RB, Lee RG (1981). Tremor and clonus. In: VB motor thalamus. J Neurophysiol 71: 14981513.
Brooks (Ed.), Handbook of Physiology. Section I: Ner- Vitek JL, Kaneoke Y, Hashimoto T et al. (1995). Neuronal
vous System. Volume II: Motor Control Part II. American activity in the pallidum of a patient with hemiballismus.
Physiological Society, Bethesda, pp. 325343. Neurology 38: 296. Ref Type: Abstract.
260 F. A. LENZ

Vitek JL, Bakay RA, Hashimoto T et al. (1998a). Microelectrode- Wester K, Hauglie-Hanssen E (1990). Stereotaxic thalamot-
guided pallidotomy: technical approach and its application in omy experiences from the levodopa era. J Neurol Neuro-
medically intractable Parkinsons disease. J Neurosurg 88: surg Psychiatry 53: 427430.
10271043. Wolbarsht ML, MacNichol EF Jr, Wagner HG (1960). Glass
Vitek JL, Zhang J, Evatt M et al. (1998b). GPi pallidotomy insulated platinum microelectrode. Science 132: 13091310.
for dystonia: clinical outcome and neuronal activity, Adv Young AB, Penney JB (1984). Neurochemical anatomy of
Neurol 78: 211219. movement disorders. Neurol Clin 2: 417433.
Vitek JL, Chockkan V, Zhang J-Y et al. (1999). Neuronal Young RF, Shumway-Cook A, Vermeulen SS et al. (1998).
activity in the basal ganglia in patients with generalized Gamma knife radiosurgery as a lesioning technique in
dystonia and hemiballismus. Ann Neurol 46: 2235. movement disorder surgery. J Neurosurg 89: 183193.
Vitek JL, Bakay RA, Freeman A et al. (2003). Randomized Zirh AT, Lenz FA, Reich SG et al. (1998). Patterns of
trial of pallidotomy versus medical therapy for Parkinsons bursting occurring in thalamic cells during parkinsonian
disease. Ann Neurol 53: 558569. tremor. Neuroscience 83: 107121.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 43

Deep brain stimulation

J. VOLKMANN* AND G. DEUSCHL

Department of Neurology, Christian-Albrechts-University, Kiel, Germany

43.1. Introduction worldwide in the year 2005, which may underline the
importance of this therapy in the treatment algorithm of
Deep brain stimulation (DBS) is an alternative to abla- PD. In 2004 sufficient scientific evidence had accumu-
tive stereotaxy, which was first introduced in the lated for the Movement Disorder Society to conclude
1970s but has not been routinely used for the treatment in its evidence-based medical review update that DBS
of movement disorders until the pioneering work of of the STN was efficacious for the symptomatic control
Benabid and colleagues in Grenoble became public of parkinsonism. Although the status of DBS has chan-
in the late 1980s. The subthalamic nucleus (STN), ged from an investigational therapy to an evidence-based
which was introduced as a target for DBS in 1993, routine treatment, a number of issues still need to be
quickly changed the scope of DBS from a sympto- addressed in controlled clinical trials, such as the target
matic treatment of tremor to a highly effective therapy selection and the safety in subgroups of patients or the
for all cardinal symptoms of Parkinsons disease (PD) timing of DBS within the course of disease.
and levodopa-induced motor complications.
DBS is based on the empirical observation that 43.2. Physiological mechanisms
high-frequency electrical stimulation of specific brain
targets can mimic the effect of a lesion without the Despite the clinical success of DBS and the rapidly
need for destroying brain tissue. DBS is accomplished expanding application for different neuropsychiatric
by permanently implanting an electrode into the target disorders, its mechanism of action is still poorly under-
area and connecting it to an internal pulse generator. stood. DBS mimics the clinical effects of lesioning in all
The stimulator settings can be adjusted telemetrically three target structures currently used for the treatment of
with respect to electrode configuration, current ampli- PD (ventrolateral thalamus, internal pallidum and STN),
tude, pulse width and pulse frequency. DBS has rapidly when high-frequency (> 100 Hz) stimulation (HFS) is
replaced ablative stereotactic surgery in movement applied. Stimulation at lower frequencies (< 50 Hz) results
disorders with several advantages: (1) DBS does not in little or no clinical benefit or may even aggravate par-
require a destructive lesion to be made in the brain; kinsonian symptoms (Moro et al., 2000; Fogelson et al.,
(2) it can be performed bilaterally with relative safety, 2005).
in contrast to most lesioning procedures; (3) stimulation When stimulating the central nervous tissue, the
parameters can be adjusted postoperatively to improve electrode is placed within a complex volume conductor
efficacy, to reduce adverse effects and to adapt DBS containing three main elements, which could be
to the course of disease; and (4) DBS is in principle affected by stimulation: (1) local cells, that have their
reversible and does not preclude the use of possible cell body close to the stimulating electrode; (2) afferent
future therapies in PD requiring integrity of the basal fibers making synaptic contact to the local cells; and
ganglia circuitry. (3) fibers of passage, where both the cell body and axon
The number of DBS procedures for PD has been stea- terminal are far from the stimulating electrode. Given
dily increasing over the past decade and has reached the stimulation parameters and electrode configurations
a cumulative number of approximately 22 000 patients presently used, it is likely that DBS directly affects all

*Correspondence to: Dr Jens Volkmann, Department of Neurology, Christian-Albrechts-University, Schittenhelmstr. 10, 24105
Kiel, Germany. E-mail: j.volkmann@neurologie.uni-kiel.de, Tel: 49-431-597-8509, Fax: 49-431-597-8506.
262 J. VOLKMANN AND G. DEUSCHL
three elements. The inability to stimulate a single thalamic DBS in parkinsonian tremor have revealed
element selectively complicates our ability to under- conflicting results. Parker et al. (1992) could demon-
stand the contribution of each of these elements to the strate that suppression of parkinsonian tremor by thala-
final behavioral effect of DBS. mic DBS was associated with significant reduction of
Electrical stimulation of nervous tissue, in general, is regional cerebral blood flow (rCBF) in the ipsilateral
more likely to activate large myelinated fibers before putamen, sensorimotor cortex, supplementary motor
small axons or cell bodies, axons near the cathode before area (SMA) and contralateral cerebellum. They argued
those near the anode and axons oriented parallel to the that DBS could inactivate the involuntary running of a
electrode before axons oriented transversely (Ranck, central motor program for alternating movements
1975). Presently, there exist five principal hypotheses involving the basal gangliathalamocortical loop and
to explain the therapeutic mechanism of DBS, that have that cerebellar deactivation might be secondary to
derived from in vitro and in vivo experiments in animals reduced proprioceptive input. In contrast, Deiber
or intraoperative recordings in humans: (1) non-synaptic et al. (1993) found reduced cerebellar activity exclu-
blocking of neural output through inactivation of vol- sively during thalamic DBS and therefore assumed
tage-dependent ion channels near the stimulating elec- a tremor generator within the cerebellothalamic
trode (Benazzouz et al., 1995; Beurrier et al., 2001); network.
(2) antidromic stimulation of inhibitory afferents to the Tremor-locked neuronal discharges have been
target nucleus and local release of gamma-aminobutyric found in the internal pallidum and STN (Rodriguez-
acid (GABA) (Dostrovsky et al., 2000); (3) driving of Oroz et al., 2001) during stereotactic surgery, but most
efferents and masking (or jamming) of pathological net- prominently in areas of the ventrolateral thalamus
work activity (Montgomery and Baker, 2000; Hashimoto receiving cerebellar input (Lenz et al., 1994). Tremor
et al., 2003); (4) modulation of afferent input to the target in PD, however, is unlikely to originate from the olivo-
nucleus (Strafella et al., 1997; Anderson et al., 2006); and cerebellar loop, because cerebellectomy does not
(5) synaptic transmission failure of the efferent output of abolish resting tremor, but rather transforms it into a
stimulated neurons as a result of synaptic depletion slow resting, postural and intention tremor (Deuschl
(synaptic depression) (Brock et al., 1952; Urbano et al., et al., 1999). Nevertheless, most neurosurgeons believe
2002). Which of these mechanisms alone or in combina- that the cerebellar relay nucleus, ventrointermediate
tion is involved in the behavioral effect of DBS is nucleus (VIM), represents the optimal site for thalamic
unknown and may depend on the anatomy of the target lesioning or stimulation in various types of tremor,
structure (e.g. the thalamic target contains a local inhibi- including parkinsonian tremor. The regional anatomy,
tory circuitry of interneurons and projections from reticu- however, is complex and pallidothalamic afferents to
lar thalamic nuclei, which does not exist in the pallidal the adjacent nucleus ventrooralis anterior (Voa)
and subthalamic target) and the exact location of the sti- receiving pallidal input cross at the base of the VIM
mulating electrode. First clinical evidence in line with or even pass through (Krack et al., 2002), such that
the mechanisms proposed above suggests that the opti- this site may rather reflect a strategic anatomical bot-
mal target point for DBS may not be within the target tleneck where interventions may affect either loop of
nuclei themselves but rather at entry or exit sites of fiber the extrapyramidal motor system. Because postmortem
tracts or adjacent crossings of large fiber bundles studies are rare and inconclusive, it is difficult to dis-
(Velasco et al., 2001; Saint-Cyr et al., 2002; Voges cern currently whether thalamotomy or DBS effects
et al., 2002; Hamel et al., 2003; Murata et al., 2003; in the various types of tremors are mediated by inhibi-
Herzog et al., 2004; Plaha et al., 2004; Kitagawa et al., tion of abnormal activity originating from the basal
2005). These preliminary results, however, await further ganglia, cerebellum or both.
confirmation in larger series and for other target sites.
For this reason, the exact documentation of the final 43.2.2. Systemic effects of pallidal
electrode location postoperatively and the corresponding or subthalamic DBS
clinical effects not only serves for internal quality control
but is also essential to create a sufficiently large database The pathophysiological hallmarks of the parkinsonian
to address the important controversy about DBS mechan- state in animal models of PD are abnormally increased
isms and the optimal electrode location. neuronal discharge rates in the STN and globus palli-
dus internal segment (GPi). Lesioning or HFS of
43.2.1. Systemic effects of thalamic DBS these structures reverses the symptoms of 1-methyl-
4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced
Two studies using positron emission tomography (PET) parkinsonism, thus supporting the functional signifi-
to reveal the cerebral activity changes associated with cance of these changes. The increased output activity
DEEP BRAIN STIMULATION 263
of the basal ganglia results from opposite effects of
the striatal dopaminergic depletion on the direct and
indirect basal ganglia pathway according to the cur-
rent model of basal ganglia circuitry: The activity of
the direct basal ganglia pathway is reduced, resulting
in decreased inhibition of the GPi. Along the indirect
basal ganglia pathway excitatory afferents from STN
are driving GPi neurons to be overactive as a result of
decreased inhibitory input to the STN from globus
pallidus external segment (GPe) (DeLong, 1990).
The hyperactive inhibitory influence of the GPi on
thalamic nuclei and via this path on cortical motor
areas linked with the corresponding thalamic areas
is thought to cause bradykinesia and other PD
symptoms.
Pallidotomy can restore a normal level of activity in
the thalamocortical motor pathways by decreasing the
excessive inhibitory drive from the GPi to the ventro-
lateral thalamus. PET studies after successful pallidot-
omy showed increased rCBF and metabolism in
ipsilateral SMA and dorsolateral prefrontal cortex of Fig. 43.1. Axial (A) and coronal section (B) of a T2-weighted
parkinsonian patients (Eidelberg et al., 1996; Samuel magnetic resonance imaging scan at the level of the subthala-
mic nucleus (STN). On the upper images the STN is visible
et al., 1997; Ceballos Baumann et al., 1999). Similar
preoperatively in relation to the red nucleus (RN) and substan-
reversible changes of rCBF in SMA and dorsolateral
tia nigra (SN). The lower rows depict the artifact caused by
prefrontal cortex were described after HFS of the the deep brain stimulation electrode implanted bilaterally
internal pallidum (Davis et al., 1997) or STN within the STN.
(Ceballos Baumann et al., 1999).
Although these observations may explain the alle-
viation of akinesia, they provide little insight into the
43.3. Methods
mechanisms by which tremor, rigidity or dyskinesias
might be improved. The simple rate model of abnor- 43.3.1. Surgical procedure
mal neuronal activity in PD would predict reduced pal-
lidal discharge rates in hyperkinetic states and thus a Accurate electrode placement requires the use of a
further worsening with pallidal lesioning or DBS stereotactic head frame. Imaging is one of the most
(Marsden and Obeso, 1994). Clinically, however, the critical aspects of the stereotactic procedure and gener-
most consistent effect of pallidal surgery is a marked ally includes ventriculography, computed tomography
reduction of contralateral hyperkinesias (Volkmann (CT) and/or magnetic resonance imaging (MRI). His-
et al., 2004). It has therefore been suggested that palli- torically, ventriculography had been the radiological
dal or STN surgery in more general terms is effective gold standard for a reliable identification of the ante-
by removing the disturbing influence of a noisy basal rior and posterior commissure (AC, PC) and landmark-
ganglia operator on to thalamocortical motor areas based, indirect stereotactic targeting, but today it has
(Marsden and Obeso, 1994). Noisy or unphysiologi- largely been replaced by CT and MRI. The advantage
cal basal ganglia activity could result not only from of MRI lies in the direct visualization of the target
changes in discharge rate but also from abnormal pat- (STN, GPi) and neighboring structures (Fig. 43.1).
terning, oscillation or synchronization of neuronal fir- Moreover, three-dimensional reconstructions from
ing (Bergman and Deuschl, 2002; McIntyre et al., MRI or CT allow coronal, axial and sagittal determina-
2004b; Brown and Williams, 2005). Furthermore, des- tion of the AC and PC, planning of the entry point and
cending projections from the basal ganglia to the a control of the entire trajectory for possible conflicts
brainstem nuclei and spinal cord are often neglected with deep and superficial vessels. The radiologically
and may also play an important role in the pathophy- derived target is further refined intraoperatively by
siology of rigidity, postural instability and gait disor- neurophysiological techniques. Intraoperative stimula-
der of PD (Delwaide et al., 2000; Pahapill and tion predicts the effect of chronic stimulation and
Lozano, 2000; Nandi et al., 2002a, b; Potter et al., is crucial to determine the final site of electrode
2004). implantation. To allow for clinical evaluation of the
264 J. VOLKMANN AND G. DEUSCHL
stimulation response, the implantation is usually per- dorsolateral STN (Saint-Cyr et al., 2002; Voges
formed under local anesthesia in awake patients. et al., 2002; Herzog et al., 2004; Zonenshayn et al.,
Microelectrode recordings may provide additional 2004). This target corresponds to the sensorimotor
information on the nuclear boundaries and allow map- territory of the nucleus, which may be identified by
ping of the target area along several passes (Fig. 43.2). the presence of movement-responsive cells in micro-
Microelectrode mapping is particularly useful when electrode recordings (Rodriguez-Oroz et al., 2001).
clinical testing is difficult or inconclusive due to a In this location the electrode also passes Forelss field
microlesioning effect or in uncooperative patients. H and the zona incerta, which are dorsally adjacent to
There is some debate on whether microelectrode map- the STN. Whether current spread into the subthalamic
ping increases the surgical bleeding risk (Hariz and white matter is necessary for optimal clinical success
Fodstad, 1999). However, leading centers in the field is a matter of debate. However, exclusive stimulation
routinely explore multiple microelectrode trajectories of subthalamic white matter by placing the electrode
and have reported low complication rates along with dorsal to the STN boundary results in an unfavorable
excellent clinical results in DBS (Krack et al., 2003; relation between clinical improvement and current
Lyons et al., 2004; Mehdorn et al., 2005; Schupbach consumption (Herzog et al., 2004).
et al., 2005; Goodman et al., 2006). After insertion of The usual coordinates for pallidotomy are 2021 mm
the permanent DBS electrode and attachment of the lateral to the intercommissural line (ACPC line), 56
extension cable, the system is connected to the internal mm below and 3 mm anterior to the mid commissural
pulse generator. This is either done immediately or in point (Laitinen et al., 1992). This corresponds to the
a staged fashion. ventrolateral aspect of the GPi, where microelectrode
recordings demonstrate movement-related cells
43.3.2. Target location (Iacono et al., 1997; Lozano et al., 1997). To what
extent the topography of pallidal stimulation effects
Optimal targeting is the prerequisite for clinical suc- is related to the anatomical subdivision of GPi or the
cess in DBS. Clinically effective stimulation is most anatomy of pallidal fiber tracts remains a matter of
commonly directed at the anterior segment of the speculation. Neither is it known whether the optimal

Zona Incerta (4.3 mm)

White matter (3.7 mm)

VLp
d STN (3.5 mm)
CL
VA
R
MDpl

VLp STN (2 mm)


PuA v
VLa
CM
VM
VPM
ZI STN (0.7 mm)
Po pc
STN
Anatomical
target
point SNr (0.3 mm)
SN

0.1 sec
Fig. 43.2. Typical traces of microelectrode recordings along a trajectory to the subthalamic nucleus (STN) are displayed in
relation to the recording site. Proximal to the STN sparse neuronal activity can be found in the zona incerta alternating with
traces of white-matter noise, when the electrode passes fiber tracts. STN is identified by a marked increase in background noise
and large-amplitude, burst-like spike discharges. This pattern changes to tonic, high-frequency discharges when the microelec-
trode enters substantia nigra (SNr). The characteristic difference in neuronal discharge patterns of STN and SNr may help to
refine the anatomical target, because the ventral border of STN is difficult to delineate on magnetic resonance images, as it
was in this case, where the anatomical target was chosen too deep.
DEEP BRAIN STIMULATION 265
sites for pallidal lesioning and stimulation are identi- events (induced by stimulation or medication changes)
cal. Two independent studies on the acute effects of depend on the stimulated area and will be discussed
pallidal stimulation have found a better reduction of with the clinical results of each target.
parkinsonian symptoms by stimulating the distal con-
tacts of the quadrupolar stimulating electrode (Bejjani 43.3.3.1. Surgery-related adverse events
et al., 1997; Krack et al., 1998b), which were located in
DBS requires a craniotomy and one or several needle
the dorsolateral aspect of GPi, closely neighboring
passes through the brain, carrying with it the risks of
GPe. Stimulation of the ventromedial GPi, in contrast,
intracranial hemorrhage and damage to adjacent brain
was antidyskinetic but also blocked the beneficial effect
structures. The risk of unintended injury to adjacent
of levodopa on akinesia. In our own experience, the posi-
brain structures, however, is much smaller than in abla-
tion of the most beneficial electrode pole used for
tive procedures, where the exact size of the final thera-
chronic pallidal stimulation was on average slightly peutic lesion may be difficult to predict. In the only
more lateral and dorsal than the standard pallidotomy
prospective controlled study randomizing patients to
target (Volkmann et al., 1998). This corresponds to a
either thalamic stimulation or lesioning for the treatment
location lateral and dorsal to the optic tract in postopera-
of tremor, the cumulative rate of neurological adverse
tive MRI controls. Given a current spread of approxi-
events was 47% in the thalamotomy group and 16% in
mately 3 mm around the cathode, based on average
the DBS group. All adverse events in the DBS group
electrical parameters for pallidal stimulation (Ranck,
were reversible when stimulation was turned off. One
1975; McIntyre et al., 2004a), it is likely that stimulation
death related to an intracranial hemorrhage in the DBS
at these coordinates will not remain restricted to the sen- group, however, cautions about the inherent risk of any
sorimotor region of GPi, but may spread to the efferent
stereotactic intervention (Schuurman et al., 2001).
fiber tracts of GPi and medial regions of GPe.
In the literature intracranial hemorrhages are reported
A reduction of parkinsonian tremor may be achieved
in 14% of patients undergoing functional stereotactic
by stimulation within a rather larger volume encom-
procedures (Favre et al., 2002; Binder et al., 2003; Terao
passing the ventrolateral thalamus and subthalamic
et al., 2003). The proportion of asymptomatic and symp-
areas. There is no consensus in the literature regarding
tomatic bleedings within this percentage is unclear, but
the clinically most effective site. Most neurosurgeons
the prevalence of persistent neurological deficits seems
favor the thalamic VIM as the optimal site for thalamic to be considerably lower compared with the total sum
lesioning or stimulation in various types of tremor,
of hemorrhagic complications. Transient postoperative
including parkinsonian tremor. Others have reported
confusion is present in up to 20% of the patients after
excellent outcome with DBS of the subthalamic white
implantation of STN leads. This proportion is higher
matter, including the prelemniscal radiation and zona
than the rate of confusion previously reported for other
incerta (Velasco et al., 2001; Plaha et al., 2004;
targets and may be related to the usual STN trajectory,
Kitagawa et al., 2005). These clinical observations are
which passes the head of the caudate on both sides
complemented by recent physiological data suggesting
(Woods et al., 2002). An additional source of surgery-
that DBS might be effective for tremor by blocking related morbidity is infection with an incidence of
afferent synaptic input to thalamic tremor cells, thereby
34% (Lyons et al., 2004; Goodman et al., 2006).
stopping propagation of abnormal oscillations beyond
the thalamus (Anderson et al., 2006). Finally, DBS of
the STN is considered an alternative to thalamic surgery 43.3.3.2. Device-related adverse events
and reduces the tremor score by approximately 80% The reported rate of hardware-related problems (e.g.
even in PD patients with severe high-amplitude tremor lead dislocation, lead breakage, internal pulse generator
(Krack et al., 1998a; Rodriguez et al., 1998). Because dysfunction, skin erosion) varies between different cen-
no comparative trials exist, it is difficult to conclude ters in the range of 530% (Hariz et al., 1999; Oh et al.,
on a better efficacy of either target. 2001; Lyons et al., 2004; Goodman et al., 2006) and
must not be underestimated. Most hardware-related
43.3.3. Safety problems, however, occurred in the first patients of a
series and were less frequent afterwards. This sheds
Adverse events associated with DBS must be divided light on the sophisticated nature of the procedure, which
into those related to the surgical procedure, to the requires extensive neurosurgical skill learning before
implanted device, to stimulation and to medication getting into a routine with DBS surgery. Device-related
changes necessitated by DBS. In this section the safety complications are usually manageable, but often require
of the operative procedure and hardware-associated an additional surgical intervention. The permanent
problems are discussed. Therapy-related adverse morbidity associated with these problems is low.
266 J. VOLKMANN AND G. DEUSCHL
43.3.3.3. Summary (2000, 2001) demonstrated after 2 years of follow-up
The risk of permanent neurological deficits associated that both procedures resulted in about equal sympto-
with DBS is lower than in lesional stereotaxy. Most lar- matic relief of tremor, but that functional outcome
ger controlled series report permanent neurological according to the Frenchay activity index was signifi-
morbidity in the region of 23%. Mortality is extremely cantly better in the DBS group. The authors related
rare. Therefore, the benefit-to-risk ratio of the DBS pro- this difference to permanent mild neurological seque-
cedure seems favorable, at least in severely disabled lae of thalamotomy such as impairment of fine motor
patients. A careful selection of candidates for increased skills due to lesions encroaching upon the internal
neurosurgical risks should help to maintain a relatively capsule.
low level of morbidity.
43.4.2. Pallidal deep brain stimulation
43.4. Clinical efficacy
The consistent effect of pallidal stimulation in all current
43.4.1. Thalamic deep brain stimulation reports is a marked reduction of contralateral levodopa-
induced dyskinesias. Improvement of off-period symp-
Experience with DBS in the thalamus for the treatment toms of parkinsonism is more variable but significant in
of parkinsonian tremor has been generally safe and most studies. Figure 43.3. summarizes the results of 15
effective. Clinical success is usually defined as a com- studies in the literature, including a total of 276 patients
plete abolition or a reduction of contralateral tremor to (Gross et al., 1997; Pahwa et al., 1997; Tronnier et al.,
grade 1 on the tremor-rating scale (mild and intermittent 1997; Ghika et al., 1998; Krack et al., 1998c; Kumar
tremor) with chronic stimulation. The success rates of et al., 1998; Burchiel et al., 1999; Merello et al., 1999;
DBS in the following paragraph relate to this definition. The Deep-Brain Stimulation for Parkinsons Disease
For tremor in PD, Benabid et al. (1991) reported a suc- Study Group, 2001; Volkmann et al., 2001; Durif et al.,
cess rate of 88% in 26 patients and very low morbidity 2002; Loher et al., 2002; Ogura et al., 2004; Peppe
even after bilateral surgery. These findings were con- et al., 2004; Anderson et al., 2005). In these studies the
firmed in a European multicenter study with 74 patients median improvement of off-period motor symptoms
and a follow-up of 1 year (Limousin et al., 1999). The induced by pallidal stimulation was 36% (lower quartile
tremor reduction is sustained in the long term for up 31%, upper quartile 43.1%) and the median reduction of
to 7 years, but thalamic DBS does not improve other dyskinesias in the on-state 71% (lower quartile 52.5%,
symptoms of PD, nor does it alleviate long-term com- upper quartile 78.5%). These data leave little doubt
plications of levodopa treatment. Akinesia, rigidity, gait about the clinical efficacy of palllidal DBS.
and postural problems may progress despite successful In our own experience bilateral GPi stimulation
treatment of tremor and can cause new therapeutic (n 11) led to a 54  33.1% improvement of the
difficulties (Rehncrona et al., 2003). off-period Unified Parkinsons Disease Rating Scale
In a double-blind randomized study comparison of (UPDRS) motor score at 1-year follow-up (Volkmann
thalamotomy and thalamic DBS Schuurman et al. et al., 1998, 2001). In UPDRS subscores we found

Fig. 43.3. Summary of results of pallidal deep brain stimulation (DBS) in the literature: 15 publications from 1997 to 2005:
276 patients, 54 unilateral. For each study the average reduction of off-period motor symptoms by levodopa before surgery
(levodopa response in %), by DBS at the final visit, the reduction of dyskinesias and the reduction of the levodopa-equivalent
daily dose (LEDD) are represented by a symbol. Horizontal lines denote the median of all studies. STIM, stimulation.
DEEP BRAIN STIMULATION 267
significant improvements for bradykinesia, tremor and tions for therapeutic long-term stimulation can only
posture and gait and a tendency towards improvement be determined in future studies carefully relating the
of rigidity. On-period motor symptoms did not sig- efficacy of chronic pallidal stimulation to the exact tar-
nificantly change after surgery except for dyskinesias, get location within GPi determined by neuroimaging
which were reduced by 83% at 1-year follow-up. The techniques and microelectrode recordings.
alleviation of dyskinesias and the reduction of off- Despite the common assumption that DBS is equally
period motor symptoms in combination led to a signif- effective but safer than lesioning techniques, there is
icant reduction of self-perceived motor fluctuations in only one clinical trial currently available addressing this
our patients. Unfortunately, the initial benefit on off- issue. In a group of 13 patients randomized to either
period symptoms of PD tended to decrease in the long unilateral pallidal DBS or radiofrequency lesioning of
term, whereas the antidyskinetic effect remained stable the GPi, Merello and colleagues (1999) found about
for up to 5 years. The worsening of akinetic-rigid equal improvement of the UPDRS motor score after
symptoms of PD had to be compensated for by 3-month follow-up. There was greater reduction of
increases in dopaminergic medication (Volkmann contralateral dyskinesias after pallidotomy, whereas
et al., 2004), which kept the total duration of off-time bilateral hand-tapping scores improved more with DBS.
after surgery at a stable level. As a result, both hyper-
and hypokinetic motor fluctuations were still reduced 43.4.3. Subthalamic nucleus deep brain stimulation
after 5 years but the increases in dopaminergic medi-
cation caused additional problems in some patients, DBS of the STN has replaced pallidal DBS in most
such as delusions or gambling. centers for the treatment of advanced PD. This prefer-
Interestingly, one group reported, in contrast to all ence for the subthalamic target is rarely based on own
other available studies, a reduction of dyskinesias but comparative experience. Centers that started a surgical
a worsening of motor function during the medication program for advanced PD with STN DBS have little
on period (Tronnier et al., 1997) after bilateral palli- incentive to explore another target, if they obtain good
dal stimulation. Because the exact position of the sti- results in the STN. For this reason, there is a reporting
mulating electrodes is uncertain in this and most bias in the literature in favor of STN DBS in recent years.
other studies, it is difficult to discern how much of Figure 43.4. summarizes 39 clinical studies of STN DBS
the variable effect of pallidal stimulation results from in advanced PD, reporting on a total of 1129 patients
different target locations within GPi. Two studies (Moro et al., 1999; Pinter et al., 1999; Houeto et al.,
(Bejjani et al., 1997; Krack et al., 1998b) claim, based 2000; Molinuevo et al., 2000; Broggi et al., 2001;
on the observation of acute stimulation effects, that Katayama et al., 2001; Krause et al., 2001; Lopiano
DBS of the ventral GPi may block dyskinesias but et al., 2001; The Deep-Brain Stimulation for Parkinsons
aggravate akinesia, whereas dorsal GPi stimulation Disease Study Group, 2001; Volkmann et al., 2001;
ameliorates akinesia on account of being prodyski- Doshi et al., 2002; Figueiras-Mendez et al., 2002; Iansek
netic. The importance of these experimental observa- et al., 2002; Kleiner-Fisman et al., 2002; Lanotte et al.,

Fig. 43.4. Summary of results of subthalamic nucleus deep brain stimulation (DBS) in the literature: 39 publications, from
2000 to 2005: 1129 patients. For each study, the average reduction of off-period motor symptoms by levodopa before surgery
(levodopa response in %), by DBS at the final visit, the reduction of dyskinesias and the reduction of the levodopa-equivalent
dose (LEED) are represented by a symbol. Horizontal lines denote the median of all studies. STIM, stimulation.
268 J. VOLKMANN AND G. DEUSCHL
2002; Martinez-Martin et al., 2002; Ostergaard et al., that significantly improved were stigma (54.4  18.1%),
2002; Romito et al., 2002b; Simuni et al., 2002; Tavella activities of daily living (51.6  18.2%), mobility (38.5 
et al., 2002; Thobois et al., 2002; Valldeoriola et al., 18.2%), bodily discomfort (35.8  15.4%) and emotional
2002; Vesper et al., 2002; Vingerhoets et al., 2002; well-being (32.1  18.1%). Dimensions with modest
Herzog et al., 2003b; Krack et al., 2003; Landi et al., benefit included social support (17.0  19.1%), cognition
2003; Pahwa et al., 2003; Tamma et al., 2003; Varma (16.5  15.0%) and communication (13.0  26.9%).
et al., 2003; Esselink et al., 2004; Ford et al., 2004; Jaggi
et al., 2004; Peppe et al., 2004; Capecci et al., 2005; 43.4.4. Comparison of the different targets
Lyons and Pahwa, 2005; Minguez-Castellanos et al.,
2005; Rodriguez-Oroz et al., 2005; Visser-Vandewalle Few studies have compared the effect of STN and GPi
et al., 2005). The median improvement in the UPDRS DBS. Most of them are parallel-group comparisons,
motor score in the off-period was 50.2% (lower quartile except for one randomized but underpowered clinical
42%, upper quartile 60.8%) More specifically, improve- trial (Burchiel et al., 1999; Anderson et al., 2005).
ment is seen in off-phase akinesia, rigidity, tremor Krack et al. (1998c) reported a 71% improvement of
(Krack et al., 1998c; Rodriguez et al., 1998), gait (Allert the off-period UPDRS motor score with STN stimu-
et al., 2001; Faist et al., 2001; Stolze et al., 2001) and bal- lation but only 39% with GPi stimulation in a group of
ance. Bilateral STN stimulation improves most axial fea- 13 patients with young-onset PD. This significant dif-
tures of PD that responded to levodopa before surgery ference resulted from a greater reduction of akinesia
(Bejjani et al., 2000b). Hypokinetic motor fluctuations in the STN-stimulated group, whereas other parkinso-
tend to disappear and patients show marked improve- nian symptoms showed about equal improvement.
ment in the activities of daily living (median improve- In contrast, Burchiel and colleagues (Burchiel et al.,
ment 37.5%). Off-period dystonia is immediately 1999; Anderson et al., 2005) found no difference in
alleviated, synchronously with stimulation (Krack reduction of akinetic-rigid symptoms of PD or dyski-
et al., 1999). The on-period dyskinesias are reduced in nesias between STN- and GPi-stimulated patients in
parallel with a marked reduction of the equivalent daily a well-designed, randomized prospective trial. In our
levodopa dose (LEED). The median reduction of dyski- own retrospective analysis of the initial 11 patients
nesias was 73% (lower quartile 64%, upper quartile implanted within GPi and 16 patients implanted within
91%) in our survey and the median reduction of the STN, the main finding was a 54  33.1% improve-
LEED 54% (lower quartile 40%, upper quartile 66%). ment of off-period motor symptoms in the first and
In the long term, the antidyskinetic effect of STN stimu- 67  22.6% improvement in the later group after
lation may be equivalent to or superior to that of GPi sti- 1 year of follow-up (Volkmann et al., 2001). The 10
mulation if the levodopa dose remains reduced. Whereas 15% difference between both groups was not signifi-
STN stimulation has a direct effect on off-period dysto- cant and power analysis suggested that, based on the
nia, on-period dyskinesias are decreased by a more com- group variances, a much larger trial including a mini-
plex mechanism, involving the decrease in levodopa mum of 135 patients in each arm would have been
dosage and possibly adaptive neuronal changes induced needed to prove significance of this possible small dif-
by continuous HFS. Bejjani et al. (2000a) demonstrated ference in favor of STN stimulation. A non-rando-
that the sensitization phenomenon resulting from long- mized multicenter study enrolled 96 patients with
term intermittent levodopa administration presumably STN DBS and 38 with GPi DBS (The Deep-Brain Sti-
causing dyskinesias is partially reversible with STN mulation for Parkinsons Disease Study Group, 2001).
DBS. His group used a challenging dose to induce dys- Except for levodopa-induced dyskinesias, which were
kinesias before surgery and, after 6 months of continu- greatly alleviated in both groups, all other outcome
ous STN DBS, found that in the stimulation off- variables favored the STN group, although some of
condition, the severity of levodopa-induced dyskinesias the differences were small. At 3-month follow-up,
was greatly reduced compared to baseline. double-blind, cross-over evaluations demonstrated that
The motor changes after STN DBS can have a signifi- STN stimulation was associated with a median
cant impact on patients quality of life (Just and Oster- improvement in the motor score (as compared with no
gaard, 2002; Lagrange et al., 2002; Martinez-Martin stimulation) of 49% and GPi stimulation with a median
et al., 2002; Spottke et al., 2002; Tamma et al., 2003; improvement of 37%. Between the preoperative and
Troster et al., 2003; Esselink et al., 2004; Lezcano et al., 6-month visits, the percentage of time during the day
2004; Drapier et al., 2005; Lyons and Pahwa, 2005). In that patients had good mobility without involuntary
studies using the Parkinsons Disease Questionnaire movements increased from 27 to 74% with STN sti-
PDQ-39, an average improvement of the summary index mulation and from 28 to 64% with GPi stimulation.
of approximately 35% was reported. PDQ-39 dimensions Interestingly, these differences in the severity of
DEEP BRAIN STIMULATION 269
off-period symptoms did not affect the total on-time is not achieved without side-effects, patient and physician
after surgery, which was still significantly increased may deliberately accept a certain degree of stimulation-
34 years after surgery to an identical extent after induced adverse effects.
DBS of the STN or GPi (Rodriguez-Oroz et al., 2005). In thalamic DBS, paresthesias and dysarthria are
In a small group of PD patients that had simultaneous among the most common stimulation-induced side-
bilateral electrode implants in the STN and the GPi, effects. Especially with bilateral DBS some dysarthria
Peppe et al. (2004) recently confirmed the slightly better will have to be accepted in up to 10% of cases if opti-
efficacy of STN DBS in reducing off-period symptoms. mal tremor control is desired. Less frequent are real
They found a larger decrease in off-period UPDRS dystonia or pseudodystonia resulting from costimula-
III scores with DBS of the STN (54.5%) than during tion of the pyramidal tract (Limousin et al., 1999;
DBS in the GPi (43.1%), when either target was stimu- Schuurman et al., 2000, 2001; Rehncrona et al., 2003).
lated separately. The additive effect of stimulation in Stimulation-induced side-effects in pallidal DBS
both targets was small (54.5%) and not significant. are rare and mostly transient during the immediate
The available data, therefore, suggest a better effi- postoperative adaptation of stimulation parameters.
cacy of STN DBS in reducing off-period motor symp- They include visual field disturbances from current
toms, but formal confirmation in a larger prospective spread to the optic tract, tetanic muscle contractions
randomized trial has to be awaited (Follett et al., (pseudodystonia) from costimulation of the pyramidal
2005). Other consistent differences between pallidal tract and nausea or dizziness.
and STN stimulation concern medication requirements Possible therapeutic problems with DBS of the STN
and stimulation parameters. Patients with pallidal sti- result from the complex interactions of medical ther-
mulation continue to require preoperative anti-PD apy and electrical stimulation. One of the challenges
medication doses and in some cases even higher doses in follow-up treatment is to distinguish between gen-
are introduced postoperatively. In contrast, STN uine stimulation-induced side-effects and pre-existing
stimulation allows an average reduction of dopaminergic symptoms of the disease that are uncovered by a com-
medication in the range of 5060% and approximately bination of reduced dopaminergic therapy and inade-
10% of patients are able to discontinue all dopaminergic quate stimulation effects. Postoperative speech, gait
drugs (Moro et al., 1999). This gives STN DBS a favor- and balance problems fall within this particular cate-
able benefit-to-cost ratio, but these advantages contrast gory. Stimulation-induced dyskinesias are one of the
with a need for more intensive postoperative monitoring most frequent and important specific side-effects of
and a higher incidence of adverse events (Volkmann STN stimulation. The appearance of dyskinesias indi-
et al., 2001; Rodriguez-Oroz et al., 2005). cates correct lead placement and an additive effect
In summary, no general advice can be given yet has been observed in bilateral stimulation. Stimula-
about the optimal target for the treatment of advanced tion-induced dyskinesias are worsened by levodopa.
PD. The STN will be the preferred choice for younger Therefore initial programming should always be done
patients in most centers based on the expected early in the morning when the patient is off medica-
levodopa savings and the lower energy expenditure. tion. Stimulation is continuously increased over a per-
Pallidal stimulation, however, is an effective proce- iod of days or weeks until a satisfactory effect on
dure and may still be considered in older patients, bradykinesia in the off-phase is achieved and at the
who may not tolerate levodopa withdrawal or time- same time the levodopa dosage is lowered (Volkmann
consuming programming of STN-DBS. Mild cognitive et al., 2000).
impairment or a history of depression (as outlined in Either current diffusion due to excessive stimula-
section 43.3.3.1) could also be arguments in favor of tion parameters or incorrect lead placement may result
GPi DBS. in reversible stimulation-induced side-effects, such as
tonic muscle contractions, dysarthria, eyelid-opening
43.4.5. Therapy-related adverse effects apraxia, ocular deviation, ipsilateral mydriasis, flush-
ing, unilateral (ipsilateral) perspiration (contralateral)
Optimal surgical positioning of the stimulating elec- paresthesias, worsening of akinesia and a reversal of
trode helps to reduce the risk of stimulation-induced the levodopa effect. These reversible side-effects may
side-effects which mostly result from unintended cur- help to define the optimal target intraoperatively or
rent spread to adjacent fiber tracts or nuclei. Stimula- to track the deviation of a misplaced electrode after
tion-induced side-effects are fully reversible when surgery (Volkmann et al., 2000).
stimulation is stopped and can be improved in most Despite the theoretical concern that pallidal or STN
cases by changing stimulation parameters or electrode stimulation could interfere with the functioning of
configuration. If sufficient control of motor symptoms cognitive basal ganglia loops, most studies found no
270 J. VOLKMANN AND G. DEUSCHL
or clinically insignificant changes in neuropsychologi- 2. Levodopa has psychotropic effects in addition to
cal functioning after DBS (Ardouin et al., 1999; the well-known motor effects. Patients describe
Jahanshahi et al., 2000; Pillon et al., 2000; Trepanier the action of levodopa as pleasantly euphoria-
et al., 2000; Alegret et al., 2001). Some reports, how- and drive-enhancing. In extreme cases, mania
ever, have warned that STN DBS has a risk of inducing and hypersexuality may be affective and beha-
cognitive decline with a frontal executive dysfunction vioral side-effects of dopaminergic therapy. The
similar to progressive supranuclear palsy in older medication reduction after STN stimulation may,
patients (above 70 years) or those with minimal cogni- therefore, cause withdrawal phenomena with an
tive dysfunction prior to surgery (Saint-Cyr et al., impact on mood and drive.
2000; Dujardin et al., 2001). 3. The basal ganglia are integrated into associative-
Mood disorders are among the most frequently cognitive and limbic regulatory systems, so that
observed postoperative side-effects in STN stimulation psychiatric symptoms could also result as a direct
(Limousin et al., 1998; Volkmann et al., 2001; side-effect of stimulation. However, the acute
Rodriguez-Oroz et al., 2005) but the true prevalence is emotional effect of STN DBS is mood-enhancing
still difficult to estimate due to small sample sizes and (Funkiewiez et al., 2003; Schneider et al., 2003)
possible biases in reporting on adverse events. The and high stimulation amplitudes may cause laughing
incidence of depression in the first postoperative spells (Krack et al., 2001; Funkiewiez et al., 2003;
months has been up to 25% in some reports (Volkmann Schneider et al., 2003). There is currently no
et al., 2001; Berney et al., 2002), but there may be a evidence for other emotional or behavioral effects
considerable overlap with apathy, that can present after of HFS within STN. The two spectacular cases
surgery as reduced drive without sadness. Depression of stimulation-induced depression and aggressive
was associated with suicidal ideation in some patients behavior resulted from misplaced electrodes
(Berney et al., 2002; Doshi et al., 2002). One center (Bejjani et al., 1999, 2002) within the substantia
described a suicide rate of 4% following DBS for PD, nigra pars reticulata and the triangle of Sano.
which is probably not representative for the therapy in 4. Successful surgery reduces disability and may
general. More recently, Foncke et al. (2006) reported enable the patient to regain independence. This
on 2 suicides out of 16 patients that were included in a may affect partnership, social bonds and profes-
clinical trial for pallidal DBS for dystonia. This observa- sional life and, at the same time, cause a loss of
tion indicates that suicide risk after DBS may not be primary and secondary gains from the illness
associated with specific targets or indications, but rather (Perozzo et al., 2001). The psychological and beha-
with more general characteristics of the patient group vioral consequences of social readaptation have
suffering from long-standing and severe motor disability been extensively studied in patients undergoing
and often regarding DBS as a last-resort treatment. epilepsy surgery. The term burden of normality
Manic disorders are less frequent, somewhere describes a syndrome of social maladjustment caus-
around 25% (Kulisevsky et al., 2002; Romito et al., ing most of the psychiatric problems after successful
2002a; Daniele et al., 2003; Herzog et al., 2003a) after treatment (Wilson et al., 2001). Whether a similar
STN DBS. Other reports about behavioral abnormal- concept applies to movement disorder surgery
ities after DBS of the STN include hypersexuality, remains to be determined.
gambling or aggressiveimpulsive behavior in indivi-
dual patients (Houeto et al., 2002). Many of the
patients with behavioral abnormalities after surgery 43.5. Patient selection
had a pre-existing psychiatric condition (Houeto
et al., 2002). Therefore, careful preoperative psychia- DBS has not been evaluated for other forms of par-
tric assessment is indicated to identify patients at risk kinsonism than idiopathic PD. From the experience
and to follow these patients closely after DBS. in few patients with progressive supranuclear palsy,
The high incidence of psychiatric problems after STN multiple system atrophy or other atypical parkinso-
DBS is most likely multifactorial, but the relevance of nian syndroms who underwent DBS, one can con-
the individual factors still needs to be determined: clude that the overall progression of these disorders
rapidly counteracts a possible transient benefit from
1. PD is a neuropsychiatric disease most frequently surgery. Up to 10% of the therapeutic failures after
associated with depression and anxiety. Mood DBS may result from an inappropriate diagnosis of
and behavioral abnormalities after surgery often PD (Okun et al., 2005), which underlines the impor-
reflect a reactivation of a pre-existing psychiatric tance of involving movement disorder specialists in
condition (Houeto et al., 2002). the selection process.
DEEP BRAIN STIMULATION 271
The objective of the selection process in general is to agonist, clozapine up to 75 mg/day and propanolol or
identify those individuals in whom the expected benefit primidone in cases of predominant action tremor
will outlast the inherent risks of the surgical procedure. (Deuschl and Volkmann, 2002).
Functional stereotactic surgery strives to improve Thalamic DBS is effective in reducing tremor, but
motor function and to reduce disability by alleviating does not improve other symptoms of PD. As an alter-
symptoms of a movement disorder. Benefit in the native, STN DBS should be advised in younger
context of movement disorder surgery is therefore a patients who are at risk of developing akinesia and
complex variable, addressing the multidimensional levodopa-related motor complications in the further
components of health and health-related well-being course of disease or in patients already suffering from
in an individual. In the World Health Organization these symptoms. Older patients (> 70 years) with tre-
(2005) WHO International Classification of Function- mor-dominant PD are less likely to progress and may
ing, Disability and Health (ICF), these health-related undergo thalamic DBS with good benefit. The advan-
domains are described from the perspective of the tage of thalamic DBS in these cases lies in a more
body, the individual and society. Disability is used rapid and predictable therapeutic response and the less
as an umbrella term for impairments of body struc- complicated adjustment of stimulation and medication
ture or function, resulting activity limitations and par- compared to STN DBS.
ticipation restrictions (social handicap). The degree of
activity limitations and social handicap a patient may 43.5.2. Advanced Parkinsons disease
experience from a physical impairment is influenced
by individual factors (such as profession and family Pallidal or subthalamic DBS are symptomatic treat-
status) within this framework. ments for motor fluctuations and dyskinesias in
The challenge that any neurologist or neurosurgeon advanced PD. Levodopa-resistant symptoms of PD
is facing in the selection process for movement disor- do not respond to DBS either. Hence, ideal candi-
der surgery is: dates should suffer from idiopathic PD with an excel-
lent levodopa response but side-effects of long-term
1. to determine whether the target symptom for sur-
medical treatment. Dementia, acute psychosis and
gery is the predominant source of disability in this
major depression are usually exclusion criteria.
patient,
The general health condition of the patient needs to
2. to identify other potential sources of disability,
be good enough to withstand the operation and to
3. to estimate the likelihood of improving the target
maintain cooperation during prolonged awake-sur-
symptom by surgery,
gery. The presurgical levodopa test helps to predict
4. to estimate the individual risk of suffering from
the individual response profile of a patient to bilateral
complications,
STN or GPi stimulation (Welter et al., 2002). Symp-
5. to formulate realistic goals for the rehabilitation of
toms other than dyskinesias or tremor which persist
the patient on the different levels of functioning,
during best on after taking a challenging dose of
6. to relate the patients own expectation from sur-
levodopa (11.5 times the equivalent of the regular
gery to these goals and to correct unrealistic
antiparkinsonian morning medication in the form of
expectations.
short-acting, soluble levodopa) are less likely to bene-
Patients need to be informed about their individual fit from DBS. Most centers have operated primarily on
risk/benefit analysis and possible alternative treatments. patients suffering from young-onset PD, because they
fulfill these criteria best. Older patients may also ben-
43.5.1. Tremor efit from surgery, but levodopa-resistant symptoms are
more often encountered in this group; axial symptoms
Tremor is seldom the most incapacitating symptom in may respond less to DBS than in younger patients
PD. Social embarrassment may be the driving force for (Russmann et al., 2004), surgical adverse events are
patients seeking surgical treatment rather than func- more frequent, motor rehabilitation is slower and
tional impairment. In our view, the involved risks do comorbidities (such as orthopedic problems secondary
not justify surgery in these cases unless the patient is to a parkinsonian postural or gait disorder) may limit
experiencing relevant disability from the social restric- the degree of functional restitution.
tions implied by tremor. Before advising surgery, the The treating physician should be informed about the
following medical therapies should have been tried: patients personal expectations about surgery and cor-
levodopa with a sufficiently high daily dose (1000 rect unrealistic perspectives. Finally, the patient needs
1500 mg/day) for a period of at least 4 weeks to to understand that the therapeutic benefits of DBS for
observe the long-term levodopa effect, a dopamine advanced PD are not immediately obtained after
272 J. VOLKMANN AND G. DEUSCHL
surgery. Programming the device and adjusting medica-
Benabid AL, Pollak P, Gervason C et al. (1991). Long-term
tion may be time-consuming and tedious and the patient suppression of tremor by chronic stimulation of the ventral
must be ready to cooperate during this process. intermediate thalamic nucleus. Lancet 337 (8738): 403406.
Benazzouz A, Piallat B, Pollak P et al. (1995). Responses of
43.6. Conclusion substantia nigra pars reticulata and globus pallidus com-
plex to high frequency stimulation of the subthalamic
DBS is one of the most promising new therapies for nucleus in rats: electrophysiological data. Neurosci Lett
the treatment of PD. There is sufficient evidence in 189 (2): 7780.
the literature that DBS of the STN is an effective and Bergman H, Deuschl G (2002). Pathophysiology of Parkinsons
relatively safe therapy for all cardinal symptoms of disease: from clinical Neurology to basic neuroscience and
back. Mov Disord 17 (Suppl 3): S28S40.
PD and levodopa-induced motor complications. The
Berney A, Vingerhoets F, Perrin A et al. (2002). Effect on
improvements in motor symptoms are so profound that
mood of subthalamic DBS for Parkinsons disease: a conse-
quality of life is significantly improved by STN DBS. cutive series of 24 patients. Neurology 59 (9): 14271429.
With respect to possible side-effects of DBS, especially Beurrier C, Bioulac B, Audin J et al. (2001). High-frequency
behavioral and psychiatric adverse events, larger regis- stimulation produces a transient blockade of voltage-gated
tries or multicenter trials are needed, because single currents in subthalamic neurons. J Neurophysiol 85 (4):
centers would take several years to recruit sufficient data 13511356.
for a safety evaluation. Finally, future trials have to clar- Binder DK, Rau G, Starr PA (2003). Hemorrhagic complica-
ify the differential indications for the three targets in tions of microelectrode-guided deep brain stimulation.
which DBS can be applied for treating PD (Follett Stereotact Funct Neurosurg 80 (14): 2831.
et al., 2005). Brock LG, Coombs JS, Eccles JC (1952). The recording of
potentials from motoneurons with intracellular electrode.
J Physiol 117: 431460.
References Broggi G, Franzini A, Ferroli P et al. (2001). Effect of
bilateral subthalamic electrical stimulation in Parkinsons
Alegret M, Junque C, Valldeoriola F et al. (2001). Effects of disease. Surg Neurol 56 (2): 8994; discussion 9496.
bilateral subthalamic stimulation on cognitive function in Brown P, Williams D (2005). Basal ganglia local field poten-
Parkinson disease. Arch Neurol 58 (8): 12231227. tial activity: character and functional significance in the
Allert N, Volkmann J, Dotse S et al. (2001). Effects of bilat- human. Clin Neurophysiol 116 (11): 25102519.
eral pallidal or subthalamic stimulation on gait in advanced Burchiel KJ, Anderson VC, Favre J et al. (1999). Comparison
Parkinsons disease. Mov Disord 16 (6): 10761085. of pallidal and subthalamic nucleus deep brain stimulation
Anderson TR, Hu B, Iremonger K et al. (2006). Selective for advanced Parkinsons disease: results of a randomized,
attenuation of afferent synaptic transmission as a mechan- blinded pilot study. Neurosurgery 45 (6): 13751382.
ism of thalamic deep brain stimulation-induced tremor Capecci M, Ricciuti RA, Burini D et al. (2005). Functional
arrest. J Neurosci 26 (3): 841850. improvement after subthalamic stimulation in Parkinsons
Anderson VC, Burchiel KJ, Hogarth P et al. (2005). Pallidal disease: a non-equivalent controlled study with 1224
vs subthalamic nucleus deep brain stimulation in Parkin- month follow up. J Neurol Neurosurg Psychiatry 76 (6):
son disease. Arch Neurol 62 (4): 554560. 769774.
Ardouin C, Klinger H, Limousin P et al. (1999). The effect Ceballos Baumann AO, Boecker H, Bartenstein P et al.
of bilateral subthalamic nucleus stimulation on cognitive (1999). A positron emission tomographic study of subtha-
functions [abstract]. Neurology 52 (Suppl 2): A514. lamic nucleus stimulation in Parkinson disease: enhanced
Bejjani B, Damier P, Arnulf I et al. (1997). Pallidal stimula- movement-related activity of motor-association cortex
tion for Parkinsons disease. Two targets? [see comments]. and decreased motor cortex resting activity. Arch Neurol
Neurology 49 (6): 15641569. 56 (8): 9971003.
Bejjani BP, Damier P, Arnulf I et al. (1999). Transient acute Daniele A, Albanese A, Contarino MF et al. (2003). Cogni-
depression induced by high-frequency deep-brain stimula- tive and behavioural effects of chronic stimulation of the
tion. N Engl J Med 340 (19): 14761480. subthalamic nucleus in patients with Parkinsons disease.
Bejjani BP, Arnulf I, Demeret S et al. (2000a). Levodopa- J Neurol Neurosurg Psychiatry 74 (2): 175182.
induced dyskinesias in Parkinsons disease: is sensitization Davis KD, Taub E, Houle S et al. (1997). Globus pallidus sti-
reversible? Ann Neurol 47 (5): 655658. mulation activates the cortical motor system during allevia-
Bejjani BP, Gervais D, Arnulf I et al. (2000b). Axial parkin- tion of parkinsonian symptoms. Nat Med 3 (6): 671674.
sonian symptoms can be improved: the role of levodopa Deiber MP, Pollak P, Passingham R et al. (1993). Thalamic
and bilateral subthalamic stimulation. J Neurol Neurosurg stimulation and suppression of parkinsonian tremor. Evi-
Psychiatry 68 (5): 595600. dence of a cerebellar deactivation using positron emission
Bejjani BP, Houeto JL, Hariz M et al. (2002). Aggressive tomography. Brain 116 (Pt 1): 267279.
behavior induced by intraoperative stimulation in the tri- DeLong MR (1990). Primate models of movement disorders
angle of Sano. Neurology 59 (9): 14251427. of basal ganglia origin. Trends Neurosci 13: 281285.
DEEP BRAIN STIMULATION 273

Delwaide PJ, Pepin JL, De Pasqua V et al. (2000). Projections blinded assessments at one year follow up. J Neurol Neu-
from basal ganglia to tegmentum: a subcortical route for rosurg Psychiatry 75 (9): 12551259.
explaining the pathophysiology of Parkinsons disease Funkiewiez A, Ardouin C, Krack P et al. (2003). Acute psycho-
signs? J Neurol 247 (Suppl 2): 7581. tropic effects of bilateral subthalamic nucleus stimulation
Deuschl G, Volkmann J (2002). Tremors: differential diagno- and levodopa in Parkinsons disease. Mov Disord 18 (5):
sis, physiology and pharmacology. In: J Jankovic, E Tolosa 524530.
(Eds.), Parkinsons Disease and Movement Disorders. Ghika J, Villemure JG, Fankhauser H et al. (1998). Efficiency
4th edn., Lippincott Williams and Wilkins, Baltimore, and safety of bilateral contemporaneous pallidal stimulation
pp. 270290. (deep brain stimulation) in levodopa-responsive patients
Deuschl G, Wilms H, Krack P et al. (1999). Function of the with Parkinsons disease with severe motor fluctuations:
cerebellum in Parkinsonian rest tremor and Holmes tremor. a 2-year follow-up review. J Neurosurg 89 (5): 713718.
Ann Neurol 46 (1): 126128. Goodman RR, Kim B, McClelland S 3rd et al. (2006). Opera-
Doshi PK, Chhaya N, Bhatt MH (2002). Depression leading tive techniques and morbidity with subthalamic nucleus
to attempted suicide after bilateral subthalamic nucleus deep brain stimulation in 100 consecutive patients with
stimulation for Parkinsons disease. Mov Disord 17 (5): advanced Parkinsons disease. J Neurol Neurosurg Psychia-
10841085. try 77 (1): 1217.
Dostrovsky JO, Levy R, Wu JP et al. (2000). Microstimula- Gross C, Rougier A, Guehl D et al. (1997). High-frequency
tion-induced inhibition of neuronal firing in human globus stimulation of the globus pallidus internalis in Parkinsons
pallidus. J Neurophysiol 84 (1): 570574. disease: a study of seven cases. J Neurosurg 87 (4): 491498.
Drapier S, Raoul S, Drapier D et al. (2005). Only physical Hamel W, Fietzek U, Morsnowski A et al. (2003). Deep brain
aspects of quality of life are significantly improved by stimulation of the subthalamic nucleus in Parkinsons
bilateral subthalamic stimulation in Parkinsons disease. disease: evaluation of active electrode contacts. J Neurol
J Neurol 252 (5): 583588. Neurosurg Psychiatry 74: 10361046.
Dujardin K, Defebvre L, Krystkowiak P et al. (2001). Influ- Hariz MI, Fodstad H (1999). Do microelectrode techniques
ence of chronic bilateral stimulation of the subthalamic increase accuracy or decrease risks in pallidotomy and
nucleus on cognitive function in Parkinsons disease. deep brain stimulation? A critical review of the literature.
J Neurol 248 (7): 603611. Stereotact Funct Neurosurg 72 (24): 157169.
Durif F, Lemaire JJ, Debilly B et al. (2002). Long-term fol- Hariz MI, Shamsgovara P, Johansson F et al. (1999). Toler-
low-up of globus pallidus chronic stimulation in advanced ance and tremor rebound following long-term chronic tha-
Parkinsons disease. Mov Disord 17 (4): 803807. lamic stimulation for Parkinsonian and essential tremor.
Eidelberg D, Moeller JR, Ishikawa T et al. (1996). Regional Stereotact Funct Neurosurg 72 (24): 208218.
metabolic correlates of surgical outcome following unilat- Hashimoto T, Elder CM, Okun MS et al. (2003). Stimulation
eral pallidotomy for Parkinsons disease. Ann Neurol of the subthalamic nucleus changes the firing pattern of
39 (4): 450459. pallidal neurons. J Neurosci 23 (5): 19161923.
Esselink RA, de Bie RM, de Haan RJ et al. (2004). Unilateral Herzog J, Reiff J, Krack P et al. (2003a). Manic episode with
pallidotomy versus bilateral subthalamic nucleus stimula- psychotic symptoms induced by subthalamic nucleus
tion in PD: a randomized trial. Neurology 62 (2): 201207. stimulation in a patient with Parkinsons disease. Mov
Faist M, Xie J, Kurz D et al. (2001). Effect of bilateral sub- Disord 18 (11): 13821384.
thalamic nucleus stimulation on gait in Parkinsons Herzog J, Volkmann J, Krack P et al. (2003b). Two year
disease. Brain 124 (Pt 8): 15901600. follow up of subthalamic deep brain stimulation in Parkin-
Favre J, Taha JM, Burchiel KJ (2002). An analysis of the sons disease. Mov Disord 18 (11): 13321337.
respective risks of hematoma formation in 361 consecu- Herzog J, Fietzek U, Hamel W et al. (2004). Most effective
tive morphological and functional stereotactic procedures. stimulation site in subthalamic deep brain stimulation for
Neurosurgery 50 (1): 4856; discussion 5657. Parkinsons disease. Mov Disord 19 (9): 10501054.
Figueiras-Mendez R, Regidor I, Riva-Meana C et al. (2002). Houeto JL, Damier P, Bejjani PB et al. (2000). Subthalamic
Further supporting evidence of beneficial subthalamic stimu- stimulation in Parkinson disease: a multidisciplinary
lation in Parkinsons patients. Neurology 58 (3): 469470. approach. Arch Neurol 57 (4): 461465.
Fogelson N, Kuhn AA, Silberstein P et al. (2005). Frequency Houeto JL, Mesnage V, Mallet L et al. (2002). Behavioural
dependent effects of subthalamic nucleus stimulation in disorders, Parkinsons disease and subthalamic stimula-
Parkinsons disease. J Neurophysiol 382 (12): 59. tion. J Neurol Neurosurg Psychiatry 72 (6): 701707.
Follett K, Weaver F, Stern M et al. (2005). Multisite rando- Iacono RP, Carlson JD, Kuniyoshi SM et al. (1997). Electro-
mized trial of deep brain stimulation. Arch Neurol 62 (10): physiologic target localization in posteroventral pallidot-
16431644. omy. Acta Neurochir (Wien) 139 (5): 433441.
Foncke EM, Schuurman PR, Speelman JD (2006). Suicide Iansek R, Rosenfeld JV, Huxham FE (2002). Deep brain
after deep brain stimulation of the internal globus pallidus stimulation of the subthalamic nucleus in Parkinsons
for dystonia. Neurology 66 (1): 142143. disease. Med J Aust 177 (3): 142146.
Ford B, Winfield L, Pullman SL et al. (2004). Subthalamic Jaggi JL, Umemura A, Hurtig HI et al. (2004). Bilateral stimu-
nucleus stimulation in advanced Parkinsons disease: lation of the subthalamic nucleus in Parkinsons disease:
274 J. VOLKMANN AND G. DEUSCHL

surgical efficacy and prediction of outcome. Stereotact Laitinen LV, Bergenheim AT, Hariz MI (1992). Leksells
Funct Neurosurg 82 (23): 104114. posteroventral pallidotomy in the treatment of Parkinsons
Jahanshahi M, Ardouin CM, Brown RG et al. (2000). The disease. J Neurosurg 76 (1): 5361.
impact of deep brain stimulation on executive function Landi A, Antonini A, Parolin M et al. (2003). [Chronic sub-
in Parkinsons disease. Brain 123 (Pt 6): 11421154. thalamus stimulation for the treatment of Parkinsons dis-
Just H, Ostergaard K (2002). Health-related quality of life in ease. Analysis of results by classes of symptoms and
patients with advanced Parkinsons disease treated with adverse effects]. J Neurosurg Sci 47 (1 Suppl 1): 2427.
deep brain stimulation of the subthalamic nuclei. Mov Lanotte MM, Rizzone M, Bergamasco B et al. (2002). Deep
Disord 17 (3): 539545. brain stimulation of the subthalamic nucleus: anatomical,
Katayama Y, Kasai M, Oshima H et al. (2001). Subthalamic neurophysiological, and outcome correlations with the
nucleus stimulation for Parkinson disease: benefits effects of stimulation. J Neurol Neurosurg Psychiatry
observed in levodopa-intolerant patients. J Neurosurg 72 (1): 5358.
95 (2): 213221. Lenz FA, Kwan HC, Martin RL et al. (1994). Single unit
Kitagawa M, Murata J, Uesugi H et al. (2005). Two-year fol- analysis of the human ventral thalamic nuclear group. Tre-
low-up of chronic stimulation of the posterior subthalamic mor-related activity in functionally identified cells. Brain
white matter for tremor-dominant Parkinsons disease. 117 (Pt 3): 531543.
Neurosurgery 56 (2): 281289; discussion 281289. Lezcano E, Gomez-Esteban JC, Zarranz JJ et al. (2004).
Kleiner-Fisman G, Saint-Cyr JA, Miyasaki J et al. (2002). Improvement in quality of life in patients with advanced
Subthalamic DBS replaces levodopa in Parkinsons dis- Parkinsons disease following bilateral deep-brain stimula-
ease. Neurology 59 (8): 12931294. tion in subthalamic nucleus. Eur J Neurol 11 (7): 451454.
Krack P, Benazzouz A, Pollak P et al. (1998a). Treatment of Limousin P, Krack P, Pollak P et al. (1998). Electrical stimu-
tremor in Parkinsons disease by subthalamic nucleus lation of the subthalamic nucleus in advanced Parkinsons
stimulation. Mov Disord 13 (6): 907914. disease. N Engl J Med 339: 11051111.
Krack P, Pollak P, Limousin P et al. (1998b). Opposite motor Limousin P, Speelman JD, Gielen F et al. (1999). Multicentre
effects of pallidal stimulation in Parkinsons disease. Ann European study of thalamic stimulation in parkinsonian
Neurol 43 (2): 180192. and essential tremor. J Neurol Neurosurg Psychiatry
Krack P, Pollak P, Limousin P et al. (1998c). Subthalamic 66 (3): 289296.
nucleus or internal pallidum stimulation in young onset Loher TJ, Burgunder JM, Pohle T et al. (2002). Long-term
Parkinsons disease. Brain 121: 451457. pallidal deep brain stimulation in patients with advanced
Krack P, Pollak P, Limousin P et al. (1999). From off-period Parkinson disease: 1-year follow-up study. J Neurosurg
dystonia to peak-dose chorea. The clinical spectrum of 96 (5): 844853.
varying subthalamic nucleus activity. Brain 122 (Pt 6): Lopiano L, Rizzone M, Bergamasco B et al. (2001). Deep
11331146. brain stimulation of the subthalamic nucleus: clinical
Krack P, Kumar R, Ardouin C et al. (2001). Mirthful laugh- effectiveness and safety. Neurology 56 (4): 552554.
ter induced by subthalamic nucleus stimulation. Mov Dis- Lozano AM, Lang AE, Hutchison WD et al. (1997). Micro-
ord 16 (5): 867875. electrode recording-guided posteroventral pallidotomy in
Krack P, Dostrovsky J, Ilinsky I et al. (2002). Surgery of the patients with Parkinsons disease. Adv Neurol 74: 167174.
motor thalamus: problems with the present nomenclatures. Lyons KE, Pahwa R (2005). Long-term benefits in quality of
Mov Disord 17 (Suppl 3): S2S8. life provided by bilateral subthalamic stimulation in patients
Krack P, Batir A, Van Blercom N et al. (2003). Five-year with Parkinson disease. J Neurosurg 103 (2): 252255.
follow-up of bilateral stimulation of the subthalamic Lyons KE, Wilkinson SB, Overman J et al. (2004). Surgical
nucleus in advanced Parkinsons disease. N Engl J Med and hardware complications of subthalamic stimulation:
349 (20): 19251934. a series of 160 procedures. Neurology 63 (4): 612616.
Krause M, Fogel W, Heck A et al. (2001). Deep brain stimu- Marsden CD, Obeso JA (1994). The functions of the basal
lation for the treatment of Parkinsons disease: subthala- ganglia and the paradox of stereotaxic surgery in Parkinsons
mic nucleus versus globus pallidus internus. J Neurol disease. Brain 117 (Pt 4): 877897.
Neurosurg Psychiatry 70 (4): 464470. Martinez-Martin P, Valldeoriola F, Tolosa E et al. (2002).
Kulisevsky J, Berthier ML, Gironell A et al. (2002). Mania Bilateral subthalamic nucleus stimulation and quality of
following deep brain stimulation for Parkinsons disease. life in advanced Parkinsons disease. Mov Disord 17 (2):
Neurology 59 (9): 14211424. 372377.
Kumar R, Lozano AM, Montgomery E et al. (1998). Pallidot- McIntyre CC, Mori S, Sherman DL et al. (2004a). Electric
omy and deep brain stimulation of the pallidum and sub- field and stimulating influence generated by deep brain
thalamic nucleus in advanced Parkinsons disease. Mov stimulation of the subthalamic nucleus. Clin Neurophysiol
Disord 13 (Suppl 1): 7382. 115 (3): 589595.
Lagrange E, Krack P, Moro E et al. (2002). Bilateral subtha- McIntyre CC, Savasta M, Walter BL et al. (2004b). How
lamic nucleus stimulation improves health-related quality does deep brain stimulation work? Present understanding
of life in PD. Neurology 59 (12): 19761978. and future questions. J Clin Neurophysiol 21 (1): 4050.
DEEP BRAIN STIMULATION 275

Mehdorn HM, Pinsker MO, Volkmann J et al. (2005). Deep Pahwa R, Wilkinson SB, Overman J et al. (2003). Bilateral
brain stimulation for idiopathic or secondary movement subthalamic stimulation in patients with Parkinson dis-
disorders. Acta Neurochir Suppl 93 (Suppl): 105111. ease: long-term follow up. J Neurosurg 99 (1): 7177.
Merello M, Nouzeilles MI, Kuzis G et al. (1999). Unilateral Pahwa R, Wilkinson SB, Overman J et al. (2005). Preopera-
radiofrequency lesion versus electrostimulation of postero- tive clinical predictors of response to bilateral subthalamic
ventral pallidum: a prospective randomized comparison. stimulation in patients with Parkinsons disease. Stereotact
Mov Disord 14 (1): 5056. Funct Neurosurg 83 (23): 8083.
Minguez-Castellanos A, Escamilla-Sevilla F, Katati MJ et al. Parker F, Tzourio N, Blond S et al. (1992). Evidence for a
(2005). Different patterns of medication change after sub- common network of brain structures involved in parkinso-
thalamic or pallidal stimulation for Parkinsons disease: nian tremor and voluntary repetitive movement. Brain Res
target related effect or selection bias? J Neurol Neurosurg 584 (12): 1117.
Psychiatry 76 (1): 3439. Peppe A, Pierantozzi M, Bassi A et al. (2004). Stimulation of
Molinuevo JL, Valldeoriola F, Tolosa E et al. (2000). Levo- the subthalamic nucleus compared with the globus palli-
dopa withdrawal after bilateral subthalamic nucleus stimu- dus internus in patients with Parkinson disease. J Neuro-
lation in advanced Parkinson disease. Arch Neurol 57 (7): surg 101 (2): 195200.
983988. Perozzo P, Rizzone M, Bergamasco B et al. (2001). Deep
Montgomery EB Jr, Baker KB (2000). Mechanisms of deep brain stimulation of subthalamic nucleus: behavioural
brain stimulation and future technical developments. modifications and familiar relations. Neurol Sci 22 (1):
Neurol Res 22 (3): 259266. 8182.
Moro E, Scerrati M, Romito LM et al. (1999). Chronic subtha- Pillon B, Ardouin C, Damier P et al. (2000). Neuropsy-
lamic nucleus stimulation reduces medication requirements chological changes between off and on STN or GPi
in Parkinsons disease. Neurology 53 (1): 8590. stimulation in Parkinsons disease. Neurology 55 (3):
Moro E, Esselink RA, Van Blercom N et al. (2000). Bilateral 411418.
subthalamic nucleus stimulation in a parkinsonian patient Pinter MM, Alesch F, Murg M et al. (1999). Deep brain sti-
with previous unilateral pallidotomy and thalamotomy. mulation of the subthalamic nucleus for control of extra-
Mov Disord 15 (4): 753755. pyramidal features in advanced idiopathic Parkinsons
Murata J, Kitagawa M, Uesugi H et al. (2003). Electrical disease: one year follow-up. J Neural Transm 106 (78):
stimulation of the posterior subthalamic area for the treat- 693709.
ment of intractable proximal tremor. J Neurosurg 99 (4): Plaha P, Patel NK, Gill SS (2004). Stimulation of the subtha-
708715. lamic region for essential tremor. J Neurosurg 101 (1):
Nandi D, Aziz TZ, Giladi N et al. (2002a). Reversal of aki- 4854.
nesia in experimental parkinsonism by GABA antagonist Potter M, Illert M, Wenzelburger R et al. (2004). The effect
microinjections in the pedunculopontine nucleus. Brain of subthalamic nucleus stimulation on autogenic inhibition
125 (Pt 11): 24182430. in Parkinson disease. Neurology 63 (7): 12341239.
Nandi D, Liu X, Winter JL et al. (2002b). Deep brain stimula- Ranck JB (1975). Which elements are excited in electrical
tion of the pedunculopontine region in the normal non-human stimulation of mammalian central nervous system?
primate. J Clin Neurosci 9 (2): 170174. A review. Brain Res 98: 417440.
Ogura M, Nakao N, Nakai E et al. (2004). The mechanism Rehncrona S, Johnels B, Widner H et al. (2003). Long-term
and effect of chronic electrical stimulation of the globus efficacy of thalamic deep brain stimulation for tremor:
pallidus for treatment of Parkinson disease. J Neurosurg double-blind assessments. Mov Disord 18 (2): 163170.
100 (6): 9971001. Rodriguez MC, Guridi OJ, Alvarez L et al. (1998). The
Oh MY, Kim SH, Abosch A et al. (2001). Hardware related subthalamic nucleus and tremor in Parkinsons disease.
complications of deep brain stimulaiton in 100 consecutive Mov Disord 13 (Suppl 3): 111118.
electrodes. J Neurosurg 94: (Proceedings of the AANS Rodriguez-Oroz MC, Rodriguez M, Guridi J et al. (2001).
meeting) 412A. The subthalamic nucleus in Parkinsons disease: somatoto-
Okun MS, Tagliati M, Pourfar M et al. (2005). Management pic organization and physiological characteristics. Brain
of referred deep brain stimulation failures: a retrospective 124 (Pt 9): 17771790.
analysis from 2 movement disorders centers. Arch Neurol Rodriguez-Oroz MC, Obeso JA, Lang AE et al. (2005).
62 (8): 12501255. Bilateral deep brain stimulation in Parkinsons disease:
Ostergaard K, Sunde N, Dupont E (2002). Effects of bilateral a multicentre study with 4 years follow-up. Brain
stimulation of the subthalamic nucleus in patients with 128 (Pt 10): 22402249.
severe Parkinsons disease and motor fluctuations. Mov Romito LM, Raja M, Daniele A et al. (2002a). Transient
Disord 17 (4): 693700. mania with hypersexuality after surgery for high frequency
Pahapill PA, Lozano AM (2000). The pedunculopontine nuc- stimulation of the subthalamic nucleus in Parkinsons
leus and Parkinsons disease. Brain 123 (Pt 9): 17671783. disease. Mov Disord 17 (6): 13711374.
Pahwa R, Wilkinson S, Smith D et al. (1997). High-fre- Romito LM, Scerrati M, Contarino MF et al. (2002b). Long-
quency stimulation of the globus pallidus for the treatment term follow up of subthalamic nucleus stimulation in
of Parkinsons disease. Neurology 49 (1): 249253. Parkinsons disease. Neurology 58 (10): 15461550.
276 J. VOLKMANN AND G. DEUSCHL

Russmann H, Ghika J, Villemure JG et al. (2004). Sub- The Deep-Brain Stimulation for Parkinsons Disease Study
thalamic nucleus deep brain stimulation in Parkinson dis- Group (2001). Deep-brain stimulation of the subthalamic
ease patients over age 70 years. Neurology 63 (10): nucleus or the pars interna of the globus pallidus in
19521954. Parkinsons disease. N Engl J Med 345 (13): 956963.
Saint-Cyr JA, Trepanier LL, Kumar R et al. (2000). Neurop- Thobois S, Mertens P, Guenot M et al. (2002). Subthalamic
sychological consequences of chronic bilateral stimulation nucleus stimulation in Parkinsons disease: clinical eva-
of the subthalamic nucleus in Parkinsons disease. Brain luation of 18 patients. J Neurol 249 (5): 529534.
123 (Pt 10): 20912108. Trepanier LL, Kumar R, Lozano AM et al. (2000). Neurop-
Saint-Cyr JA, Hoque T, Pereira LC et al. (2002). Localization sychological outcome of GPi pallidotomy and GPi or
of clinically effective stimulating electrodes in the human STN deep brain stimulation in Parkinsons disease. Brain
subthalamic nucleus on magnetic resonance imaging. Cogn 42 (3): 324347.
J Neurosurg 97 (5): 11521166. Tronnier VM, Fogel W, Kronenbuerger M et al. (1997). Pal-
Samuel M, Ceballos-Baumann AO, Turjanski N et al. (1997). lidal stimulation: an alternative to pallidotomy? J Neuro-
Pallidotomy in Parkinsons disease increases supplementary surg 87: 700705.
motor area and prefrontal activation during performance of Troster AI, Fields JA, Wilkinson S et al. (2003). Effect of
volitional movements an H2(15)O PET study. Brain motor improvement on quality of life following subthala-
120 (Pt 8): 13011313. mic stimulation is mediated by changes in depressive
Schneider F, Habel U, Volkmann J et al. (2003). Deep brain symptomatology. Stereotact Funct Neurosurg 80 (14):
stimulation of the subthalamic nucleus enhances emo- 4347.
tional processing in Parkinson disease. Arch Gen Psychia- Urbano FJ, Leznki E, Linas RR (2002). Cortical activation
try 60 (3): 296302. patterns evoked by afferent axons stimuli at different fre-
Schupbach WM, Chastan N, Welter ML et al. (2005). Stimu- quencies: an in vitro voltage-sensitive dye imaging study.
lation of the subthalamic nucleus in Parkinsons disease: Thalamus Rel Syst 1: 371378.
a 5 year follow up. J Neurol Neurosurg Psychiatry Valldeoriola F, Pilleri M, Tolosa E et al. (2002). Bilateral
76 (12): 16401644. subthalamic stimulation monotherapy in advanced Parkin-
Schuurman PR, Bosch DA, Bossuyt PM et al. (2000). sons disease: long-term follow-up of patients. Mov Dis-
A comparison of continuous thalamic stimulation and ord 17 (1): 125132.
thalamotomy for suppression of severe tremor. N Engl J Varma TR, Fox SH, Eldridge PR et al. (2003). Deep brain
Med 342 (7): 461468. stimulation of the subthalamic nucleus: effectiveness in
Schuurman P, Bosch D, Speelman J (2001). Thalamic stimu- advanced Parkinsons disease patients previously reliant
lation versus thalamotomy: long-term follow-up. Parkin- on apomorphine. J Neurol Neurosurg Psychiatry 74 (2):
sonism Relat Disord 7 (Suppl): S85(Abstract). 170174.
Simuni T, Jaggi JL, Mulholland H et al. (2002). Bilateral stimu- Velasco F, Jimenez F, Perez ML et al. (2001). Electrical sti-
lation of the subthalamic nucleus in patients with Parkinson mulation of the prelemniscal radiation in the treatment of
disease: a study of efficacy and safety. J Neurosurg 96 (4): Parkinsons disease: an old target revised with new techni-
666672. ques. Neurosurgery 49 (2): 293306; discussion 306308.
Spottke EA, Volkmann J, Lorenz D et al. (2002). Evaluation Vesper J, Klostermann F, Stockhammer F et al. (2002).
of healthcare utilization and health status of patients with Results of chronic subthalamic nucleus stimulation for
Parkinsons disease treated with deep brain stimulation Parkinsons disease: a 1-year follow-up study. Surg Neu-
of the subthalamic nucleus. J Neurol 249 (6): 759766. rol 57 (5): 306311; discussion 311313.
Stolze H, Klebe S, Poepping M et al. (2001). Effects of bilat- Vingerhoets FJ, Villemure JG, Temperli P et al. (2002).
eral subthalamic nucleus stimulation on parkinsonian gait. Subthalamic DBS replaces levodopa in Parkinsons dis-
Neurology 57 (1): 144146. ease: two-year follow-up. Neurology 58 (3): 396401.
Strafella A, Ashby P, Munz M et al. (1997). Inhibition of Visser-Vandewalle V, van der Linden C, Temel Y et al.
voluntary activity by thalamic stimulation in humans: (2005). Long-term effects of bilateral subthalamic nucleus
relevance for the control of tremor. Mov Disord 12 (5): stimulation in advanced Parkinson disease: a four year fol-
727737. low-up study. Parkinsonism Relat Disord 11 (3): 157165.
Tamma F, Rampini P, Egidi M et al. (2003). Deep brain Voges J, Volkmann J, Allert N et al. (2002). Bilateral high-
stimulation for Parkinsons disease: the experience of the frequency stimulation in the subthalamic nucleus for the
Policlinico-San Paolo Group in Milan. Neurol Sci treatment of Parkinson disease: correlation of therapeutic
24 (Suppl 1): S41S42. effect with anatomical electrode position. J Neurosurg
Tavella A, Bergamasco B, Bosticco E et al. (2002). Deep 96 (2): 269279.
brain stimulation of the subthalamic nucleus in Parkinsons Volkmann J, Sturm V, Weiss P et al. (1998). Bilateral high-
disease: long-term follow-up. Neurol Sci 23 (Suppl 2): frequency stimulation of the internal globus pallidus in
S111S112. advanced Parkinsons disease. Ann Neurol 44: 953961.
Terao T, Takahashi H, Yokochi F et al. (2003). Hemorrhagic Volkmann J, Fogel W, Krack P (2000). Postoperatives
complication of stereotactic surgery in patients with move- neurologisches Management bei Stimulation des Nucleus
ment disorders. J Neurosurg 98 (6): 12411246. subthalamicus. Aktuelle Neurolog 27 (Suppl 1): 2339.
DEEP BRAIN STIMULATION 277

Volkmann J, Allert N, Voges J et al. (2001). Safety and effi- Woods SP, Fields JA, Troster AI (2002). Neuropsychological
cacy of pallidal or subthalamic nucleus stimulation in sequelae of subthalamic nucleus deep brain stimulation in
advanced PD. Neurology 56 (4): 548551. Parkinsons disease: a critical review. Neuropsychol Rev
Volkmann J, Allert N, Voges J et al. (2004). Long-term 12 (2): 111126.
results of bilateral pallidal deep brain stimulation in World Health Organization (2005). The International Classi-
Parkinsons disease. Ann Neurol 55: 871875. fication of Functioning, Disability and HealthICF.
Welter ML, Houeto JL, Tezenas du Montcel S et al. (2002). WHO, Geneva.
Clinical predictive factors of subthalamic stimulation in Zonenshayn M, Sterio D, Kelly PJ et al. (2004). Location of
Parkinsons disease. Brain 125 (Pt 3): 575583. the active contact within the subthalamic nucleus (STN) in
Wilson S, Bladin P, Saling M (2001). The burden of nor- the treatment of idiopathic Parkinsons disease. Surg Neu-
mality: concepts of adjustment after surgery for seizures. rol 62 (3): 216225; discussion 225226.
J Neurol Neurosurg Psychiatry 70 (5): 649656.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 44

Transplantation

CURT R. FREED*, W. MICHAEL ZAWADA, MAUREEN LEEHEY,


WENBO ZHOU AND ROBERT E. BREEZE

University of Colorado School of Medicine, Denver, CO, USA

44.1. Introduction to survive in large numbers because of the insurmoun-


table problem of xenograft rejection.
Neurotransplantation offers a novel treatment strategy In the USA, during the presidency of Bill Clinton,
for Parkinsons disease (PD), replacing lost dopamine there was a window of federal funding to test systema-
neurons with new cells. The fact that there are so many tically the value of embryonic dopamine cell trans-
successful therapies for PD is extraordinary. This book plants. Two double-blind studies were performed
has summarized both pharmacologic and neurosurgical (Freed et al., 2001; Olanow et al., 2003). Although
procedures, including lesions and deep brain stimula- neither study achieved significance in primary outcome
tion. Neurotransplantation is fundamentally different variables, both showed long-term survival of embryo-
from these strategies, because only neurotransplanta- nic dopamine cells. In the first of these, we found that
tion repairs a primary defect of PD, the loss of transplants significantly improved Unified Parkinsons
dopamine neurons. Disease Rating Scale (UPDRS) and Schwab and
Transplantation of dopamine neurons into humans England off scores in patients under age 60 (Freed
has developed from a comprehensive series of experi- et al., 2001). A proportion of patients in our study as
ments in animal models of PD, particularly rats. Most well as the later study by Olanow et al. (2003) devel-
principles established in the rat have proven applicable oped persistent dyskinesias in a pattern similar to that
to humans. In the rat, embryonic dopamine neurons from seen after levodopa therapy in the same patients. Thus,
a narrow window of development, 1315 days after transplants replicated the effects of levodopa, including
conception, are suitable for transplantation. For human the tendency to produce dyskinesias in patients with an
transplantation, the equivalent stage of embryonic devel- earlier history of drug-induced dyskinesias.
opment is 68 weeks after conception. All successful To explore the value of dopamine cell transplanta-
allografts of human embryonic tissue into patients with tion for PD comprehensively, a more reliable tissue
PD have come from tissue in this developmental window source must be found. Efforts to produce dopamine
(for reviews, see Clarkson and Freed, 1999; Bjorklund neurons from human embryonic stem cells are promis-
et al., 2003; Lang and Obeso, 2004). ing (Buytaert-Hoefen et al., 2004; Perrier et al., 2004;
Tissues other than fetal dopamine neurons have Yan et al., 2005). If a homogeneous supply of dopamine
been transplanted into PD patients. Autotransplants neurons can be generated, cell replacement can be
of adrenal chromaffin cells were pursued intensively systematically studied in large groups of patients, better
in the late 1980s, without success. Because the supply defining the potential of this therapeutic strategy.
of human fetal tissue is limited and variable in quality,
some researchers looked to other species as a source of 44.2. Experimental basis for
fetal dopamine neurons. Ventral mesencephalon from neurotransplantation
fetal pig was transplanted into patients who were
immunosuppressed with ciclosporin and prednisone. Developing a good rodent model of PD was critical to
Despite efforts to prevent rejection, porcine cells failed the development of neurotransplantation. Ungerstedt

*Correspondence to: Curt R. Freed, University of Colorado Health Sciences Center, Box C237, 4200 E. Ninth Avenue, Denver,
CO 80262, USA. E-mail: curt.freed@uchsc.edu, Tel: 1-303-315-8455, Fax: 1-303-315-3272.
280 C. R. FREED ET AL.
and Arbuthnott (1970) demonstrated that unilateral small groups of striatal neurons, the model does not
injection of the neurotoxin 6-hydroxydopamine assure homogeneous reinnervation by dopamine neu-
(6-OHDA) into the medial forebrain bundle of rat brain rons. Since the grafted dopamine neurons are ectopi-
can destroy dopamine neurons in the substantia nigra cally placed in the striatum and do not have normal
pars compacta with loss of dopamine nerve terminals afferents, the dopamine neurons do not receive normal
in the striatum. Nearly complete destruction of dopa- inputs regulating their own firing rates. If those inputs
mine neurons can be achieved on one side of the brain are needed for fully normal control of movement,
without damaging other neural systems or dopamine dopamine cell transplants will be limited in value.
neurons on the contralateral side. Animals maintain Based on current results in humans, as described
nearly normal eating, drinking and grooming behaviors. below, fetal cell transplants into putamen appear to
When doses of methamphetamine are administered that mimic all of the effects of levodopa. At the present
ordinarily produce stereotypic behavior (5 mg/kg time, transplants can be viewed as equivalent to a
subcutaneously), the unilaterally lesioned animals will continuous infusion of levodopa. This outcome is quite
begin circling toward their lesioned side at rates as high remarkable, given the fact that dopamine is being
as 10 rpm. Dopamine released asymmetrically from the made available only to putamen.
intact side of striatum drives the circling behavior. Only Ultrastructural studies have shown that fetal grafts
rats with >95% unilateral dopamine depletion will reinnervate denervated striatum with both host-to-graft
circle at these high rates. By contrast, the dopamine and graft-to-host synapses (Mahalik et al., 1985).
agonist apomorphine stimulates the supersensitive Dopaminergic nerve terminals synapse on dendritic
dopamine receptors in the denervated striatum and spines of medium spiny neurons. Transplanted cells
leads to circling in the direction contralateral to the synthesize and release dopamine (Schmidt et al.,
lesion. 1982). Grafted dopamine neurons exhibit electrical fir-
This rat model made it possible to explore cell ing patterns and pharmacologic responses to dopamine
replacement therapies for PD. Cells from a wide devel- agonists similar to the intrinsic dopamine cells of the
opmental range from early embryonic to adult tissue substantia nigra pars compacta of adult animals
were tested for their ability to survive in brain. Critical (Wuerthele et al., 1981).
experiments by Bjorklund and Stenevi (1979) and Prior to transplanting human embryonic dopamine
Perlow et al. (1979) were the first to show that rat neurons into patients, human cells were transplanted
mesencephalic dopamine cells transplanted into the into the rat model of PD. In ciclosporin-immunosup-
denervated striatum could survive, extend neurites pressed rats, human fetal dopamine neurons were
and stop the circling response to amphetamine as shown to survive and produce behavioral effects
dopamine concentrations were restored. equivalent to rat dopamine cells, albeit with a time
These early experiments were followed by others course delayed to 820 weeks compared to the 46
that showed that dopamine neurite outgrowth occurred weeks seen with the faster-developing rat neurites
in response to factors produced by the dopamine (Brundin et al., 1986; Stromberg et al., 1986).
denervated striatum. Only mesencephalic dopamine Xenograft transplants of human to rat predicted the
neurons will reinnervate the striatum and transplant future difficulty of xenograft transplants in humans.
growth occurs only after striatum has been denervated. Although allografts of human embryonic dopamine
The behavioral effects of transplantation in rats are neurons are not rejected following transplant, even
specific to mesencephalic dopamine cells. Rats trans- with no immunosuppression (Freed et al., 2001), xeno-
planted with serotonergic neurons from mesencephalic graft transplants of human into rat require continuous
raphe or dopamine neurons from hypothalamus have treatment with ciclosporin.
no improvement in behavior (Dunnett et al., 1988; In the search for alternatives to mesencephalic
Hudson et al., 1994). These experiments showed that dopamine neurons, cells from the adrenal medulla
innervation of the target is a specific interaction were harvested and transplanted into the rat model of
between the appropriate dopamine neuron and factors PD (Freed et al., 1981; Freed, 1983; Stromberg et al.,
released from the denervated striatum. 1985). Ordinarily, these cells produce norepinephrine
This important principle provides an argument for and epinephrine, not dopamine. Separated from
the safety of dopamine cell transplants, suggesting that adrenal cortex, adrenal medullary tissue generates a
a wide range of tissue doses could produce an accepta- substantial amount of dopamine. Although these cells
ble behavioral response since the host striatum would usually secrete neurotransmitters directly into the cir-
secrete neurotrophic factors until appropriate dopami- culation and do not resemble neurons, isolated adrenal
nergic input was achieved. Because this response is medulla cells adopt a neuronal morphology, particu-
likely to be very local, even confined to individual or larly when exposed to a source of nerve growth factor
TRANSPLANTATION 281
(NGF) (Freed, 1983; Stromberg et al., 1985). Survival difficult to obtain than human fetal tissue, the number
of cells in striatum was much better with a source of of monkey transplants reported in the literature has
NGF or equivalent neurotrophic factor. Behavioral been relatively small.
effects were not as robust as seen with mesencephalic The question of targeting transplants to putamen,
dopamine neurons (Freed, 1983). caudate or both is based on electrophysiological and
Humans and monkeys were shown to be at risk for transplant studies in monkeys. Alexander and DeLong
chemically induced PD after catastrophic incidents in (1985) used microstimulation of the caudate and puta-
drug addicts. Initially, a young man who tried to pro- men to show that the putamen had motor responses
duce synthetic narcotics inadvertently injected himself specific to parts of the body with a homonculus that
with a mixture that was subsequently shown to include resembled that of motor and sensorimotor cortex.
the neurotoxin 1-methyl-4-phenyl-1, 2, 3, 6-tetrahy- Leg and trunk movements were dorsolateral, arm
dropyridine (MPTP) (Davis et al., 1979). Shortly movements intermediate and orofacial movements
thereafter, he developed bradykinetic signs that were ventromedial. By contrast, microstimulation of
initially diagnosed as catatonic schizophrenia but later caudate produced no limb movements. Hikosaka and
rediagnosed as parkinsonism. Because he was a single colleagues (1989a) performed a series of experiments
case and the phenomenon could not be reproduced in in monkeys showing that caudate controls saccadic
rats, the report gained little attention. Had the investi- eye movements. They also showed that caudate has a
gators picked any other mammal, such as mouse, cat, role in more complex behavioral responses to reward,
dog or monkey, they would have seen a parkinsonian perhaps reflecting its connections with prefrontal
syndrome. Instead, this critical observation had to be cortex (Hikosaka et al., 1989b). The same group has
rediscovered after a group of drug addicts obtained found that monkeys with dopamine depletion of cau-
MPTP from a single dealer (Langston et al., 1983). date show impaired eye movements, similar to
From these unfortunate events came a non-human PD patients, but no abnormalities of limb movement
primate model of PD (Burns et al., 1983). Monkeys (Kori et al., 1995). Only one study in monkeys has
lesioned by MPTP showed signs of bradykinesia, directly compared transplants into caudate versus
rigidity and tremor with dopamine depletion in the putamen. In the marmoset, dopamine neurons placed
caudate and putamen. Levodopa improved these signs. in caudate reversed amphetamine-induced circling,
Although the systemically lesioned monkey closely whereas transplants introduced into putamen improved
resembled idiopathic PD, the condition was highly motor function of the contralateral limb (Annett et al.,
variable. Some animals recovered to normal, making 1995). Taken together, these experiments in monkeys
it difficult to distinguish a therapeutic intervention indicate that dopamine replacement in the putamen
from spontaneous improvement. Others, unable to feed will directly improve movement of the limbs,
themselves or maintain an upright posture, died of the whereas dopamine cell transplants into the caudate
complications of immobility. A more predictable may have effects on eye movements and complex
lesioning method was developed by Bankiewicz et al. motor behaviors.
(1986). By unilaterally infusing MPTP into the inter-
nal carotid artery, dopamine neurons on one side of 44.3. Clinical experience with adrenal
the brain could be destroyed, leading to contralateral medulla transplantation
signs of parkinsonism. As with the 6-OHDA-lesioned
rat, these animals could care for themselves. In 1987, an article by Madrazo et al. described a remark-
Transplanting fetal dopamine neurons into dener- able improvement in 2 patients after transplantation of
vated striatum reduced the parkinsonian signs in both adrenal medulla fragments recovered from the patients
systemically and unilaterally lesioned monkeys, if the own adrenals and placed into the head of the caudate on
embryonic dopamine cells were obtained from early one side of the brain. Although the clinical improvement
in embryogenesis at a developmental stage equivalent was confounded by also initiating levodopa treatment,
to days 1316 in the embryonic rat (Bakay et al., the fact that the report appeared in the New England
1985; Freed et al., 1988; Annett et al., 1994, 1997). Journal of Medicine brought a great deal of attention.
By contrast, if mesencephalic tissue from later fetal Immediately, many groups began performing trans-
stages was transplanted, cells failed to survive and ani- plants of adrenal medulla, with more than 200 patients
mals did not improve (Redmond et al., 1986; Freed receiving surgery over the next 2 years. Although these
et al., 1988). Monkey experiments were valuable for open clinical trials reported some improvement for up to
showing that the principles established in rats also 6 months, there were no sustained effects of transplant.
applied to primates. Because embryonic monkey tissue The morbidity of the surgery was significant. During
of the appropriate early gestational stage is even more recovery from the abdominal surgery on the adrenal
282 C. R. FREED ET AL.
gland, patients were often unable to take their oral Assessment Program for Intracerebral Transplantations
antiparkinsonian medications. Typically, patients spent (Langston et al., 1992).
10 days in the intensive care unit and up to 30 days in Kordower and colleagues (1995), who did bilateral
hospital. One-year mortality was 10% and 2-year mor- transplants of fragments of embryonic mesencephalon
tality was 20%. In a registry of 61 patients, only 19% into postcommissural putamen, were the first to
were judged improved 2 years after transplant (Goetz demonstrate surviving dopamine cells postmortem.
et al., 1991). Autopsy revealed few surviving adrenal The patient was immunosuppressed with ciclosporin
chromaffin cells (Hurtig et al., 1989). Since no source for 6 months and died 18 months after transplant.
of NGF had been provided, significant cell survival Large numbers of surviving dopamine neurons were
was improbable. seen in the transplant tracks.
Adrenal medulla transplants using cografts of The biggest problems with most clinical efforts
peripheral nerve as a source of trophic factors have were that patient numbers were small and trials uncon-
been conducted with reportedly better cell survival trolled. Therefore, statistically valid group compari-
and longer duration of clinical effects (Date et al., sons could not be made. Results were anecdotal
1996; Watts et al., 1997). Nonetheless, enthusiasm assertions rather than statistically valid evaluations.
based on flawed case reports turned to disappointment. Only the two clinical trials sponsored by the National
Grafting of cells from the adrenal medulla was Institutes of Health have had experimental designs
abandoned because of the lack of efficacy and the with enough patients, consistent methodology and
significant morbidity associated with the surgical control groups to make it possible to draw conclusions.
procedure itself. Our double-blind, placebo-controlled trial trans-
planted dopamine neurons from ventral mesencephalon
44.4. Clinical experience with human fetal recovered from four embryos 78 weeks postconception
dopamine cell transplantation and transplanted into four sites in the putamen, bilater-
ally under stereotaxic control. Tissue had been kept as
Since transplantation of human fetal tissue was first strands in culture for up to 4 weeks prior to transplant.
undertaken in the late 1980s, several groups have A 34-cm-long column of cells was placed in a dorsal
reported results using a variety of methods (Freed and a ventral site in putamen on each side of the brain
et al., 1990, 1992, 1995, 2001; Langston et al., 1992; via twist drill holes in the forehead under local anesthe-
Spencer et al., 1992; Widner et al., 1992; Peschanski sia. Sham surgery patients received the identical proce-
et al., 1994; Freeman et al., 1995a; Kordower et al., dure except there was no needle penetration of the brain
1995; Kopyov et al., 1996; Lindvall et al., 2001; and no tissue was deposited. All patients gave informed
Olanow et al., 2003). Comparisons among studies have consent to receive either tissue transplants or sham sur-
been difficult because technical strategies and clinical gery. All recovery of embryonic tissue from elective
evaluations have differed so widely. Tissue has been abortions was done only after women had consented to
placed in putamen as well as caudate and unilaterally the abortion and after they had given a second consent
as well as bilaterally. It has been transplanted as solid to donate fetal tissue per federal law and Institutional
tissue fragments as well as suspensions. Because trans- Review Board (IRB) requirements. There were no surgi-
plants of allografts in outbred rats and in monkeys are cal complications that led to breaking the blind. There
not rejected even without immunosuppression, the need were no infections.
for immunosuppressants was uncertain. Therefore, We found that, 12 months after surgery, transplant
some groups used immunosuppression, some did not subjects showed improvements in UPDRS motor
and others compromised with short-term antirejection off scores for the group as a whole and for the
regimens. Patients with parkinsonism provide addi- subgroup under age 60 compared to the sham surgery
tional variability. Age is an important variable, as is controls (Fig. 44.1) (Freed et al., 2001). As shown in
the specific disease in the individual patient. To define Figure 44.2, transplants were detectable in 85% of
patients as having idiopathic PD, most investigators patients by blind scoring of FDOPA PET scans and
have chosen patients who were responsive to levodopa. grew equally well in younger and older patients
Some investigators have used fluorodopa positron (Nakamura et al., 2001). Patients under the age of 60
emission tomography (FDOPA PET) to image the char- were most likely to benefit and changes in the UPDRS
acteristic pattern of dopamine depletion in putamen correlated with changes in PET signal. The signs that
with relative sparing in the caudate nucleus. No single improved were bradykinesia and rigidity. Although
clinical rating scale has been used, though there was tremor showed a trend for improvement, statistical
an effort to define an evaluation strategy using a combi- significance was not reached. For each patient, the
nation of the UPDRS scale and timed tests called Core best on state was carefully determined after the first
TRANSPLANTATION 283

All subjects Age 60 Age > 60


50 50 50
= Sham
45 = Real 45 45
Motor UPDRS "OFF"

40 40 40

35 35 35

30 30 30

25 25 25
p = 0.015 p = 0.0003 p = 0.989
20 20 20
b12 p04 p08 p12 b12 p04 p08 p12 b12 p04 p08 p12
Visit
Fig. 44.1. Changes in Unified Parkinsons Disease Rating Scale motor off scores in the first 12 months after transplant dur-
ing the double-blind phase. Results show significant improvement for the transplant group as a whole and for the younger
transplant patients, compared to the sham surgery group.

Fetal mesencephalic cell implant


FDOPA PET

Preop. Postop.

Sham surgery

Normal

Preop. Postop.
Fig. 44.2. For full color figure, see plate section. Fluorodopa positron emission tomography (FDOPA PET) scans before and after
transplantation. The left-hand panel shows the FDOPA PET signal of a normal person scanned in a horizontal plane that includes
caudate and putamen. Stored F-dopamine is shown in false color red. The two right upper panels show PET scans in a patient
before and 12 months after implantation with embryonic dopamine neurons. The preoperative panel demonstrates the profound
depletion of putamenal signal. Caudate signal is relatively higher. Twelve months after transplant, the FDOPA PET signal in puta-
men has increased toward the normal range. The two right lower panels show PET signals in a sham-surgery patient before and
12 months after transplant. As shown, there is no improvement in putamenal dopamine. The caudate dopamine is further depleted.
Reproduced from Freed et al. (2001), by permission of the New England Journal of Medicine. Copyright # 2001 Massachusetts
Medical Society. All rights reserved.

morning dose of levodopa. After transplant, there was levodopa cannot be altered with transplants. Because
no change in the best on state. The sham surgery transplants were placed only into putamen, we can say
group had no changes in UPDRS or Schwab and with some confidence that the motor effects of levo-
England scores after surgery (Freed et al., 2001). dopa are mediated through actions on the putamen.
In patients with the best clinical responses, trans- The primary outcome variable in our study, a global
plants could equal but not exceed the best effect of rating scale, proved to be an unreliable measure of
levodopa seen preoperatively. Therefore, transplants patient outcome. While still blinded at 12 months post-
generate a purely dopaminergic effect. Just as with deep transplant, patients were asked to pick terms that
brain stimulation, parkinsonian signs not improved by described their current state compared to 1 year before.
284 C. R. FREED ET AL.
Those descriptors were then assigned whole-number Dyskinesias have appeared in some transplant
values and averaged as though they were continuous recipients who had levodopa-induced dyskinesias prior
variables. Scores were recorded. Patients were then to transplant (Freed et al., 1990, 1992, 2001; Hagell
shown videos of themselves 1 year before and all et al., 2002; Olanow et al., 2003). In nearly all patients
groups changed their ratings to more positive values. who respond to transplants, there is an initial period of
Our study demonstrated that a global rating scale can- increased dyskinesias which resolve as drug doses are
not provide reliable assessments of long-term outcome reduced. In some patients, dyskinetic responses per-
in PD patients. sisted even after substantial reduction or even elimina-
Because some subjects are now more than 10 years tion of levodopa and other dopamine agonist therapy.
posttransplant, long-term analysis is possible. As noted In our double-blind study, all patients who developed
above, the most important predictor of response to persistent off dyskinesias were from the younger
transplant was the magnitude of the preoperative transplant group and all had had levodopa-induced
response to levodopa. Patients who had less than 50% dyskinesias prior to transplantation. It appears that
improvement after the first morning dose of levodopa the transplantation of tissue from four embryos pro-
were unlikely to respond to transplant. In our group of vided enough dopamine to trigger dyskinesias. In the
subjects, levodopa had a much broader range of effects first year after transplant, these patients had had
in those over age 60 compared to younger. Some older remarkable resolution of their parkinsonian signs.
patients had only the minimal 30% improvement in FDOPA PET scans showed some asymmetry of dopa-
UPDRS scores required to enter the study whereas mine fiber outgrowth in these patients, with greater
others had up to 90% improvement. All subjects under signal in left ventral putamen than was seen in other
age 60 had greater than 50% improvement after levo- patients who did not develop dyskinesias (Ma et al.,
dopa. Transplant results showed that only patients with 2002). Patients did not show supranormal concentra-
greater than 50% levodopa response had improvement tions of dopamine. Although asymmetric transplant
after surgery. When corrected for levodopa responsive- growth may be an explanation for dyskinesias, it is
ness, the older and younger patients had similar trans- also possible that the dyskinetic response has its origin
plant outcomes. By 2 or 3 years after transplantation, in other brain structures such as the subthalamic
UPDRS motor off scores showed an average improve- nucleus, the pallidum or the pars reticulata of the sub-
ment of 50% of the best response to levodopa seen stantia nigra. An imbalance between the restored dopa-
before transplant, regardless of age. mine innervation of putamen and the persistent
At the current stage of development, the average denervation of other brain regions may be responsible
response to transplant is equivalent to about half of for the dyskinetic responses. We reported a tendency
the levodopa effect. These long-term changes are toward dyskinetic movements in our first transplant
similar to what we first described in 1992 (Freed recipient in 1990 (Freed et al., 1990), as well as in
et al., 1992) and they are similar to what other groups 6 of 7 patients described in 1992 (Freed et al., 1992).
have subsequently reported. One could argue that a In most of these patients, dyskinesias were controlled
transplant which produces at least 50% of the best levo- by reductions in drug doses.
dopa effect is an appropriate target response. This effect Neurophysiologic and cognitive effects of transplant
is large enough to prevent severe off symptoms while were evaluated during the double-blind phase. Results
still not leading to dyskinetic movements (Freed et al., showed significant improvement in reaction plus
2001). With the transplant producing 50% of the movement times (Gordon et al., 2004). This benefit
needed dopamine, the optimum clinical response in was seen in both younger and older subjects. Cognitive
the individual patient can be achieved by administra- performance showed no change in any measure 1 year
tion of levodopa and other dopamine agonists. after transplant (Trott et al., 2003).
Individual variability of transplants has been a pro- In the second double-blind clinical trial, the goal of
blem. Using tissue obtained from elective abortion, the study was to compare placebo surgery with two
transplant outcome has varied from no effect to com- doses of tissue, a low dose of cells from two embryos
plete replacement of the need for levodopa. If stem and a high dose of tissue from up to eight embryos
cell research leads to a source of authentic midbrain transplanted into putamen, bilaterally (Olanow et al.,
dopamine neurons with more predictable growth and 2003). The primary outcome variable was the UPDRS
survival characteristics, then outcome might become motor off score, which had shown significant
more uniform. If cell transplantation could reliably changes in our earlier study. Surprisingly, at the 2-year
replace 5080% of the levodopa requirement in the endpoint, there were no differences in the transplant
individual patient, then transplantation could become groups compared to the placebo surgery controls.
an important component of the management of PD. FDOPA PET scans showed greater reinnervation of
TRANSPLANTATION 285
the putamen with the higher tissue dose at 1 year after in the form of a strand (made by extruding mesencepha-
transplant but similar reinnervation at 2 years after lic tissue through a tapered glass cannula with a 0.2-mm
transplant. This result is compatible with animal bore) into 6-OHDA-lesioned rats produced greater
studies which indicate that transplant growth occurs behavioral improvement and better dopamine neuron
in response to signals generated by denervated stria- survival than transplants of dissociated mesencephalon
tum (Dunnett et al., 1988). Presumably, the transplants (Clarkson et al., 1998b).
from the low-dose group continued to have axon
arborization between year 1 and year 2 in order to sup- 44.4.3. Age as a predictor of transplant outcome
ply portions of putamen inadequately innervated in the
first year. By this mechanism, the FDOPA PET scans Although PD is primarily a disease of elderly people,
of the low-dose group caught up with the high-dose some researchers have been hesitant to perform trans-
group by the second year. plantation in older patients. In our 1999 review, we
The authors could not account for the failure of the noted that the average age at the time of transplanta-
transplants to have effects on UPDRS scores. Because tion was 54 years, with average disease duration of
immunosuppression was instituted for only 6 months, about 13 years (Clarkson and Freed, 1999). Because
they speculated that transplants might have been in PD, the average age of onset is 55 years and the
rejected after drugs were stopped. This conjecture is average patient with PD is estimated to be 67, clinical
unlikely, since this same group has shown transplant trials have been biased toward younger patients.
survival at 18 months after transplant (Kordower Our double-blind study specifically addressed the
et al., 1995) and FDOPA PET scan signals did not fall issue of age with half of recruited patients 60 or over
as they would if transplant rejection had occurred. and half younger than 60. As noted above, it was not
They did find that patients with more severe baseline age per se but the preoperative response to levodopa
signs (higher UPDRS off scores) failed to improve that correlated with transplant outcome. In the aging
after transplant (Olanow et al., 2003). brain, the symptoms of PD are likely to be one mani-
Olanow et al. did note dyskinesias present in the festation of a more global neuropathologic process.
off state in a large proportion of their transplant sub-
jects and in none of the sham-surgery patients. 44.4.4. Number of donors
Although the dyskinesias were of minor significance
in most, some patients required the placement of deep The number of surviving dopaminergic neurons needed
brain stimulating electrodes in the subthalamic nucleus to improve motor function significantly in a patient
to control excess movements. with PD is unknown. The normal adult human
substantia nigra contains approximately 500 000
44.4.1. Unilateral versus bilateral transplantation dopamine-producing neurons (Pakkenberg et al.,
1991). Unfortunately, about 95% of dopamine neurons
Unilateral transplantation of fetal tissue was used in die after transplantation of rat or human tissue. In
the first clinical PD transplant trials (Lindvall et al., human patients, no more than 20 00025 000 dopamine
1989, 1990; Freed et al., 1990). Typically, implants cells survive from each embryo transplanted (Kordower
were made into the side of the brain contralateral to et al., 1995; Freed et al., 2001). Some investigators
the side of the body with the worse parkinsonian have argued that such a high rate of cell death requires
symptoms. In an attempt to provide more symmetric that tissue from six or more donors be transplanted into
innervation and with the goal of improving axial func- each putamen to restore a complete complement of
tions such as walking, we and others began transplant- dopamine-producing neurons. Studies examining the
ing fetal tissue bilaterally in the early 1990s (Freed ideal volume of grafted tissue in patients with PD are
et al., 1992). limited (Kopyov et al., 1997; Olanow et al., 2003) and
the issue is still widely debated. Because PD symptoms
44.4.2. Solid versus suspension graft only develop after a 50% loss of nigral neurons and a
6080% reduction in striatal dopamine (Bernheimer
Fetal mesencephalic tissue can be transplanted either et al., 1973; Kish et al., 1988), complete replacement
as a suspension of dissociated mesencephalon or as a of dopamine-producing neurons may not be required
solid graft. In some studies, human fetal tissue trans- to improve motor skills and to reduce the need for
planted into immunosuppressed 6-OHDA-lesioned levodopa.
rats showed comparable survival of both solid and We have counted the dopamine neurons that have
suspension grafts (Freeman et al., 1995b). However, survived in transplant tracks in postmortem brain. In
our studies showed that rat mesencephalon transplanted patients with bilateral putamenal implants who had
286 C. R. FREED ET AL.
clinical improvement sufficient to discontinue all levo- vior (Zawada et al., 1998a). Immortalized dopaminer-
dopa, the total number of surviving dopamine neurons gic cell lines that do not generate tumors can reduce
was 30 00040 000 (Freed et al., 2005). The two behavioral deficits and may offer an additional alter-
individuals had received tissue from two and four native to human fetal tissue (Clarkson et al., 1998a).
embryos, respectively. Figure 44.3 shows transplanted Because xenograft rejection remains an unsolved pro-
dopamine neurons in a subject who received cells from blem, aggressive immunosuppression is required for
two embryos via the frontal surgery approach. He transplantation of all non-human tissue. Efforts to
discontinued all levodopa 15 months after transplant. blunt the immune response by methods such as using
He was never immunosuppressed. He died 10 years fragments of a monoclonal antibody to major histo-
after surgery. These data provide experimental rather compatibility complex class I (Palzaban et al., 1995)
than hypothetical evidence for the amount of embryo- or class II antigens may reduce xenograft rejection.
nic tissue and the number of surviving cells needed to Encapsulating dopamine-producing xenografts may
have maximum clinical effect. also prevent rejection, although such anatomically
isolated grafts have no capacity to reinnervate the
44.5. Alternative tissue sources host brain (Emerich et al., 1992). Xenografts carry
the potential risk of animal virus transmission to
In addition to the scientific controversies about techni- humans. With the concern about CreutzfeldtJakob
ques for transplantation of human fetal tissue, ethical disease, special efforts to monitor and control this
concerns remain about the use of fetal tissue. There risk are required.
are logistical problems to recovering tissue after elec- Non-neuronal dopamine cells from retinal pigment
tive abortion. With the demonstration that a few fetal epithelium have been proposed and are under clinical
pig neural cells can survive transplantation into a evaluation (Stover et al., 2005). This biotechnology
patient with PD (Deacon et al., 1997), xenografts approach has expanded dopamine-producing retinal
have been proposed as an alternative to human cell pigment epithelial cells and made them adhere to micro-
transplants, although the immunologic hurdles are spheres which have then been transplanted into PD
enormous and unresolved. We reported that cloned patient striatum. An open clinical trial has suggested
transgenic cow embryos could survive transplantation some clinical benefits from this approach and a double-
in immunosuppressed rat and improve circling beha- blind study is underway.

Dopamine neurons-10 yrs after transplant

Fig. 44.3. Surviving dopamine neurons in a putamenal transplant track of a patient who died 10 years after implant.
This patient received dopamine neurons from two embryos via four needle passes in the putamen using a frontal surgical
approach. He received no immunosuppression. The left-hand panel shows a low-power view of one of the transplant tracks
with large numbers of dopamine neurons in the field (35). The right-hand panel shows a high-power view of the same trans-
plant with profiles of individual tyrosine hydroxylase-positive dopamine neurons evident (400). Cell counts revealed nearly
40 000 surviving dopamine neurons from the two embryos. Cells and processes are immunostained with antibody to tyrosine
hydroxylase (Pel Freez) followed by a secondary antibody with peroxidase diaminobenzidine visualization.
TRANSPLANTATION 287
The most promising source of an unlimited supply of reported. Several principles have emerged. Mesencepha-
dopamine neurons is from differentiated human embryo- lic tissue must be from early in embryonic development,
nic stem cells. Since 2000, mouse and non-human pri- typically 78 weeks after conception. Bilateral transplan-
mate embryonic stem cells have been successfully tation into putamen can be done safely during a single
converted to neurons with some dopaminergic charac- operation. Unilateral transplantation leads to asymmetric
teristics and transplanted into experimental animals transplant effects. Immunosuppression is not required.
(Kawasaki et al., 2000; Lee et al., 2000; Kim et al., Two double-blind studies have shown that trans-
2002; Shim et al., 2004; Takagi et al., 2005). Although plants can survive and improve FDOPA PET scans in
strategies have differed in details, the basic concept has most subjects. One has shown that UPDRS motor
been to differentiate embryonic stem cells from neuro- scores (bradykinesia and rigidity) can significantly
progenitor cells, then expand that cell population and improve in transplant patients compared to placebo
finally guide differentiation to dopamine neurons using controls (Freed et al., 2001). Well-controlled double-
known differentiation factors such as sonic hedgehog blind studies must be used to compare the outcome of
and fibroblast growth factor 8 (FGF8). The same prin- the transplant strategies yet to be tested.
ciples have been applied to human embryonic stem Although the clinical benefit in individual patients
cells with similar results, although with a slower time- has made drug elimination possible, there is substantial
line because of the longer cell cycle of human cells variability in outcome. Contributing to this variability
(Buytaert-Hoefen et al., 2004; Perrier et al., 2004; Yan are differences in pathologic processes in individual
et al., 2005). patients and in dopamine neuron survival and out-
For reasons that remain uncertain, transplant survi- growth. The best predictor of transplant outcome is
val of dopamine neurons derived from embryonic stem the preoperative response to levodopa (Freed et al.,
cells has been even worse than from fetal mesencepha- 2004). Transplantation can duplicate but cannot exceed
lon. Some success has been achieved by transplanting the best preoperative response to levodopa. Some
small numbers of mouse embryonic stem cells into patients with a prior history of levodopa-induced dys-
immunosuppressed rat models of PD, though teratomas kinesias may have persistent dyskinesias even after
inevitably form in a proportion of the animals (Bjork- the reduction or elimination of levodopa. These dyski-
lund et al., 2002). Selecting only neural progenitors at nesias respond to inhibitors of dopamine synthesis such
an earlier stage of differentiation may eliminate the ter- as metyrosine or to deep brain stimulation of pallidum
atoma risk (Chung et al., 2006). or subthalamic nucleus.
Interest has also focused on the delivery of neuro-
trophic factors to help reduce the number of dopamine 44.7. Future prospects for neural
neurons that die after transplantation. Cotransplantation transplantation
of fibroblasts infected to produce basic fibroblast
growth factor with mesencephalic grafts increased the Where shall human transplant research go from here? To
number of dopamine neurons in these transplants evaluate fully the effectiveness of dopamine cell trans-
(Takayama et al., 1995). Other survival factors, includ- plants for PD, a uniform source of dopamine neurons
ing insulin-like growth factor I and glial cell line- must be developed. Differentiation of embryonic stem
derived neurotrophic factor and caspase inhibitors, can cells from dopamine neurons may produce an unlimited
protect transplanted cells from apoptotic cell death number of dopamine neurons for transplant. Unlike most
(Clarkson et al., 1995; Zawada et al., 1996, 1998b; somatic tissue transplants, allografts of human fetal
Schierle et al., 1999). Inhibitors of lipid peroxidation neural tissue are not rejected. Therefore, donor-specific
have been tested in humans and may promote improved embryonic stem cells will probably not be required.
dopamine cell survival (Brundin et al., 2000). Although less likely, some of the other cell sources
described in this chapter may prove useful. Because
44.6. Summary of immunologic barriers, successful xenografts are
improbable.
Since 1988, neurotransplantation with embryonic Results of the double-blind studies indicate that
mesencephalic dopamine neurons has been tried as a transplants only into the putamen can produce benefi-
treatment for patients with advanced PD. Although cial motor effects. On the other hand, clinical benefits
transplant methods have differed substantially among have been incomplete in most patients. Many PD
centers, most reports have found some value to patients have cognitive deterioration either early or late
implants made into putamen. in their disease. In an effort to improve the outcome of
Open clinical trials of small numbers of patients have neurotransplantation, we are obliged to consider new
shown benefit from transplantation in most patients targets for transplantation. Because the region of dopa-
reported. Reduction of levodopa dose is frequently mine cell loss, the substantia nigra pars compacta, has
288 C. R. FREED ET AL.
important regional connections with the pars reticulata,
Bjorklund LM, Sanchez-Pernaute R, Chung S et al. (2002).
the subthalamic nucleus and the pallidum, dual tran- Embryonic stem cells develop into functional dopaminer-
plants into putamen and substantia nigra should be con- gic neurons after transplantation in a Parkinson rat model.
sidered. Because the caudate nucleus has a role in Proc Natl Acad Sci USA 99: 23442349.
cognitive function and eye movements, comparison of Brundin P, Nilsson OG, Strecker RE et al. (1986). Behavioral
dual transplants into caudate and putamen could be com- effects of human fetal dopamine neurons grafted in a rat
pared to putamen alone. Since dyskinesias in both model of Parkinsons disease. Exp Brain Res 65: 235240.
experimental animals and in humans are most often seen Brundin P, Pogarell O, Hagell P et al. (2000). Bilateral cau-
after priming with levodopa, a clinical trial of transplan- date and putamen grafts of embryonic mesencephalic tis-
tation early in PD may show that transplantation pre- sue treated with lazaroids in Parkinsons disease. Brain
123: 13801390.
vents the development of levodopa-induced dyskinesias.
Burns RS, Chiueh CC, Markey SP et al. (1983). A primate
Although restoring uniform anatomic integrity of the
model of parkinsonism: selective destruction of dopami-
nigrostriatal dopamine system is a daunting challenge, nergic neurons in the pars compacta of the substantia nigra
transplantation has already demonstrated successful by N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc
repair of the human PD brain. Nonetheless, transplant Natl Acad Sci USA 80: 45464550.
effects are limited to the replacement of dopamine. Buytaert-Hoefen KA, Alvarez E, Freed CR (2004). Genera-
There is no treatment for the progressive, non-dopami- tion of tyrosine hydroxylase positive neurons from human
nergic signs of PD. The ideal treatment for PD would embryonic stem cells after coculture with cellular sub-
stop the loss of dopamine neurons and other affected strates and exposure to GDNF. Stem Cells 22: 669674.
cell types at the earliest stage of the disease. Chung S, Shin BS, Hedlund E et al. (2006). Genetic selection
of sox1GFP-expressing neural precursors removes residual
References tumorigenic pluripotent stem cells and attenuates tumor
formation after transplantation. J Neurochem 97:
Alexander GE, DeLong MR (1985). Microstimulation of the 14671480.
primate neostriatum. II. Somatotopic organization of stria- Clarkson ED, Freed CR (1999). Minireview: development of
tal microexcitable zones and their relation to neuronal fetal neural transplantation as a treatment for Parkinsons
response properties. J Neurophysiol 53: 14171430. disease. Life Sci 65: 24272437.
Annett LE, Martel FL, Rogers DC et al. (1994). Behavioral Clarkson ED, Zawada WM, Freed CR (1995). GDNF
assessment of the effects of embryonic nigral grafts in reduces apoptosis in dopaminergic neurons in vitro. Neu-
marmosets with unilateral 6-OHDA lesions of the nigros- roreport 7: 145149.
triatal pathway. Exp Neurol 125: 228246. Clarkson ED, La Rosa FC, Edwards-Prasad J et al. (1998a).
Annett LE, Torres EM, Ridley RM et al. (1995). A comparison Improvement of neurological deficits in 6-hydroxydopamine-
of the behavioural effects of embryonic nigral grafts in the lesioned rats after transplantation with allogeneic simian virus
caudate nucleus and in the putamen of marmosets with uni- 40 large tumor antigen gene-induced immortalized dopamine
lateral 6-OHDA lesions. Exp Brain Res 103: 355371. cells. Proc Natl Acad Sci USA 95: 12561270.
Annett LE, Torres EM, Clarke DJ et al. (1997). Survival of Clarkson ED, Zawada WM, Adams FS et al. (1998b). Strands
nigral grafts within the striatum of marmosets with 6- of embryonic mesencephalic tissue show greater dopamine
OHDA lesions depends critically on donor embryo age. neuron survival and better behavioral improvement than
Cell Transplant 6: 557569. cell suspensions after transplantation in parkinsonian rats.
Bakay RAE, Fiandaca MS, Barrow DL et al. (1985). Preli- Brain Res 806: 6068.
minary report of the use of fetal tissue transplantation to Date I, Imaoka T, Miyoshi Y et al. (1996). Chromaffin cell
correct MPTP-induced Parkinson-like symptoms in survival and host dopaminergic fiber recovery in a patient
primates. Appl Neurophysiol 48: 358361. with Parkinsons disease treated by cografts of adrenal
Bankiewicz KS, Oldfield EH, Chiueh CC et al. (1986). Hemi- medulla and pretransected peripheral nerve. Case report.
parkinsonism in monkeys after unilateral internal carotid J Neurosurg 84: 685689.
artery infusion of 1-methyl-4-phenyl-1,2,3,6-tetrahydro- Davis GC, Williams AC, Markey SP et al. (1979). Chronic
pyridine (MPTP). Life Sci 39: 716. Parkinsonism secondary to intravenous injection of meper-
Bernheimer H, Birkmayer W, Hornykiewicz O et al. (1973). idine analogues. Psychiatry Res 1: 249254.
Brain dopamine and the syndromes of Parkinson and Deacon T, Schumacher J, Dinsmore J et al. (1997). Histological
Huntingtons: clinical, morphological and neurochemical evidence of fetal pig neural cell survival after transplantation
correlations. J Neurol Sci 20: 415455. into a patient with Parkinsons disease. Nat Med 3: 350353.
Bjorklund A, Steveni U (1979). Reconstruction of the Dunnett SB, Hernandez TO, Summerfield A et al. (1988).
nigrostriatal dopamine pathway by intracerebral nigral Graft-derived recovery from 6-OHDA lesions: specificity
transplants. Brain Res 177: 555560. of ventral mesencephalic graft tissues. Exp Brain Res 71:
Bjorklund A, Dunnett S, Brundin P et al. (2003). Neural 411424.
transplantation for the treatment of Parkinsons disease. Emerich DF, Winn SR, Christenson L et al. (1992). A novel
Lancet Neurol 2: 437445. approach to neural transplantation in Parkinsons disease:
TRANSPLANTATION 289

use of polymer-encapsulated cell therapy. Neurosci Hikosaka O, Sakamoto M, Usui S (1989a). Functional
Biobehav Rev 16: 437447. properties of monkey caudate neurons. I. Activities
Freed CR, Richards JB, Sabol KE et al. (1988). Fetal substan- related to saccadic eye movements. J Neurophysiol 61:
tia nigra transplants lead to dopamine cell replacement and 780798.
behavioral improvement in Bonnet monkeys with MPTP Hikosaka O, Sakamoto M, Usui S (1989b). Functional prop-
induced Parkinsonism. In: PM Beart, G Woodruff, DM erties of monkey caudate neurons. III. Activities related to
Jackson (Eds.), Pharmacology and Functional Regulation expectation of target and reward. J Neurophysiol 61:
of Dopaminergic Neurons. Macmillan Press, London, pp. 814832.
353360. Hudson JL, Bickford P, Johansson M et al. (1994). Target
Freed CR, Breeze RE, Rosenberg NL et al. (1990). Trans- and neurotransmitter specificity of fetal central nervous
plantation of human fetal dopamine cells for Parkinsons system transplants: importance for functional reinnerva-
disease. Results at 1 year. Arch Neurol 47: 505512. tion. J Neurosci 14: 283290.
Freed CR, Breeze RE, Rosenberg NL et al. (1992). Survival Hurtig H, Joyce J, Sladek JR Jr et al. (1989). Postmortem ana-
of implanted fetal dopamine cells and neurologic improve- lysis of adrenal-medulla-to-caudate autograft in a patient
ment 12 to 46 months after transplantation for Parkinsons with Parkinsons disease. Ann Neurol 25: 607614.
disease. N Engl J Med 327: 15491555. Kawasaki H, Mizuseki K, Nishikawa S et al. (2000). Induction
Freed CR, Breeze RE, Schneck SA et al. (1995). Fetal neural of midbrain dopaminergic neurons from ES cells by stromal
transplantation for Parkinsons disease. In: RR Rich, cell-derived inducing activity. Neuron 28: 3140.
(Ed.), Clinical Immunology; Principles and Practice. Kim JH, Auerbach JM, Rodriguez-Gomez JA et al. (2002).
Mosby-Year Book, St. Louis, pp. 16771687. Dopamine neurons derived from embryonic stem cells
Freed CR, Greene PE, Breeze RE et al. (2001). Transplanta- function in an animal model of Parkinsons disease.
tion of embryonic dnpamine neurons for severe Parkin- Nature 418: 5056.
sons disease. N Engl J Med 344: 710719. Kish SJ, Shannak K, Hornykiewicz O (1988). Uneven pat-
Freed CR, Breeze RE, Fahn S et al. (2004). Preoperative tern of dopamine loss in the striatum of patients with idio-
response to levodopa is the best predictor of transplant pathic Parkinsons disease. N Engl J Med 318: 876880.
outcome. Ann Neurol 55: 896. Kopyov OV, Jacques O, Lieberman A et al. (1996). Clinical
Freed CR, Zawada M, DeMasters BK et al. (2005). Human study of fetal mesencephalic intracerebral transplants
embryonic dopamine neurons transplanted into Parkinson for the treatment of Parkinsons disease. Cell Transplant
patients survive and improve motor function for at least 5: 327337.
10 years without immunosuppression. Program No. Kopyov OV, Jacques DS, Lieberman A et al. (1997). Out-
329.6. 2005 Abstract Viewer/Itinerary Planner. Society come following intrastriatal fetal mesencephalic grafts
for Neuroscience, Washington, DC [Online]. for Parkinsons patients is directly related to the volume
Freed WJ (1983). Functional brain tissue transplantation: of grafted tissue. Exp Neurol 146: 536545.
reversal of lesion-induced rotation by intraventricular Kordower JH, Freeman TB, Snow BJ et al. (1995). Neuro-
substantia nigra and adrenal medulla grafts, with a pathological evidence of graft survival and striatal reinner-
note on intracranial retinal grafts. Biol Psychiatry 18: vation after the transplantation of fetal mesencephalic
12051267. tissue in a patient with Parkinsons disease. N Engl J
Freed WJ, Morihisa JM, Spoor E et al. (1981). Transplanted Med 332: 11181124.
adrenal chromaffin cells in rat brain reduce lesion-induced Kori A, Miyashita N, Kato M et al. (1995). Eye movements
rotational behaviour. Nature 292: 351352. in monkeys with local dopamine depletion in the caudate
Freeman TB, Olanow CW, Hauser BA et al. (1995a). Bilat- nucleus. II. Deficits in voluntary saccades. J Neurosci
eral fetal nigral transplantation into the postcommissural 15: 928941.
putamen in Parkinsons disease. Ann Neurol 38: Lang A, Obeso O (2004). Challenges in Parkinsons disease:
379388. restoration of the nigrostriatal dopamine system is not
Freeman TB, Sanberg PR, Nauert CM et al. (1995b). The enough. Lancet Neurology 3: 309316.
influence of donor age on the survival of solid and suspen- Langston JW, Ballard P, Tetrud JW et al. (1983). Chronic
sion inrraparenchymal human embryonic nigral cells. Cell Parkinsonism in humans due to a product of meperidine-
Transplant 4: 141154. analog synthesis. Science 219: 979980.
Goetz CG, Stebbins GT 3rd, Klawans HL et al. (1991). United Langston JW, Widner H, Goetz CG et al. (1992). Core
Parkinson Foundation Neurotransplantation Registry on assessment program for intracerebral transplantations
adrenal medullary transplants: presurgical, and 1- and 2-year (CAPIT). Mov Disord 7: 213.
follow-up. Neurology 41: 17191722. Lee SH, Lumelsky N, Studer L et al. (2000). Efficient gen-
Gordon PH, Yu Q, Qualls C et al. (2004). Reaction time and eration of mid-brain and hindbrain neurons from mouse
movement time after embryonic cell implantation in Par- embryonic stem cells. Nat Biotechnol 18: 675679.
kinson disease. Arch Neurol 61: 858861. Lindvall O, Rehncrona S, Brundin P et al. (1989). Human
Hagell P, Piccini P, Bjorklund A et al. (2002). Dyskinesias fetal dopamine neurons grafted into the striatum in two
following neural transplantation in Parkinsons disease. patients with severe Parkinsons disease. Arch Neurol 46:
Nat Neurosci 5: 627628. 615631.
290 C. R. FREED ET AL.

Lindvall O, Brundin P, Widner H et al. (1990). Grafts of Spencer DD, Robbins RJ, Naftojin F et al. (1992). Unilateral
fetal dopamine neurons survive and improve motor func- transplantation of human fetal mesencephalic tissue into
tion in Parkinsons disease. Science 247: 574577. the caudate nucleus of patients with Parkinsons disease.
Ma Y, Feigin A, Dhawan V et al. (2002). Dyskinesia after N Engl J Med 327: 15411548.
fetal cell transplantation for parkinsonism: a PET study. Stover NP, Bakay RA, Subramanian T et al. (2005). Intras-
Ann Neurol 52: 628634. triatal implantation of human retinal pigment epithelial
Madrazo I, Drucker-Colin T, Daiz V et al. (1987). Open micro- cells attached to microcarriers in advanced Parkinson
surgical autograft of adrenal medulla to the right caudate disease. Arch Neurol 62: 18331837.
nucleus in two patients with intractable Parkinsons disease. Stromberg I, Herrera-Marschitz M, Ungerstedt U et al.
N Eng J Med 316: 831833. (1985). Chronic implants of chromaffin tissue into the
Mahalik TJ, Finger TE, Stromberg I et al. (1985). Subsranria dopamine-denervated striatum. Effects of NGF on graft
nigra transplants into denervated srriatum of the rat ultra- survival, fiber growth and rotational behavior. Exp Brain
strucrure of graft and host interconnections. J Comp Neu- Res 60: 335349.
rol 240: 6070. Stromberg I, Bygdeman M, Goldstein M et al. (1986). Human
Nakamura T, Dhawan V, Chaly T et al. (2001). Blinded fetal substantia nigra grafted to the dopamine-denervated
positron emission tomography study of dopamine cell striatum of immunosuppressed rats: evidence for func-
implantation for Parkinsons disease. Ann Neurol 50: tional reinnervation. Neurosci Lett 71: 271276.
181187. Takagi Y, Takahashi J, Saiki H et al. (2005). Dopaminergic neu-
Olanow CW, Goetz CG, Kordower JH et al. (2003). A double- rons generated from monkey embryonic stem cells function in
blind controlled trial of bilateral fetal nigral transplantation a Parkinson primate model. J Clin Invest 115: 102109.
in Parkinsons disease. Ann Neurol 54: 403414. Takayama H, Ray J, Raymon HK et al. (1995). Basic fibro-
Pakkenberg B, Moller A, Cundersen HJ et al. (1991). The blasts growth factor increases dopaminergic graft survival
absolute number of nerve cells in substantia nigra in normal and function in a rat model of Parkinsons disease. Nat
subjects and in patients with Parkinsons disease estimated Med 1: 5358.
with unbi-based stereological method. J Neurol Neurosurg Trott CT, Fahn S, Greene P et al. (2003). Cognition follow-
Psychiatry 54: 3033. ing bilateral implants of embryonic dopamine neurons in
Palzaban P, Deacon TW, Burns LH et al. (1995). A novel PD: a double blind study. Neurology 60: 19381943.
mode of immunoprotection of neural xenotransplants: Ungerstedt U, Arbuthoott CW (1970). Quantitative recording
masking of donor major histocompatibility complex class of rotational behavior in rats after 6-hydroxy-dopamine
I enhances transplant survival in the central nervous lesions of the nigrostriatal dopamine system. Brain Res
system. Neuroscience 65: 983996. 24: 485493.
Perlow MJ, Freed WJ, Hoffer BJ et al. (1979). Brain grafts
Watts RL, Subramanian T, Freeman A et al. (1997). Effect
reduce motor abnormalities produced by destruction of
of stereotaxic intrastriatal cografts of autologous adrenal
nigrosttiatal dopamine system. Science 204: 555560.
medulla and peripheral nerve in Parkinsons disease:
Perrier AL, Tabar V, Barberi T et al. (2004). Derivation of
two-year follow-up study. Exp Neurol 147: 510517.
midbrain dopamine neurons from human embryonic stem
Widner H, Tetrud J, Rehncrona S et al. (1992). Bilateral fetal
cells. Proc Natl Acad Sci USA 101: 1254312548.
mesencephalic grafting in two patients with Parkinsonism
Peschanski M, Defer C, NGuyen JP et al. (1994). Bilateral
induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
motor improvement and lateralization of L-dopa effect in
(MPTP). N Engl J Med 327: 15561563.
two patients with Parkinsons disease following intrastria-
tal transplantation of foetal ventral mesencephalon. Brain Wuerthele SM, Freed WJ, Olson L et al. (1981). Effect of
117: 487499. dopamine agonists and antagonists on the electrical activ-
Redmond DE, Sladek JR Jr, Roth RH et al. (1986). Fetal ity of substantia nigra neurons transplanted into the lateral
neuronal grafts in monkeys given methylphenyltetrahydro- ventricle of the rat. Exp Brain Res 44: 110.
pyridine. Lancet 8490: 11251127. Yan Y, Yang D, Zarnowska ED et al. (2005). Directed dif-
Schierle GS, Hansson O, Leist M et al. (1999). Caspase inhi- ferentiation of dopaminergic neuronal subtypes from
bition reduces apoptosis and increases survival of nigral human embryonic stem cells. Stem Cells 23: 781790.
transplants. Nat Med 5: 97100. Zawada WM, Kirschman DL, Cohen JJ et al. (1996). Growth
Schmidt RH, Ingvar M, Lindvall O et al. (1982). Functional factors rescue embryonic dopamine neurons from
activity of substantia nigra grafts reinnervating the stria- programmed cell death. Exp Neurol 140: 6067.
tum: neurotransmitter metabolism and (14C)-2-deoxy-D- Zawada WM, Cibelli JB, Choi PK et al. (1998a). Somatic
glucose autoradiography. J Neurochem 38: 737748. cell cloned transgenic bovine neurons for transplantation
Shim JW, Koh HC, Chang MY et al. (2004). Enhanced in parkinsonian rats. Nat Med 4: 569574.
in vitro midbrain dopamine neuron differentiation, dopa- Zawada WM, Zastrow DJ, Clarkson ED et al. (1998b).
minergic function, neurite outgrowth, and 1-methyl-4-phe- Growth factors improve immediate survival of embryonic
nylpyridium resistance in mouse embryonic stem cells dopamine neurons after transplantation into rats. Brain
overexpressing Bcl-XL. J Neurosci 24: 843852. Res 786: 96103.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 45

Gene therapy approaches for the treatment


of Parkinsons disease

BIPLOB DASS AND JEFFREY H. KORDOWER*

Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA

Parkinsons disease (PD) is the second most common virus-glutamic acid decarboxylase (AAV-GAD), Avigen:
neurodegenerative disease, with over 1 million Ameri- AAV-aromatic acid decarboxylase (AADC) and Cere-
cans suffering from this disorder. The cardinal symptoms gene: adeno-associated virus-neurturin (AAV-NTN)).
of PD are rigidity of the cogwheel type, resting tremor, There are a number of reasons to suspect that delivery
bradykinesia and postural instability (Marsden, (1990)). of therapeutic genes would be superior to the delivery of
These symptoms result from striatal dopamine insuffi- pharmacological agents. The first reason is site specificity.
ciency secondary to degeneration of dopaminergic When delivering therapeutic drugs peripherally, you are
neurons in the substantia nigra (SN) pars compacta often dose-limited because the drug usually acts in a non-
(Ehringer and Hornykiewicz, 1960). Although it has been specific manner and activates unintended systems. For
over 50 years since its discovery, levodopa remains the example, administration of levodopa in high enough doses
gold-standard treatment by which all other therapies can lead to hallucinations due to increasing dopaminergic
are compared. Despite being an outstanding therapy, neurotransmission in the mesolimbic system. The meso-
levodopa ultimately loses its usefulness, not because it limbic system emanates from the ventral tegmental area,
can no longer provide benefit, but because benefit is a region adjacent to the SN. Axons from the ventral teg-
accompanied by debilitating side-effects that limit its uti- mental area do not terminate in the striatum but rather in
lity (Marsden and Parkes, 1977; Curtis et al., 1984). the nucleus accumbens and medial prefrontal cortex.
Therefore novel therapeutic strategies are needed for When this occurs, the dose of levodopa needs to be low-
the long-term treatment of PD patients. Towards this ered, thereby minimizing the therapeutic efficacy of this
end, a myriad of pharmacological therapies have com- powerful compound. In contrast, novel therapeutic strate-
prised the armament of treatment strategies for PD. These gies such as gene therapy can drive the dose of the thera-
have been reviewed elsewhere in this volume. Further- peutic molecule by placing the vectors in site-specific
more, a variety of surgical treatments have been tested loci and, with care, this delivery method should not affect
clinically, such as adrenal cell transplants, adrenal cell/per- unintended brain regions.
ipheral nerve cografts, human fetal nigral transplantation For prodopaminergic gene therapy approaches (see
and porcine fetal transplantation (Madrazo et al., 1987; below), it is thought that the postcommissural putamen
Lindvall et al., 1989; Quinn, (1990); Kordower et al., is the most critical site. This choice has been based
1995; Date et al., 1996; Deacon et al., 1997; Wenning upon a number of factors. First, the loss of dopamine
et al., 1997; Schumacher et al., 2000; Freed et al., 2001). in PD is greatest in the postcommissural putamen. Sec-
Out of all the surgical interventions tested to date, only ond, this region of the striatum is intimately connected
deep brain stimulation has emerged as a genuinely useful with motor cortex. In contrast, the caudate nucleus and
treatment strategy (Figueiras-Mendez et al., 1999). anterior putamen are similar embryologically, have
Recently, preclinical studies delivering therapeutic dense interconnections with association cortices and
genes have shown such promise that three strategies have are linked to higher-order functions such as cognition
already begun clinical trials (Neurologix: adeno-associated rather than motor function. Based upon this logic and

*Corresponding author: Jeffrey H. Kordower, Department of Neurological Sciences, Cohn Building, Rush University Medical
Center Chicago, IL 60612, USA. E-mail: jkordowe@rush.edu, Tel: 312-563-3570, Fax: 312-563-3571.
292 B. DASS AND J. H. KORDOWER
due to the limited availability of donor material, double- clear empirical difference between AAV and lentivirus
blinded clinical trials testing human and porcine fetal is that the former is retrogradely transported whereas
transplantation strategies have grafted cells exclusively the latter is not. The functional relevance of this differ-
to the postcommissural putamen that were based upon ence, if any, is unclear. Given their safety profile and
successful open-label studies (Lindvall et al., 1989; scientific advantages, only AAVs have currently been
Kordower et al., 1995; Deacon et al., 1997; Wenning approved for clinical trials in PD.
et al., 1997; Schumacher et al., 2000; Freed et al.,
2001). Similarly, Amgen sponsored a double-blind trial 45.2. Gene therapy for Parkinsons disease:
in which catheters were placed in the postcommissural delivery of therapeutic enzymes
putamen to deliver the trophic factor glial-derived
neurotrophic factor that were also based upon success- The first strategy proposed as a gene therapeutic treat-
ful open-label studies (GDNF, Gill et al., 2003; Slevin ment for PD was striatal levodopa delivery by ex vivo
et al., 2005). It is notable that all of these double-blind expression of tyrosine hydroxylase (TH) (Wolff et al.,
trials have failed to meet their primary endpoints. There 1989). The initial basis for the ex vivo delivery of
are a number of explanations for the failure of these TH was the known efficacy of peripheral levodopa
trials, for instance, cells not surviving in the porcine treatment combined with the neurosurgical safety
transplantation study, immune issues may have influ- demonstrated by fetal ventral mesencephalic dopamine
enced the human embryonic transplantation experi- grafting in rodents and humans (Brundin et al., 1986;
ments and dose and catheter design issues may have Lindvall et al., 1989). These studies failed due to the
influenced the outcome of the GDNF trial. However, it inability of ex vivo gene therapy approaches at that
remains troubling that this dogmatic view that the time to provide high-titer long-term gene expression,
postcommissural putamen is the essential region for as well as having issues with inflammation. Indeed,
dopaminergic reinnervation has yet to be sustained in shutdown of the transgene and only transient and
any double-blinded clinical trial in PD. low-level functional recovery was seen using this
approach. Although the rationale for proceeding with
45.1. Drug development strategy enzyme gene delivery was logical, the tools at that
time to elicit long-term high-titer gene expression
Because PD is a long-term degenerative disorder and were unavailable. To expand upon this initial study,
the gene delivery itself will require neurosurgery, a a serotype 2 AAV encoding TH was utilized in 6-
single surgical procedure that results in long-term or hydroxydopamine (6-OHDA)-lesioned rats. The use
permanent therapy will be required to make this thera- of stably lesioned animals facilitated the investigation
peutic option practical. This means that the gene trans- of the reversal of deficits induced by neurotoxin treat-
fer should lead to very long-term or permanent gene ment. TH immunoreactivity was detectable in the
expression. The issue of which vector would be ideal striata up to 4 months after vector administration and
for use in the treatment was initially a problem, since resulted in a persistent reduction of apomorphine-
each virus has strengths and drawbacks. Adenovirus induced rotational behavior for the duration of the
and herpes simplex virus (HSV) administrations were experiment (Kaplitt et al., 1994). Following this, vec-
initially shown to induce strong immunogenic res- tors encoding for combinations of genes involved in
ponses and be short-acting and were therefore non- the synthesis of levodopa were injected into the dener-
starters for clinical application in PD. AAV has a low vated striata of rats using separate AAVs. Thus, double
level of transgene expression and lentivirus has a fairly or triple transfections of TH, AADC and guanosine tri-
small capacity and the stigma of being associated with phosphate-cyclohydrolase-1 (GCH-1) were shown to
human immunodeficiency virus (HIV). However there be effective at producing levodopa in the 6-OHDA-
has been remarkable improvement in the safety and ex- lesioned striatum, as measured by microdialysis
pression of many of these vectors. The use of gutless (Mandel et al., 1998; Shen et al., 2000; Kirik et al.,
adenovirus has minimized its immunogenicity. The 2002). The success of this approach was also deter-
creation and characterization of novel AAV serotypes mined by the PD model that was employed. In fully
have resulted in robust long-term gene expression in rats lesioned animals, gene delivery of TH and GCH-1
and monkeys. For a while, empirical evidence favored did not alter apomorphine-induced rotational behavior,
lentivirus as the vector of choice as studies progressed but rotational deficits were attenuated in rats with a
towards clinical trials. However, AAV now appears partial lesion of the nigrostriatal pathway (Kirik
equal to lentivirus with regard to many of the character- et al., 2002). When three genes were delivered (TH,
istics deemed important for gene delivery, such as long- GTPCH1 and AADC), behavioral recovery, as mea-
term expression, infectivity of neurons and the absence sured by apomorphine-induced rotational behavior,
of immunogenicity and inflammation. In fact, the one could be observed in rats with complete nigrostriatal
GENE THERAPY APPROACHES FOR THE TREATMENT OF PARKINSONS DISEASE 293
lesions. The increase in striatal dopamine content was dopamine concentrations were only modestly raised in
modest in these animals, although the effects of the the lesioned striata. In contrast, triple transfection in the
gene delivery could be demonstrated for 12 months putamen of TH, AADC and GCH-1 did result in beha-
(Kirik et al., 2002). Using a lentivirus gene delivery vioral recovery of 4060% in 4 MPTP-treated primates
system based on equine infectious anemia virus, (Muramatsu et al., 2002). Dopamine synthesis in the
Azzouz and coworkers (2002) performed similar striatum was also increased approximately fivefold,
experiments. A non-human lentivirus was employed although this increase was still very low. Long-term
as a safety measure as it is less likely to recombine studies examining bi- and tricistronic delivery of thera-
and form HIV if used clinically. It is critical to note, peutic enzymes are still needed prior to the initiation of
however, that there is currently no empirical evidence clinical trials.
that lentivirus could recombine to form a pathogenic As an alternative to expressing the enzymes
HIV particle and there is also no proof that non-human required for the production of levodopa and dopamine,
forms of HIV would pose a lesser risk. Rotational the Bankiewicz group has used viral vectors (see
behavior induced by apomorphine administration was Table 45.1) to express solely AADC in the striatum
partially reversed by treatment with the tricistronic to enhance the production of dopamine following the
(TH, GTPCH1 and AADC) vector, although turning administration of levodopa. This approach is hoped
behavior was still greater than 5 per minute in these to reduce levodopa requirements and provide a more
rats. Striatal dopamine content was also partially ele- stable level of conversion to dopamine in the striatum
vated by the gene delivery, but levels were still less and therefore help avoid the incidences of levodopa-
than 10% of the intact hemisphere. induced dyskinesias. In their initial experiments, they
Since the symptoms of PD only arise after a thresh- injected AAV-AADC into the striata of MPTP-
old of approximately 70% striatal dopamine depletion lesioned monkeys and were able to observe increased
occurs, a small increase in dopamine levels could result AADC activity in vivo, coupled with increased dopa-
in significant improvements in motor function. Thus, mine metabolite production (Bankiewicz et al.,
these strategies were investigated in primates with stable 2000). Convection-enhanced delivery was used as the
lesions. When administered to 1-methyl-4-phenyl- infusion method and resulted in a high density of
1,2,3,6-tetrahydropyridine (MPTP)-treated primates, AADC immunoreactive cells throughout the striatum.
AAV-mediated gene delivery of TH and AADC was Most transfected cells had a NeuN-positive, medium
successful up to 10 weeks (During et al. (1998)). No spiny neuron-type morphology and there were very
inflammation or other adverse effects were noted after few glial fibrillary acid protein and AADC co-positive
AAV administration. However, no behavioral recovery cells. In a subsequent experiment in 6-OHDA-lesioned
was observed in these monkeys and postmortem rats, the gene transfer of AADC also resulted in

Table 45.1
Characteristics and uses of viral vectors in animal models of Parkinsons disease

Neuronal Maximum Strength of Duration of Immunogenic Current uses in


Vector preference insert expression expression response models of PD

Adenovirus 7.5 kb Weeks TH, GDNF, XIAP, JBD,


Calpastatin, Cu/Zn
SOD,
Adeno 4 kb (type 2) >2 years TH, AADC, GTP-
associated (type 2) (type 5) cyclohydrolase,
virus (type 5) BDNF, GDNF, NTN,
SHH, GAD
Herpes simplex 30150 kb Weeks TH, Bcl-2, GDNF
virus
Lentivirus 9 kb >1 year GDNF, TH, AADC,
GTPcyclohydrolase

TH, tyrosine hydroxylase; GDNF, glial-derived neurotrophic factor; XIAP, X chromosome-linked inhibitor of apoptosis; JBD, JNK binding
domain; SOD, superoxide dismutase; AADC, aromatic acid decarboxylase; GTP, guanosine triphosphate; NTN, neurturin; SHH, sonic hedge-
hog; GAD, glutamic acid decarboxylase.
294 B. DASS AND J. H. KORDOWER
enhanced conversion of levodopa to dopamine, but the has been set and currently patients enrolled in the phase
dopamine was not stored within the neurons and I trial that have received unilateral AAV-GAD into the
seemed to be released continuously (Sanchez-Pernaute STN are reported to have improved 27% on the Unified
et al., 2001). Theoretically, uncontrolled and increased Parkinsons Disease Rating Scale and have significant
dopamine release in the striatum might exacerbate decreases in fluorodeoxyglucose utilization on the side
levodopa-induced dyskinesias. However, a reduction contralateral to the injection (Neurologix press release,
in the required dose of levodopa might lead to fewer 25 September 2005).
treatment-related side-effects. Interestingly, when
AAV-AADC was delivered without using convection- 45.3. Delivery of trophic molecules
enhanced delivery at three focal points in the striatum of
MPTP-lesioned monkeys, dyskinesias were observed, Although the striatal delivery of genes encoding for
suggesting that the creation of a hotspot of dopamine dopaminergic enzymes or STN delivery of inhibitory
activity is key to the generation of levodopa-related enzymes might provide substantial therapeutic benefit,
side-effects (Bankiewicz et al., 2006). This corroborates none of these approaches has a compelling basis for
previous work with cell grafts, suggesting that focal altering the underlying disease. An alternative ap-
hotspots rather than widespread grafts can cause proach, which theoretically could alter the natural his-
dyskinesias (Steece-Collier et al., 2003). In further tory of PD, is the delivery of genes encoding for
studies with MPTP-lesioned monkeys infused with neuron-saving trophic factors. In addition to providing
AAV-AADC throughout the striatum using convec- neuroprotection, gene delivery of trophic factors has
tion-enhanced delivery, no dyskinesia was observed the added advantage of enhancing the function of cells
over a 5-year time period, although behavior in response and inducing cells to produce greater amounts of rele-
to levodopa treatment was improved in these animals vant neurotransmitters (i.e. dopamine). A wealth of data
(personal communications from Dr. Bankiewicz). has been amassed on the efficacy of many dopaminergic
As a result of the preclinical work performed in the trophic molecules, such as epidermal growth factor,
Bankiewicz laboratory using MPTP-lesioned monkeys basic fibroblast growth factor, brain-derived neuro-
and 6-OHDA-lesioned rats, a clinical trial was trophic factor (BDNF), sonic hedgehog and GDNF to
initiated by Avigen in December of 2004 using an prevent degeneration following bolus administrations
AAV to deliver the DNA for AADC into the human in models of nigrostriatal degeneration (Gash et al.,
putamen. At the time of writing, the trial is still under 1996; Pearce et al., 1996, 1999; Svendsen et al., 1996;
way and no peer review data are available for ass- Dass et al., 2002; for review, see Collier and Sortwell,
essment. However, a press release suggested that 1999). In particular, GDNF is a particularly promising
AAV-AADC was well tolerated and resulted in the candidate for clinical use, based on a series of studies
production of AADC (Avigen press release, 18 July in animal models of PD. Initial open-label and double-
2005). blind studies examining the safety and efficacy of
Currently, a phase I clinical trial utilizing an AAV monthly intraventricular injections GDNF failed. No
expressing GAD in PD patients is under way in New efficacy was seen and serious side-effects were
York (During et al., 2001). The rationale behind this observed (Kordower et al., 1999; Nutt et al., 2003).
study is that the overactivity of the excitatory subthala- However this result was to be expected, since vulner-
mic nucleus (STN) in PD patients might be reduced by able neurons are not exposed to GDNF via this delivery
the transfer of the GAD gene into this nucleus and so method. Indeed, even chronic intraventricular infusions
provide relief from thalamic inhibition. Previously, of GDNF demonstrated trivial immunoreactivity to
this group had shown that AAV-GAD transfection into GDNF protein beyond the ventricular ependyma. These
the STN of fully 6-OHDA-lesioned rats could induce a initial studies demonstrated that, even if GDNF is the
phenotypic change in these nuclei, causing them to correct protein, it would be unsuccessful if delivered
release gamma-aminobutyric acid (GABA) in addition in an ineffective manner. In an attempt to improve
to glutamate (Luo et al., 2002). Injection of an AAV- delivery parameters, two open-label trials delivered
GAD65 vector 3 weeks before a nigrostriatal lesion GDNF chronically into the postcommissural putamen
resulted in a 35% protection of nigral TH-positive of PD patients (Gill et al., 2003; Amgen press releases,
cells and decreased rotational behavior and behavioral June 2004, February 2005; Slevin et al., 2005). Both stu-
asymmetries. As with other studies utilizing AAVs, no dies reported sustained functional benefit. One trial
inflammation was found 45 months after administra- (Gill et al., 2003) also reported increases in fluoro-
tion and no antibodies against the viral capsid were dopa uptake on positron emission tomography (PET)
detected in the sera of these animals (Luo et al., 2002). scan, an effect that increased from 6 months to 1 year.
Thus, the basis for further studies utilizing AAV-GAD Disappointingly, the 1 patient who has currently come
GENE THERAPY APPROACHES FOR THE TREATMENT OF PARKINSONS DISEASE 295
to autopsy showed only marginal changes in TH, an nal infusions of GDNF need to be addressed and once
effect that was dwarfed by other procedures such as further information is gathered, a new double-blind trial
fetal transplants that have yet to be proved efficacious. using more optimal dosing and delivery parameters
As a follow-up to these open-label studies, Amgen should be initiated.
sponsored a multicenter double-blind trial examining
the safety and efficacy of bilateral intraputamenal 45.4. Gene delivery of glial-derived
GDNF in patients with PD. This trial failed to dem- neurotrophic factor in rodents before lesioning
onstrate efficacy. At first glance, one has to consider
strongly that placebo effects and experimenter bias, A superior approach to trophic factor infusion may be
phenomena that are inherent to all open-label trials gene therapy. Gene therapy does not have the hardware
and particularly those in PD (de la Fuente-Fernandez issues associated with the implantation of catheters and
et al., 2001), resulted in the positive clinical effects seen pumps, nor does it have the issue of numerous pump
in the Bristol and Kentucky studies and when they were refills over long periods of time. In this regard, numer-
controlled for by the enhanced rigor associated with a ous studies have created a body of evidence indicating
double-blind trial, these reported effects could not be that gene delivery of GDNF and GDNF-like molecules
substantiated. However, equally strongly, it needs to may be a powerful therapeutic tool to provide neuropro-
be noted that there were substantial and potentially tection and neuroaugmentation for degenerating SN
critical differences in technique between the open-label neurons. Martha Bohn and colleagues deserve credit
trials and the double-blind trial. These differences for performing the first experiments examining gene
included dosage (higher dosages ultimately used in the delivery of GDNF in animal models of PD. Initial
open-label trials), catheter design (a thicker catheter studies used an adenovirus to deliver GDNF before
(Bristol) and multiport catheter (Kentucky) in the 6-OHDA lesioning in rats and so examined the neuro-
open-label studies) and rate of trophic factor delivery protective potential of this therapeutic strategy
(convection-enhanced delivery in the open-label trials (Bilang-Bleuel et al., 1997; Choi-Lundberg et al.,
and non-convection enhanced delivery in the double- 1997; Lapchak et al., 1997). In Choi-Lundbergs study,
blind trial). Therefore it is very possible that delivery supranigral administration of Ad-GDNF (AD, adeno-
methods in the double-blind procedure were inferior to virus) 7 days before the initiation of a striatal 6-OHDA
those in the open-label trial and this critical aspect of lesion in rats resulted in a 75% protection of nigral fluor-
the study prevented the induction of functional benefit. ogold-positive dopaminergic neurons, but the size of the
In addition to the absence of clinical benefit, the Amgen striatal lesion was unchanged in these animals, com-
trial was stopped prematurely due to the presence of pared to controls. Horellous group injected Ad-GDNF
side-effects. Neutralizing antibodies against GDNF into the rat striatum 6 days before the initiation of a stria-
were observed in approximately 10% of patients and tal 6-OHDA partial lesion (Bilang-Bleuel et al., 1997).
some monkeys treated with high-dose GDNF displayed The number of TH-IR (IR, immunoreactivity) cells in
profound cerebellar degeneration. The origin of the the SN was significantly greater than in control animals,
antibody response is unclear and has been seen in all although again complete protection of the SN was not
infusion trials. It could be due to leakage of protein into achieved. In the striatum, TH immunoreactivity was
the abdomen during refilling of the pump. It also could increased following striatal Ad-GDNF administration
be due to the use of glycosylated, rather than native, and amphetamine-induced rotational asymmetry was
GDNF. It is unclear what side-effects antibodies attenuated. Thus, the site of administration of GDNF is
to GDNF might cause in adult humans and, to date, no critical to the protection of the nigrostriatal pathway fol-
side-effects have been reported in patients who received lowing a neurotoxic insult. Using GDNF protein, simi-
GDNF. With regard to the cerebellar damage, it is pos- lar results have been obtained when comparing the
sible that, due to the thin catheter and non-convection protective effects of intrastriatal, intranigral and intra-
delivery method, the GDNF backed up the catheter ventricular administration (Kirik et al., 2000a). Accord-
and GDNF flowed over the brain convexities to the cer- ingly, GDNF was only able to protect the anatomy and
ebellum. Two aspects of this phenomenon are notable. function of the nigrostriatal pathway following an
It never happens when GDNF or GDNF family mem- intrastriatal injection of Ad-GDNF and, if a viral vector
bers are delivered via viral vectors to monkeys directly encoding a trophic molecule was considered for use
to the striatum. Second, no patients who have received in PD patients, it is more likely to be efficacious to
intraputamenal GDNF have been reported to have signs administer the vector into the caudate and putamen.
of cerebellar degeneration. The only thing at this point Choi-Lundberg and colleagues (1998) repeated the
that is perfectly clear is that nothing is perfectly clear. administration of Ad-GDNF, but injected the vector
The safety and delivery issues surrounding intraputame- intrastriatally before creating a partial 6-OHDA lesion.
296 B. DASS AND J. H. KORDOWER
No improvement in the density of TH-IR fibers in the 6-OHDA lesion, the dopaminergic nigrostriatal path-
striatum was observed, but behavioral asymmetry was way was completely protected and this was accompa-
reduced and nigral TH-IR cell numbers were pro- nied by a reduction in ()-amphetamine-induced
tected. This result might be mediated by the retrograde asymmetry (Kirik et al., 2000b). This landmark study
transport of GDNF itself or Ad-GDNF into the SN and indicates that the site of GDNF delivery and the rein-
the subsequent trophic support that GDNF could pro- statement of striatal dopamine are critical if functional
vide to the functionality of the cell body. GDNF was effects are to be observed.
detectable at nanogram levels in the striatum and pico- Another alternative vector to adenovirus that
gram levels in the nigra. Since GDNF can undergo ret- induces little inflammation is lentivirus and this has
rograde transportation along the nigrostriatal tract, it also been used in the rat 6-OHDA model of nigrostriatal
was unclear if this finding was as a result of the degeneration. Georgievska and coworkers (2002a)
actions on GDNF or a function of the adenovirus demonstrated that GDNF administered 3 weeks before
(Tomac et al., 1995; Lapchak et al., 1997; Mufson a striatal injection of 6-OHDA can preserve nigral neu-
et al., 1999). Following this study, the same group pre- rons relative to rats receiving a control vector in a dose-
treated aged rats with Ad-GDNF before initiating a dependent manner. In this regard, animals treated with
striatal 6-OHDA lesion (Connor et al., 1999). GDNF a lower titer of vector preserved 65% of TH-IR neurons
increased TH-IR optical density in the striatum, indi- whereas animals administered with a higher concentra-
cating the potent phenotypic upregulation GDNF can tion of lenti-GDNF preserved 77% of TH-positive cells.
provide in addition to its neuroprotective properties. Interestingly, intense sprouting of fibers was observed
It is likely that the actions of GDNF transfected by in the medial parts of the striatum, globus pallidus and
an adenovirus not only protects cells from 6-OHDA- entopeduncular nucleus and this corresponded to the
mediated cell death but also from the toxicity induced presence of GDNF immunoreactivity, indicating that
by the virus itself, highlighting the reason to use less GDNF was transported and had effects away from the
immunogenic vectors such as AAV or lentivirus in injection site. The same group repeated this experiment
the brain. As an example of this, in the studies by with a single titer of lenti-GDNF administered into the
Choi-Lundberg et al. (1997) and Bilang-Bleuel et al. striatum (Georgievska et al., 2002b). As before, TH-
(1997), both groups reported adverse cellular reactions positive cell numbers in the nigra were preserved by
to the administration of first-generation adenovirus, lenti-GDNF treatment and deficits in amphetamine-
regardless of the particular gene sequence transfected induced rotational behavior were prevented. Tracing
by the vector. Choi-Lundberg et al. reported that 12 out the nigrostriatal pathway using fluorogold retrograde
of 14 rats injected with adenovirus possessed a mild to labeling from the striatum or AAV-GFP (GFP, green
moderate reaction in the SN, whereas Bilang-Bleuel fluorescent protein) anterograde marking from the nigra
and colleagues found that the size of the striatum was showed that lenti-GDNF administration also protected
reduced and that Ad-b-Gal (Gal, galactosidase) adminis- the axonal projections of the SN pars compacta into
tration alone reduced the number of TH-IR cells by 37%. the striatum. However TH fiber immunoreactivity was
To circumvent the adenovirus-mediated immuno- downregulated in the striatum: the authors speculated
genic response, the use of AAVs has markedly that this was due to a downregulation of TH follow-
increased in the last few years. GDNF, when expressed ing high levels of GDNF expression. Also, non-
following transfection by an AAV, has been very suc- drug-induced motor asymmetry was unchanged by
cessful at preventing dopaminergic cell death in the lenti-GDNF administration and this was attributed
SN pars compacta and reducing behavioral deficits in to the aberrant sprouting of TH-positive fibers from
6-OHDA-lesioned rats (Mandel et al., 1997; Kirik the striatum projecting to the globus pallidus, ento-
et al., 2000b; Wang et al., 2002). In the study by Man- penduncular nucleus and the SN.
del et al., the authors injected AAV-GDNF 3 weeks GDNF gene therapy has been successful at protect-
before initiating a partial 6-OHDA lesion, resulting ing dopamine neurons in other models of PD, includ-
in protection of fluorogold-labeled neurons in the ing mice receiving MPTP. MPTP, after metabolism
SN. Kirik and colleagues (2000b) found that the to 1-methyl-4-phenylpyridinium ion (MPP), selec-
expression of GDNF was stable for over 6 months tively kills dopaminergic neurons by inhibiting com-
when AAV-GDNF was injected into the SN, but no plex I in the electron transport chain and thereby
protection of the striatal dopaminergic immunoreac- stopping cellular respiration. GDNF delivered into
tive terminals was observed and behavioral recovery the striatum using an adenovirus, 2 days prior to the
did not occur following striatal 6-OHDA treatment administration of MPTP in mice, elevated striatal
(Kirik et al., 2000b). In contrast, when AAV-GDNF dopamine levels in comparison to lesioned control
was administered into the striatum of rats prior to a mice, although the dopamine content was still severely
GENE THERAPY APPROACHES FOR THE TREATMENT OF PARKINSONS DISEASE 297
reduced compared to naive animals (Kojima et al., has not been considered further (Klein et al., 1999).
1997). In another interesting study, Ad-GDNF was Sonic hedgehog has also been shown by two groups to
concurrently administered into the striatum with an protect cells in vivo from 6-OHDA lesioning. Using an
adenovirus encoding the protein caspase inhibitor, X- adenoviral construct, Hurtado-Lorenzo and colleagues
chromosome-linked inhibitor of apoptosis (XIAP) (2004) protected dopaminergic cells at the nigral level
(Eberhardt et al., 2000). Treatment of the mice with from 6-OHDA lesioning, but could not protect the stria-
Ad-GDNF alone, followed by a chronic lesioning tal axonal terminals. In an alternative study, the admin-
paradigm with MPTP injections over 5 days, resulted istration of sonic hedgehog delivered using an AAV 3
in no protection of TH-IR neurons in the SN and no ele- weeks before striatal 6-OHDA treatment reduced
vation of striatal dopamine, or dopamine metabolites amphetamine-induced rotational behavior and pro-
levels. However, when Ad-XIAP and Ad-GDNF treat- tected striatal dopamine levels and nigral TH-positive
ments were administered in combination, a synergistic cell counts (Dass et al., 2005b).
elevation in both nigral TH-IR cell numbers and
protection of the catecholamine content of the striatum 45.5. Neuroprotection in a primate model
from MPTP treatment occurred. of Parkinsons disease
GDNF has also been transfected in combination
with the antiapoptotic molecule Bcl-2, using HSV vec- Primate studies are essential prerequisites for deter-
tors (Natsume et al., 2001). When administered to 6- mining whether novel therapies for PD are ready for
OHDA-lesioned rats amphetamine-induced rotational clinical trials. Primarily, primate studies employ
behavior was significantly reduced by the administra- MPTP to induce a lesion and often repeated adminis-
tion of either HSV-GDNF or HSV-Bcl-2 or an injec- trations are required to induce a stable lesion. Using
tion of both combined. Cell survival was enhanced this method, the extent of the MPTP-induced lesion
by either vector and there was an additive effect of cannot be predicted before degeneration, making neu-
the two when co-administered. It is unclear however roprotective effects difficult to interpret. Thus, some
if this effect would be replicated in parkinsonian investigators have used sterotaxic administrations of
patients, since so little is known about the etiology of 6-OHDA in smaller primates, such as marmosets, to
the disease and this approach is primarily oriented guarantee destruction of the nigrostriatal pathway.
towards neuroprotection. Also, in a previous experi- AAV-GDNF administered into the striatum and SN
ment, the administration of either HSV-Bcl-2 or a of marmosets, before an injection of 6-OHDA into
control virus resulted in a 40% reduction in fluorogold the medial forebrain bundle (MFB), only provided a
and TH-positive cells, in the absence of 6-OHDA modest protection of dopaminergic cells in the SN.
lesioning, and so the use of HSV as a vector for gene However, motor function assessed by a modified clin-
transfer in humans is unlikely (Yamada et al., 1999). ical rating scale was reversed to pre-lesion levels 5
It is important to note that gene delivery of GDNF weeks after 6-OHDA (Eslamboli et al., 2003). Also
is not effective in all models of PD. Lo Bianco et al. ()-amphetamine induced rotational behavior and
(2004) administered lenti-GDNF to the supranigral head positional bias were attenuated at all time points
region of rats 2 weeks prior to an intranigral lentiviral after lesioning, indicating that AAV-transfected GDNF
injection of the A30P mutant human a-synuclein. protected motor function in the primates.
Although excellent expression of GDNF was obser-
ved, gene delivery of this trophic factor was unable 45.6. Neurorestoration following
to prevent the loss of nigrostriatal neurons. 6-hydroxydopamine lesioning
Two other trophic molecules have been tested in
animal models of PD. BDNF has been delivered into Of equal importance to the protection of the nigrostria-
the SN pars compacta of rats using a recombinant tal system from the unknown degenerative processes
AAV, which also encoded green fluorescent protein of PD is the ability to restore dopaminergic transmis-
(Klein et al., 1999). Expression of the green fluores- sion using trophic factors. Lapchak and colleagues
cent protein was observed at 9 months after the admin- (1997) attempted to use Ad-GDNF to restore the dopa-
istration of the viral vector. Six months after gene minergic function of the nigrostriatal pathway of rats
transfection, the animals were partially lesioned with with an established 6-OHDA lesion. Their 6-OHDA
6-OHDA and rotational behavior in response to administration paradigm resulted in a 73% loss of
amphetamine was reduced in the AAV-BDNF-treated dopamine in the SN and a 99% reduction in the stria-
animals in comparison to controls. However, no pro- tum. In animals injected with Ad-GDNF into the SN,
tection from 6-OHDA-induced dopaminergic cell the nigral dopamine content was elevated to 44% of
death in the SNpc was found and so this neurotrophin the levels on the intact side, whereas dopamine levels
298 B. DASS AND J. H. KORDOWER
in the striatum were increased to 4% of the contralat- the individual dexterity levels of MPTP-lesioned mon-
eral side. These neurochemical improvements in the keys. Overall, TH immunoreactivity in the striatum
nigrostriatal system were reflected in a moderate was significantly greater in lenti-GDNF-treated mon-
reduction in apomorphine-induced asymmetry and an keys compared to control animals. In the SN, an
improvement in locomotor activity. In a recent similar increase in TH-positive cell numbers and cell density
study, the administration of Ad-GDNF, 7 days after compared to the untreated hemisphere was observed
the initiation of a striatal 6-OHDA lesion, protected following lenti-GDNF administration, whereas lenti-
basal, potassium chloride and amphetamine-evoked b-Gal-treated animals had large decreases in cell
extracellular dopamine levels in the striatum measured counts and density on the MPTP-lesioned side. Immu-
by mcirodialysis, such that the basal level of dopamine nohistochemistry for CD45, CD3 and CD8 resulted in
was 65% of intact animal levels and the drug-induced negligible levels of positive cells, indicating that the
levels were 3540% (Smith et al., 2005). lentivirus caused no immunogenicity.
Another group investigated the effect of administra- Two young unlesioned rhesus monkeys were also
tion of AAV-GDNF after the initiation of a striatal 6- administered with lenti-GDNF into the right caudate
OHDA partial lesion in rats and examined effects of and putamen and left SN pars compacta and examined
gene therapy over a longer term. Delayed delivery of 8 months later for gene expression. Enzyme-linked
AAV-GDNF prevented nigral cell death, when admi- immunosorbent assay analysis found 2.53.5ng/mg tis-
nistered 4 weeks following the initiation of a 6-OHDA sue of GDNF in the caudate and putamen, whereas in
lesion. Behavioral recovery, assessed by apomorphine- the SN there were many GDNF immunoreactive cells,
induced rotations and contralateral limb use, was also proving long-term transgene expression. In a subse-
improved following AAV-GDNF treatment and these quent study, the effect of lenti-GDNF was examined
changes were maintained for 20 weeks (Wang et al., stereologically in the striatum of aged monkeys and
2002). Thus, the use of AAV offers a mechanism for unilaterally MPTP-lesioned monkeys (Palfi et al.,
the expression of molecules in the striatum over a long 2002). MPTP treatment alone increased the numbers
period of time, whilst inducing minimal side-effects, of intrinsic dopamine neurons in the striatum and
such as inflammation, and so could be used to achieve lenti-GDNF administration further increased the stria-
a long-term administration of trophic factors. tal TH immunoreactive cell counts sevenfold. In aged
monkeys, there was also a large increase in TH and
45.7. Neurorestoration following MPTP dopamine transporter immunoreactivity in the striatum
treatment and this correlated with GDNF expression, indicating
an autotrophic effect. The coupling of the two res-
Our group generated a GDNF-expressing lentivirus and ults suggests that lenti-GDNF is effective, regardless
injected it into the striatum and nigra of young adult rhe- of the level of downregulation of the dopaminergic
sus monkeys 1 week after MPTP lesioning, or unlesioned phenotype (Figs. 45.1 and 45.2).
aged monkeys (Kordower et al., 2000). In all monkeys,
strong GDNF immunoreactivity was observed and 45.8. Effect of glial-derived neurotrophic
this was coupled with increased striatal fluorodopa factor in an intact system
uptake, TH immunoreactivity, dopamine and HVA con-
tent. Interestingly, GDNF immunoreactivity was also Following 6-OHDA lesioning in rats, it was noticed by
observed in the globus pallidus and SN pars reticulata, Georgievska and colleagues (2002b) that in areas of
indicating anterograde transport of the transgene. strong lentivirally mediated overexpression of GNDF
MPTP-lesioned animals displayed clear motor dis- in the striatum, the expression of TH was reduced.
ability, as measured by a clinical rating scale, but the The authors then checked to see if this phenomenon
administration of lenti-GDNF resulted in a reduction was replicated in naive rats and observed that treat-
over the following 3 months. Dexterity, as measured ment with lenti-GDNF into either the striatum or nigra
by a hand-reach task, was also improved in lenti- resulted in 6070% decreases in TH expression and
GDNF-administered monkeys compared to the lenti-b- TH mRNA levels. Levodopa production remained at
Gal-treated animals. Lenti-GDNF-treated animals had normal levels, indicating that GDNF increased the
a 300% increase in fluorodopa uptake seen in PET activity of TH (Georgievska et al., 2004). However,
scans taken at 3 months postvector injection compared when GDNF is delivered using a viral vector in primates,
to control monkeys, although this was limited to the TH expression has only been observed to increase in
putamen only. naive animals, indicating that the phenotypic downregu-
Striatal TH immunoreactivity was enhanced by lation only occurs in rodents (Kordower et al., 2000;
lenti-GDNF treatment in a manner that correlated with Eslamboli et al., 2005).
GENE THERAPY APPROACHES FOR THE TREATMENT OF PARKINSONS DISEASE 299

GDNF
Immunoreactivity

TH
Immunoreactivity

Fig 45.1. Bilateral lentiviral delivery of glial-derived neurotrophic factor (GDNF) into the caudate and putamen improves
(TH-IR) in aged rhesus monkeys.

45.9. Neurturin: An alternative to glial-derived that indicate that neurturin can provide structural and
neurotrophic factor functional protection of nigrostriatal neurons (Dass
et al., 2004, 2005a; Bartus et al., 2005; Herzog et al.,
Neurturin is a protein that was discovered by Milbrandt 2005). These studies have led to the initiation of a
and coworkers (Kotzbauer et al., 1996) and is the second phase I clinical trial.
trophic factor identified in the GDNF family of ligands.
The amino acid sequence of neurturin has a high 45.10. Conclusions
sequence homology to GDNF. Although both GDNF
and neurturin signal through the RET receptor, they phy- When considering a gene therapy strategy to treat any
siologically bind to two separate receptors; GDNF uses human disease, there are two major unknowns that
GFRa1 whereas neurturin prefers GFRa2. There are no must be conquered. These are: (1) the safety of the
GFRa2 receptors in the striatum, suggesting that the gene transfer itself; and (2) the efficacy of the trans-
delivery of neurturin would not be effective. However, gene product. In addition, any new treatment, espe-
at the levels achieved following gene delivery, neurturin cially a highly experimental strategy such as gene
can bind to GFRa1 and, like GDNF, provide potent neu- therapy, must be shown to be superior to the current
roprotection for degenerating nigrostriatal neurons. gold-standard therapy of levodopa.
In this regard, neurturin has recently been delivered The most important issue to be addressed, if gene
to four sites in the rat striatum via a lentivirus, 2 weeks therapy is to be utilized clinically, is safety. Previously,
before a 6-OHDA lesion. Lenti-neurturin was shown to in 2002, a gene therapy trial involving patients with
protect over 90% of the nigral TH immunoreactive severe combined immune deficiency using a retrovirus
cells. Unfortunately, striatal innervation was not was halted after 1 out of the 11 trial subjects developed
spared and amphetamine-induced rotational behavior leukemia (Hacein-Bey-Abina et al., 2003). Analysis of
was not ameliorated (Fjord-Larsen et al., 2005). Our this patient revealed that the retrovirus had integrated
lab, in collaboration with Ceregene Inc., has per- into the host genome at 40 different sites, including
formed a series of experiments in rats and monkeys one, LMO-2, which was related to oncogenesis and
300 B. DASS AND J. H. KORDOWER

GDNF
Immunoreactivity

TH
Immunoreactivity

Fig. 45.2. Unilateral lentiviral delivery of GDNF into the caudate and putamen increases (TH-IR) in 1-methyl-4-phenyl-
1,2,3,6-tetrahydropyridine (MPTP)-lesioned rhesus monkeys. No GDNF-IR or increases in TH-IR are observed in the untreated
hemisphere (left side).

this contributed to the abnormal growth of a T cell. rigorous screening of patients must be conducted
Insertational mutagenesis is therefore a concern with before the commencement of gene therapy.
the use of integrating viral vectors, especially those To gain the greatest efficacy when treating PD, the
such as serotype 5 AAV or lentivirus, which are capable best site for administration of a viral vector must be
of infecting glial cells. Also, Nakai and colleagues ascertained. AAV, but not lentivirus, has shown a cap-
(2003) showed in mice that AAV serotype 2 integrates ability for retrograde transport. Anterograde transport
its DNA more frequently into active genes rather than can also occur with trophic factors such as GDNF.
quiescent ones. This could cause problems of unwanted expression of
More seriously, a patient suffering from ornithine transgenes in other areas of the brain if the site of
transcarbamylase (OTC) deficiency was infused into administration was the striatum. Currently this is just
the right hepatic artery with an adenovirus encoding a theoretical concept as no adverse reactions have been
the human OTC gene, as part of a phase I trial (Raper demonstrated as a result of such transport. Thus,
et al., 2003). Unfortunately, the patient suffered from a although it would be favorable to have transport of a
severe immune reaction to the adenovirus and died 5 trophic factor or dopamine-synthesizing enzyme into
days after treatment. The reason for this may have the SN pars compacta, it is unclear whether transport
been that the patient was previously exposed to adeno- to the pallidum, thalamus or the neocortex would be
virus and this may have contributed towards an beneficial, harmful or inconsequential. Diffusion, fol-
increased autoimmune response to the vector. Most lowing an injection of a vector into the nigral region,
of the other patients in this trial, who all received equally could result in major problems with alterations
lower titers of adenovirus, also rapidly developed in the firing of ventral tegmental area neurons. Thus,
fever, supporting the theory that previous exposure to the preferential site of injection and the expression
adenovirus followed by a second administration of level of transgenes away from the administration loca-
the virus could cause greater inflammation. Therefore tion need to be fully investigated. One possibility to
GENE THERAPY APPROACHES FOR THE TREATMENT OF PARKINSONS DISEASE 301
limit the expression of a transgene is to infect cells Azzouz M, Martin-Rendon E, Barber RD et al. (2002).
with a viral vector and then transplant them into the Multicistronic lentiviral vector-mediated striatal gene
brain, but currently this strategy has had mixed results transfer of aromatic L-amino acid decarboxylase, tyrosine
in promoting behavioral recovery in 6-OHDA-lesioned hydroxylase, and GTP cyclohydrolase I induces sustained
transgene expression, dopamine production, and func-
rats (Ostenfeld et al., 2002; Park et al., 2003).
tional improvement in a rat model of Parkinsons disease.
The ideal candidate for gene therapy in PD is another
J Neurosci 22 (23): 1030210312.
question that needs more study. Trophic factors, dopa- Bankiewicz KS, Eberling JL, Kohutnicka M et al. (2000).
mine-synthesizing enzymes and basal ganglia modula- Convection-enhanced delivery of AAV vector in parkinso-
tion therapies have been outlined here. The transfection nian monkeys; in vivo detection of gene expression and
of trophic factors promises the greatest benefit, in that restoration of dopaminergic function using pro-drug
it could prevent the progressive degeneration of the approach. Exp Neurol 164 (1): 214.
dopaminergic nigrostriatal system, whilst enhancing the Bankiewicz KS, Daadi M, Pivirotto P et al. (2006). Focal stria-
function of surviving neurons. The problems encoun- tal dopamine may potentiate dyskinesias in parkinsonian
tered following the administration of GDNF protein monkeys. Exp Neurol 197 (2): 363372. [Epub 2005 Dec 9].
should not discourage the site-specific gene delivery of Bartus RT, Herzog CD, Cunningham JJ et al. (2005). Biolo-
trophic molecules. Consideration must be made about gical activity of CERE-120, an AAV2 vector encoding
whether the gene therapy treatments in development human neurturin, in the rat 6-hydroxydopamine model of
today for PD are truly being tested in conditions and nigrostriatal degeneration and in the young intact and aged
rat. J Neurosci #545.1.
models that reflect the clinical scenario in which they
Bilang-Bleuel A, Revah F, Colin P et al. (1997). Intrastriatal
would actually be employed. The etiology of the disease
injection of an adenoviral vector expressing glial-cell-line-
remains unknown and therefore the neuroprotective ele- derived neurotrophic factor prevents dopaminergic neuron
ments of trophic factor therapy may become ineffectual, degeneration and behavioral impairment in a rat model of
but the well-characterized restorative effects should still Parkinson disease. Proc Natl Acad Sci USA 94 (16):
provide benefit. Other strategies for the treatment of PD 88188823.
may also provide benefit for patients, by reducing their Brundin P, Nilsson OG, Strecker RE et al. (1986). Beha-
levodopa requirements and so ameliorate the develop- vioural effects of human fetal dopamine neurons grafted
ment of levodopa-induced side-effects. Site-specific in a rat model of Parkinsons disease. Exp Brain Res
intrastriatal delivery of the enzymes involved in the pro- 65 (1): 235240.
duction of levodopa is expected to obviate side-effects Choi-Lundberg DL, Lin Q, Chang YN et al. (1997). Dopami-
resulting from global decarboxylation of levodopa in nergic neurons protected from degeneration by GDNF
gene therapy. Science 275 (5301): 838841.
non-therapeutic brain compartments, as occurs with
Choi-Lundberg DL, Lin Q, Schallert T et al. (1998). Beha-
Sinemet treatment. Thus, future research into gene ther-
vioral and cellular protection of rat dopaminergic neurons
apy is promising with regard to the development of pro- by an adenoviral vector encoding glial cell line-derived
spective treatments for PD. neurotrophic factor. Exp Neurol 154 (2): 261275.
Collier TJ, Sortwell CE (1999). Therapeutic potential of
References nerve growth factors in Parkinsons disease. Drugs Aging
14 (4): 261287 Review.
Amgen Press Release 28th June 2004. Amgens Phase 2 Connor B, Kozlowski DA, Schallert T et al. (1999). Differ-
Study of GDNF for Advanced Parkinsons Disease Fails ential effects of glial cell line-derived neurotrophic factor
to Meet Primary Endpoint; Six Months of Treatment (GDNF) in the striatum and substantia nigra of the aged
Showed Biological Effect But No Clinical Improvement. Parkinsonian rat. Gene Ther 6 (12): 19361951.
http://wwwext.amgen.com/media/media_pr_detail.jsp? Curtis L, Lees AJ, Stern GM et al. (1984). Effect of L-DOPA
year2004&releaseID585632 on course of Parkinsons disease. Lancet 2 (8396):
Amgen Press Release 11th Feb 2005. Following Complete 211212.
Review of Phase 2 Trial Data Amgen Confirms Decision to Date I, Imaoka T, Miyoshi Y et al. (1996). Chromaffin cell
Halt GDNF Study; Comprehensive Review of Scientific Find- survival and host dopaminergic fiber recovery in a patient
ings, Patient Safety, Drove Decision. http://wwwext. amgen. with Parkinsons disease treated by cografts of adrenal
com/media/media_pr_detail.jsp?year2005&releaseID medulla and pretransected peripheral nerve. Case report.
673490 J Neurosurg 84 (4): 685689.
Avigen Press Release 18th July 2005. Avigen Announces Deacon T, Schumacher J, Dinsmore J et al. (1997). Histolo-
Encouraging Early Data from Parkinsons Disease Clinical gical evidence of fetal pig neural cell survival after trans-
Trial. Evidence for First Successful Gene Transfer of plantation into a patient with Parkinsons disease. Nat
AADC Gene in Humans. http://www.avigen.com/non_ Med 3 (3): 350353.
financial_release/2005/2005_Avigen_EarlyData_PDClini- Dass B, Iravani MM, Jackson MJ et al. (2002). Behavioural
calTrial_071805.htm and immunohistochemical changes following supranigral
302 B. DASS AND J. H. KORDOWER

administration of sonic hedgehog in 1-methyl-4-phenyl- Gash DM, Zhang Z, Ovadia A et al. (1996). Functional
1,2,3,6-tetrahydropyridine-treated common marmosets. recovery in parkinsonian monkeys treated with GDNF.
Neuroscience 114 (1): 99109. Nature 380 (6571): 252255.
Dass B, Herzog CD, Gasmi M et al. (2004). Adeno-asso- Georgievska B, Kirik D, Rosenblad C et al. (2002a). Neuropro-
ciated virus mediated gene delivery of neurturin (CERE- tection in the rat Parkinson model by intrastriatal GDNF gene
120) in non-human primates. J Neurosci #790.19. transfer using a lentiviral vector. Neuroreport 13 (1): 7582.
Dass B, Herzog CD, Gasmi M et al. (2005a). Adeno-associated Georgievska B, Kirik D, Bjorklund A (2002b). Aberrant
virus mediated gene delivery of neurturin (CERE-120) sprouting and downregulation of tyrosine hydroxylase in
prevents MPTP-induced motor disability following injec- lesioned nigrostriatal dopamine neurons induced by long-
tion into the striatum of non-human primates. J Neurosci lasting overexpression of glial cell line derived neuro-
#545.6. trophic factor in the striatum by lentiviral gene transfer.
Dass B, Iravani MM, Huang C et al. (2005b). Sonic hedge- Exp Neurol 177 (2): 461474.
hog delivered by an adeno-associated virus protects dopa- Georgievska B, Kirik D, Bjorklund A (2004). Overexpression
minergic neurones against 6-OHDA toxicity in the rat. of glial cell line-derived neurotrophic factor using a lenti-
J Neural Transm 112 (6): 763778. [Epub 2004 Dec 10]. viral vector induces time- and dose-dependent downregu-
de la Fuente-Fernandez R, Ruth TJ, Sossi V et al. (2001). Expec- lation of tyrosine hydroxylase in the intact nigrostriatal
tation and dopamine release: mechanism of the placebo effect dopamine system. J Neurosci 24 (29): 64376445.
in Parkinsons disease. Science 293 (5532): 11641166. Gill SS, Patel NK, Hotton GR et al. (2003). Direct brain
During MJ, Samulski RJ, Elsworth JD et al. (1998). In vivo infusion of glial cell line-derived neurotrophic factor in
expression of therapeutic human genes for dopamine pro- Parkinson disease. Nat Med 9 (5): 589595.
duction in the caudates of MPTP-treated monkeys using Hacein-Bey-Abina S, von Kalle C, Schmidt M et al.
an AAV vector. Gene Ther 5 (6): 820827. (2003). A serious adverse event after successful gene ther-
During MJ, Kaplitt MG, Stern MB et al. (2001). Subthalamic apy for X-linked severe combined immunodeficiency.
GAD gene transfer in Parkinson disease patients who are N Engl J Med 348 (3): 255256.
candidates for deep brain stimulation. Hum Gene Ther Herzog CD, Holden JE, Bakay RA et al. (2005). Enhanced
12 (12): 15891591. 18Fdopa uptake in the striatum of aged rhesus monkeys
Eberhardt O, Coelln RV, Kugler S et al. (2000). Protection by following striatal delivery of CERE120, an AAV2 vector
synergistic effects of adenovirus-mediated X-chromosome- encoding human neurturin. J Neurosci #545.7.
linked inhibitor of apoptosis and glial cell line-derived Hurtado-Lorenzo A, Millan E, Gonzalez-Nicolini V et al.
neurotrophic factor gene transfer in the 1-methyl- (2004). Differentiation and transcription factor gene ther-
4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinsons apy in experimental Parkinsons disease: Sonic Hedgehog
disease. J Neurosci 20 (24): 91269134. and Gli-1, but not Nurr-1, protect nigrostriatal cell bodies
Ehringer H, Hornykiewicz O (1960). Verteilung von nora- from 6-OHDA-induced neurodegeneration. Mol Ther
drenalin und dopamin (3-hydroxytyramine) im gerhin des 10 (3): 507524.
menschen und ihr verhalten bei erkrantungen des extrapyr- Kaplitt MG, Leone P, Samulski RJ et al. (1994). Long-term
amidalen systems. Kiln Wschr 38: 12361239. gene expression and phenotypic correction using adeno-
Eslamboli A, Cummings RM, Ridley RM et al. (2003). associated virus vectors in the mammalian brain. Nat
Recombinant adeno-associated viral vector (rAAV) deliv- Genet 8 (2): 148154.
ery of GDNF provides protection against 6-OHDA lesion Kirik D, Rosenblad C, Bjorklund A (2000a). Preservation of
in the common marmoset monkey (Callithrix jacchus). a functional nigrostriatal dopamine pathway by GDNF in
Exp Neurol 184 (1): 536548. the intrastriatal 6-OHDA lesion model depends on the site
Eslamboli A, Georgievska B, Ridley RM et al. (2005). Con- of administration of the trophic factor. Eur J Neurosci
tinuous low-level glial cell line-derived neurotrophic fac- 12 (11): 38713882.
tor delivery using recombinant adeno-associated viral Kirik D, Rosenblad C, Bjorklund A et al. (2000b). Long-
vectors provides neuroprotection and induces behavioral term rAAV-mediated gene transfer of GDNF in the rat
recovery in a primate model of Parkinsons disease. J Neu- Parkinsons model: intrastriatal but not intranigral trans-
rosci 25 (4): 769777. duction promotes functional regeneration in the lesioned
Figueiras-Mendez R, Marin-Zarza F, Antonio, Molina J et al. nigrostriatal system. J Neurosci 20 (12): 46864700.
(1999). Subthalamic nucleus stimulation improves directly Kirik D, Georgievska B, Burger C et al. (2002). Reversal of
levodopa induced dyskinesias in Parkinsons disease. motor impairments in parkinsonian rats by continuous
J Neurol Neurosurg Psychiatry 66 (4): 549550. intrastriatal delivery of L-DOPA using rAAV-mediated
Fjord-Larsen L, Johansen JL, Kusk P et al. (2005). Efficient gene transfer. Proc Natl Acad Sci USA 99 (7):
in vivo protection of nigral dopaminergic neurons by len- 47084713. [Epub 2002 Mar 26].
tiviral gene transfer of a modified Neurturin construct. Klein RL, Lewis MH, Muzyczka N et al. (1999). Prevention
Exp Neurol 195 (1): 4960. of 6-hydroxydopamine-induced rotational behavior by
Freed CR, Greene PE, Breeze RE et al. (2001). Transplanta- BDNF somatic gene transfer. Brain Res 847 (2): 314320.
tion of embryonic dopamine neurons for severe Parkinsons Kojima H, Abiru Y, Sakajiri K et al. (1997). Adenovirus-
disease. N Engl J Med 344 (10): 710719. mediated transduction with human glial cell line-derived
GENE THERAPY APPROACHES FOR THE TREATMENT OF PARKINSONS DISEASE 303

neurotrophic factor gene prevents 1-methyl-4-phenyl- of neurodegenerative diseases. Prog Neurobiol 57 (4):
1,2,3,6-tetrahydropyridine-induced dopamine depletion in 451484 Review.
striatum of mouse brain. Biochem Biophys Res Commun Muramatsu S, Fujimoto K, Ikeguchi K et al. (2002). Beha-
238 (2): 569573. vioral recovery in a primate model of Parkinsons disease
Kordower JH, Freeman TB, Snow BJ et al. (1995). Neuro- by triple transduction of striatal cells with adeno-asso-
pathological evidence of graft survival and striatal reinner- ciated viral vectors expressing dopamine-synthesizing
vation after the transplantation of fetal mesencephalic enzymes. Hum Gene Ther 13 (3): 345354.
tissue in a patient with Parkinsons disease. N Engl J Nakai H, Montini E, Fuess S et al. (2003). AAV serotype 2
Med 332 (17): 11181124. vectors preferentially integrate into active genes in mice.
Kordower JH, Palfi S, Chen EY et al. (1999). Clinicopatholo- Nat Genet 34 (3): 297302.
gical findings following intraventricular glial-derived neu- Natsume A, Mata M, Goss J et al. (2001). Bcl-2 and GDNF
rotrophic factor treatment in a patient with Parkinsons delivered by HSV-mediated gene transfer act additively to
disease. Ann Neurol 46 (3): 419424. protect dopaminergic neurons from 6-OHDA-induced
Kordower JH, Emborg ME, Bloch J et al. (2000). Neuro- degeneration. Exp Neurol 169 (2): 231238.
degeneration prevented by lentiviral vector delivery of Neurologix Press Release 25th September 2005. Neurologix
GDNF in primate models of Parkinsons disease. Science Announces PositiveInterim Results of Landmark Gene
290 (5492): 767773. Therapy Clinical Trial for Patients with Parkinsons Disease.
Kotzbauer PT, Lampe BA, Heuckeroth RO et al. (1996). http://phx.corporate-ir.net/phoenix.zhtml?c106413p
Neurturin, a relative of glial-cell-line-derived neurotrophic irol-newsArticleID760701highlight
factor. Nature 384: 467470. Nutt JG, Burchiel KJ, Comella CL et al. (2003). ICV GDNF
Lapchak PA, Araujo DM, Hilt DC et al. (1997). Adenoviral Study Group. Implanted intracerebroventricular. Glial cell
vector-mediated GDNF gene therapy in a rodent lesion line-derived neurotrophic factor. Randomized, double-
model of late stage Parkinsons disease. Brain Res blind trial of glial cell line-derived neurotrophic factor
777 (1-2): 153160. (GDNF) in PD. Neurology 60 (1): 6973.
Lindvall O, Rehncrona S, Brundin P et al. (1989). Human Ostenfeld T, Tai YT, Martin P et al. (2002). Neurospheres
fetal dopamine neurons grafted into the striatum in two modified to produce glial cell line-derived neurotrophic
patients with severe Parkinsons disease. A detailed factor increase the survival of transplanted dopamine neu-
account of methodology and a 6-month follow-up. Arch rons. J Neurosci Res 69 (6): 955965.
Neurol 46 (6): 615631. Palfi S, Leventhal L, Chu Y et al. (2002). Lentivirally deliv-
Lo Bianco C, Deglon N, Pralong W et al. (2004). Lentiviral ered glial cell line-derived neurotrophic factor increases
nigral delivery of GDNF does not prevent neurodegenera- the number of striatal dopaminergic neurons in primate
tion in a genetic rat model of Parkinsons disease. Neuro- models of nigrostriatal degeneration. J Neurosci 22 (12):
biol Dis 17 (2): 283289. 49424954.
Luo J, Kaplitt MG, Fitzsimons HL et al. (2002). Subthalamic
Park S, Kim EY, Ghil GS et al. (2003). Genetically modified
GAD gene therapy in a Parkinsons disease rat model.
human embryonic stem cells relieve symptomatic motor
Science 298 (5592): 425429.
behavior in a rat model of Parkinsons disease. Neurosci
Madrazo I, Drucker-Colin R, Diaz V et al. (1987). Open
Lett 353 (2): 9194.
microsurgical autograft of adrenal medulla to the right
caudate nucleus in two patients with intractable Parkin- Pearce RKB, Collins P, Jenner P et al. (1996). Intraventricular
sons disease. N Engl J Med 316 (14): 831834. infusion of basic fibroblast growth factor (bFGF) in the
Mandel RJ, Spratt SK, Snyder RO et al. (1997). Midbrain MPTP treated common marmoset. Synapse 23: 192200.
injection of recombinant adeno-associated virus encoding Pearce RKB, Costa S, Jenner P et al. (1999). Chronic supra-
rat glial cell line-derived neurotrophic factor protects nigral infusion of BDNF in normal and MPTP treated
nigral neurons in a progressive 6-hydroxydopamine- common marmosets. J Neural Transm 106: 663683.
induced degeneration model of Parkinsons disease in rats. Quinn NP (1990). The clinical application of cell grafting
Proc Natl Acad Sci USA 94 (25): 1408314088. techniques in patients with Parkinsons disease. Prog
Mandel RJ, Rendahl KG, Spratt SK et al. (1998). Characteri- Brain Res 82: 619625.
zation of intrastriatal recombinant adeno-associated virus- Raper SE, Chirmule N, Lee FS et al. (2003). Fatal systemic
mediated gene transfer of human tyrosine hydroxylase and inflammatory response syndrome in a ornithine transcar-
human GTP-cyclohydrolase I in a rat model of Parkin- bamylase deficient patient following adenoviral gene
sons disease. J Neurosci 18 (11): 42714284. transfer. Mol Genet Metab 80 (1-2): 148158.
Marsden CD (1990). Parkinsons disease. Lancet 335 (8695): Sanchez-Pernaute R, Harvey-White J, Cunningham J et al.
948952. (2001). Functional effect of adeno-associated virus mediated
Marsden CD, Parkes JD (1977). Success and problems of gene transfer of aromatic L-amino acid decarboxylase into
long-term levodopa therapy in Parkinsons disease. Lancet the striatum of 6-OHDA-lesioned rats. Mol Ther 4 (4):
1 (8007): 345349. 324330.
Mufson EJ, Kroin JS, Sendera TJ et al. (1999). Distribution Schumacher JM, Ellias SA, Palmer EP et al. (2000). Trans-
and retrograde transport of trophic factors in the central plantation of embryonic porcine mesencephalic tissue in
nervous system: functional implications for the treatment patients with PD. Neurology 54 (5): 10421050.
304 B. DASS AND J. H. KORDOWER

Shen Y, Muramatsu SI, Ikeguchi K et al. (2000). Triple factor in the adult nigrostriatal system suggests a trophic
transduction with adeno-associated virus vectors expres- role in the adult. Proc Natl Acad Sci USA 92 (18):
sing tyrosine hydroxylase, aromatic-L-amino-acid decar- 82748278.
boxylase, and GTP cyclohydrolase I for gene therapy Wang L, Muramatsu S, Lu Y et al. (2002). Delayed delivery
of Parkinsons disease. Hum Gene Ther 11 (11): of AAV-GDNF prevents nigral neurodegeneration and
15091519. promotes functional recovery in a rat model of Parkinsons
Slevin JT, Gerhardt GA, Smith CD et al. (2005). Improve- disease. Gene Ther 9 (6): 381389.
ment of bilateral motor functions in patients with Parkin- Wenning GK, Odin P, Morrish P et al. (1997). Short- and
son disease through the unilateral intraputaminal infusion long-term survival and function of unilateral intrastriatal
of glial cell line-derived neurotrophic factor. J Neurosurg dopaminergic grafts in Parkinsons disease. Ann Neurol
102 (2): 216222. 42 (1): 95107.
Smith AD, Kozlowski DA, Bohn MC et al. (2005). Effect of Wolff JA, Fisher LJ, Xu L et al. (1989). Grafting fibroblasts
AdGDNF on dopaminergic neurotransmission in the stria- genetically modified to produce L-DOPA in a rat model of
tum of 6-OHDA-treated rats. Exp Neurol 193 (2): Parkinson disease. Proc Natl Acad Sci USA 86 (22):
420426. 90119014.
Steece-Collier K, Collier TJ, Danielson PD et al. (2003). Yamada M, Oligino T, Mata M et al. (1999). Herpes simplex
Embryonic mesencephalic grafts increase levodopa- virus vector-mediated expression of Bcl-2 prevents 6-
induced forelimb hyperkinesia in parkinsonian rats. Mov hydroxydopamine-induced degeneration of neurons in the
Disord 18: 14421454. substantia nigra in vivo. Proc Natl Acad Sci USA 96 (7):
Svendsen CN, Clarke DJ, Rosser AE et al. (1996). Survival 40784083.
and differentiation of rat and human epidermal growth
factor-responsive precursor cells following grafting into Further Reading
the lesioned adult central nervous system. Exp Neurol Maries E, Kordower JH, Chu Y et al. (2006). Focal not wide-
137 (2): 376388. spread grafts induce novel dyskinetic behavior in parkinsonian
Tomac A, Widenfalk J, Lin LF et al. (1995). Retrograde rats. Neurobiol Dis 21 (1): 165180. [Epub 2005 Aug 10].
axonal transport of glial cell line-derived neurotrophic
Section 8

Other parkinsonian syndromes


Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 46

Multiple system atrophy

RONALD F. PFEIFFER*

University of Tennessee Health Science Center, Memphis, TN, USA

46.1. Introduction with neuronal loss and gliosis in the putamen and cau-
date and loss of pigmented neurons in the substantia
The nosological history of what is now known as mul- nigra were evident, but Lewy bodies were conspicuously
tiple system atrophy (MSA) brings to mind the well- absent. Described in more detail 1 year later (Adams
known ancient Buddhist parable of the blind men et al., 1961), cerebellar dysfunction, pyramidal tract
examining an elephant. In the parable, each of the abnormalities and autonomic impairment were also pre-
blind men feels a different part of the elephant and sent in these individuals. Although the original authors
then, unable to visualize the entire beast, describes applied the appellation striopallidonigral degeneration
the elephant in strikingly different terms. MSA might to the condition, the abbreviated label of striatonigral
be considered the movement disorders elephant. degeneration (SND) was subsequently employed. Thus,
Descriptions of the three parts of the MSA elephant it came to be that three syndromes sporadic OPCA with
date back to the end of the 19th century, when Dejer- predominantly cerebellar dysfunction, ShyDrager syn-
ine and Thomas (1900) described two individuals drome (SDS) with striking autonomic impairment and
with a sporadic, adult-onset syndrome characterized SND with rigid-akinetic parkinsonism existed side by
primarily by ataxia, but also displaying parkinsonism, side in the neurological nomenclature as three distinct
brisk reflexes and incontinence. The autopsy findings entities.
of atrophy in the inferior olives, pons, middle cerebel- The conceptual synthesis of these three entities into a
lar peduncles and cerebellum led them to apply the single pathological process was initiated by Graham and
label of olivopontocerebellar atrophy (OPCA) to the Oppenheimer in 1969, when they described an indivi-
condition. Wenning and colleagues (1997) in their dual with progressive autonomic failure and ataxia.
review of 203 cases of pathologically proven cases of In discussing the case, they noted the clinical and patho-
MSA document even earlier descriptions by Schultze logical similarities between OPCA, SDS and SND and
(1887) and Arndt (1894) of individuals with cerebellar suggested that the term multiple system atrophy be
syndromes that would now be called MSA. In 1960, used for them all. This suggestion did not meet with
two additional conditions that would later be rec- immediate or universal acceptance and employment of
ognized as MSA were described. Shy and Drager the older three terms in publications continued for many
(1960) reported two individuals with progressive auto- years. However, the discovery of a distinctive patholo-
nomic failure accompanied by parkinsonism, dysar- gical marker, glial cytoplasmic inclusions (GCI), by
thria, ataxia, pyramidal signs, distal muscle wasting Papp and colleagues in 1989 finally provided a firm
and extraocular muscle weakness. Autopsy in one of basis for the designation of MSA as a distinct disease
the cases demonstrated degenerative changes in the process with a complex combination of clinical presen-
caudate, substantia nigra, cerebellum, inferior olives, tations (Papp et al., 1989). The subsequent identifica-
locus ceruleus and intermediolateral cell columns. In tion of a-synuclein as a major component of GCI has
the same year, van der Eecken and colleagues (1960) established MSA as a member of the family of synuclei-
detailed unique pathological findings in 3 patients with nopathies (Gai et al., 1998; Wakabayashi et al., 1998;
predominantly rigid-akinetic parkinsonism. Atrophy Dickson et al., 1999a).

*Correspondence to: Ronald F. Pfeiffer, Department of Neurology, University of Tennessee Health Science Center, 855
Monroe Avenue, Memphis TN 38163, USA. E-mail: rpfeiffer@utmem.edu, Tel: 1-901-448-5209, Fax: 1-901-448-7440.
308 R. F. PFEIFFER
The confusing clinical picture of MSA, with its male-to-female ratio of 1.3:1 in their 203 cases of
diversity of clinical presentations and the absence of MSA; Bower and colleagues (1997) a ratio of 2:1 in
any unequivocally diagnostic clinical feature or defini- 9 patients. The mean age of onset of symptoms of
tive laboratory test, has prompted formation of clinical MSA is somewhat younger than that of Parkinsons
diagnostic criteria to aid in diagnosis. The first effort disease. In most studies, the mean age of onset of
was undertaken by Quinn in 1989 (Quinn, 1989a), fol- symptoms has hovered between 53 and 56, with a
lowed a decade later by the development of the range of 3178 years (Saito et al., 1994; Wenning
currently employed Consensus Criteria assembled by et al., 1994, 1997; Ben-Shlomo et al., 1997; Testa
Gilman and colleagues (1998, 1999). et al., 2001; Watanabe et al., 2002). Quinn (2005)
This chapter will address the epidemiology, clini- has made the point that there has never been a case
cal features, pathological characteristics, diagnostic of pathologically proven MSA with onset of symptoms
evaluation and treatment approaches for MSA. before age 30. Although there is a case report of an
individual with very slowly progressive probable
MSA who experienced the onset of symptoms at age
46.2. Epidemiology
18 (Schatz, (2003)), the accuracy of the clinical diag-
46.2.1. Descriptive epidemiology nosis is thrown into some doubt by the unusually pro-
longed survival (40 years) of that patient. The report
MSA is a rare disease and relatively few formal stu- of Bower and colleagues (1997) stands somewhat
dies of its prevalence have been completed. In two stu- apart from the other studies, with a more advanced
dies reported by the same group of investigators, the median age of symptom onset of 66, with a range of
prevalence rate of MSA was approximately 1.9 per 5182 years.
100 000 (Tison et al., 2000; Chrysostome et al., In addition to earlier age of onset, individuals with
2004). In a report by another group of investigators, MSA possess a considerably poorer prognosis than per-
the crude prevalence (including probable and possible sons with Parkinsons disease, with more rapid dete-
cases) was 3.3 per 100 000 and the age-adjusted preva- rioration in neurological function. The mean survival
lence 4.4 per 100 000 (Schrag et al., 1999). Schrag and time for someone with MSA is only 67 years (Ben-
colleagues (1999) also extrapolated MSA prevalence Shlomo et al., 1997; Wenning et al., 1997; Kurisaki,
rates of 2.3 per 100 000 (Wermuth et al., 1997) and 1999). Viewed from another standpoint, survival rates
4.9 per 100 000 (Chio et al., 1998) from two studies of 90% at 3 years, 83.5% at 5 years, 54% at 6 years
that were primarily investigating the prevalence of and 2939.9% at 10 years have been documented (Saito
Parkinsons disease in the Faroe Islands and in north- et al., 1994; Testa et al., 2001; Watanabe et al., 2002).
west Italy respectively. Not all studies have arrived Watanabe and colleagues (2002) also reported median
at rates this low. Trenkwalder and colleagues (1995) intervals from disease onset until requiring aids for
investigated the prevalence of different types of par- walking to be 3 years, to wheelchair requirement 5 years
kinsonism in individuals over age 65 by performing a and to being bed-ridden 8 years.
door-to-door survey in two Bavarian villages and
found a markedly higher prevalence of MSA, with a 46.2.2. Analytical epidemiology
rate over 300 per 100 000. Whether this difference is
due to the age limitation employed in this study, its MSA is generally considered a sporadic disease.
door-to-door design leading to improved ascertain- According to the Consensus Criteria formulated for
ment, or simple chance is unclear. the diagnosis of MSA (Gilman et al., 1998, 1999;
Studies of annual incidence of MSA are equally Quinn, 2005), a family history of a similar disorder is
scarce. Bower and colleagues (1997) employed the considered an exclusion criterion. Nevertheless, some
medical records linkage system of the Rochester Epi- reports have suggested the possibility of a genetic
demiology Project to estimate an overall annual in- component to MSA. Nee and colleagues (1991) com-
cidence rate of 0.6 per 100 000; in their study no pleted a case-control study of 60 patients with MSA
patient developed MSA prior to age 50, whereas and 60 control subjects and noted that first-degree
between ages 50 and 99 incidence was 3.0 per 100 relatives of persons with MSA were more likely to
000. Schrag and colleagues (1999) obtained a similar report clinical symptoms of MSA than were relatives
figure (approximately 0.5 per 100 000) when they of the control subjects.
indirectly measured MSA annual incidence. There have also been several descriptions of
Although MSA affects both sexes, studies seem to families with possible autosomal-dominant MSA.
indicate that men are at somewhat higher risk than Lewis (1964) reported a kindred with familial ortho-
women. Wenning and colleagues (1997) reported a static hypotension (the term multiple system atrophy
MULTIPLE SYSTEM ATROPHY 309
had not yet been created) involving six family mem- (1991) performed a case-control study of 60 indivi-
bers over three generations. However, the accuracy duals with MSA and 60 controls, using a self-adminis-
of the portrayal of this family as having MSA has tered questionnaire and found statistically significant
recently been questioned (Berciano and Wenning, increased exposure to metal dusts and fumes (OR
2005). More recently, another family with an autoso- 14.75), pesticides (OR 5.8), plastic monomers and
mal-dominant disorder possessing the clinical phenotype additives (OR 5.25) and organic solvents (OR
of MSA has been reported (Wullner et al., 2004). 2.41) in the MSA group. A more recent case-control
Although no pathological confirmation of the diagnosis study, using the Consensus Criteria for the diagnosis
of MSA is yet available in this family, extensive clinical of MSA, has been carried out by the European Study
testing that has included autonomic function testing, Group for Atypical Parkinsonism (Vanacore et al.,
magnetic resonance imaging (MRI) and even single- 2001, 2005). A total of 73 patients with MSA, 73
photon emission computed tomography (SPECT) in sev- population controls and 146 hospital controls with
eral affected individuals has been consistent with a diag- non-neurodegenerative neurological diseases were
nosis of MSA. enrolled. A statistically significant difference between
The possibility that genetic polymorphisms might the MSA group and both the population and hospital
play a role in the development of MSA has received control groups was uncovered for an occupational his-
considerable attention in recent years but the search tory of farming (adjusted OR 2.52 versus hospital
thus far has been largely fruitless. Multiple studies, controls; adjusted OR 4.53 versus population
looking at multiple genes (cytochrome P4502D6, controls). Additionally, a doseresponse effect was
cytochrome P4501A1, N-acetyltransferase 2, dopa- demonstrable with regard to duration of farming
mine transporter, glutathione S-transferase M1, ciliary exposure. The specific component(s) of farming
neurotrophic factor, insulin-like growth factor-1, exposure responsible for the increased risk could not
hiGIRK2, HLA-A32, a-synuclein, apolipoprotein E, be determined from this study, but the authors nomi-
synphilin, tau, alcohol dehydrogenase 7, UCHL-1) nated pesticides, solvents and mycotoxins as candi-
have not demonstrated any association with the dev- date risk factors for MSA. In the same study,
elopment of MSA (Plante-Bordeneuve et al., 1995; increased but not statistically significant ORs for
Bandmann et al., 1997; Cairns et al., 1997; Nicholl several other occupational categories (iron and steel,
et al., 1999; Morris et al., 2000; Kim and Lee, 2003; textile and clothing, leather and shoe) were also
Healy et al., 2005). However, Combarros and collea- documented. In another investigation in which medi-
gues (2003) have reported that individuals homo- cal records of 100 patients with MSA were reviewed,
zygous for interleukin-1A (IL-1A) allele 2 in the significant environmental toxin exposure was docu-
regulatory region of the IL-1A gene have a fivefold mented in 11 individuals (Hanna et al., 1999). No
(odds ratio (OR) 5.1) increased risk of developing single suspect toxin was again identified, but a his-
MSA; persons with only one copy of the allele also tory of significant exposure to pesticides was docu-
have an increased risk (OR 1.6), but considerably mented in 6 of the 11 individuals, and exposure to a
less than those with two copies. Even more recently, variety of other chemicals in the other 5. Prolonged
the same group of investigators has provided evidence occupational exposure to carbon disulfide in a person
that the IL-8 and intercellular adhesion molecule-1 with MSA has also been reported (Frumkin, 1998).
(ICAM-1) genes may also be susceptibility genes for In contrast, Chrysostome and colleagues (2004) did
MSA (Infante et al., 2005). The presence of two not find any relationship between occupational expo-
copies of the IL-8 T allele increased the risk of sure to pesticides and MSA in their study of 50 MSA
MSA over threefold (OR 3.89). Polymorphism of patients, 50 Parkinsons disease patients and 50
the ICAM-1 gene did not independently affect the risk healthy controls.
of MSA, but individuals carrying two copies of the As has been reported with Parkinsons disease, an
ICAM-1 K allele and two copies of the IL-8 T allele inverse association between MSA and smoking has
had an even higher risk of MSA (OR 11.0). Since been identified by several groups of investigators
both studies involved relatively small numbers of (Vanacore et al., 2000; Chrysostome et al., 2004).
MSA patients (30 and 41 respectively), the authors The mechanism by which smoking might reduce the
point out that further studies, both to confirm their risk of MSA is not known. One patient has been
findings and to investigate the potential mechanisms reported in whom the symptoms of MSA were exacer-
by which these polymorphisms might act to increase bated by cigarette smoking (Johnsen and Miller,
risk of MSA, need to be undertaken. 1986), but this phenomenon could not account for the
Possible environmental risk factors for MSA have higher numbers of never-smokers in the ranks of
also received some attention. Nee and colleagues MSA patients.
310 R. F. PFEIFFER
46.3. Primary clinical features tremor does not occur in MSA. It does. In fact, tremor
of some type develops in the majority of patients; even
The pleomorphic propensity of MSA produces a rest tremor is seen in 2940% of cases, although a
potentially confusing picture for the clinician, who classic pill-rolling tremor is evident in only 79%
can be faced with a bewildering array of symptoms (Colosimo et al., 2005a). What has been branded tre-
and signs in the patient with MSA. The difficulty in mor in MSA, however, may not always actually be
diagnosing MSA was amply illustrated in a study so. The term jerky tremor is sometimes used to
carried out by Litvan and colleagues (1997), in which describe postural tremor in MSA (Gouider-Khouja
the diagnostic accuracy of the treating neurologist in et al., 1995; Bower, 2000), but accelerometric and
16 patients with autopsy-proven MSA had been only electromyographic recordings of such small-ampli-
25% at the first clinic visit and 50% at the final visit. tude, non-rhythmic movements of the fingers that
In this and another series, however, the diagnostic occur when holding a posture or initiating movement
accuracy of movement disorders specialists was demonstrate that they are actually a form of postu-
considerably higher (Hughes et al., 2002). ral and action myoclonus (labeled minipolymyocl-
The clinical features of MSA generally fall into onus by the authors) rather than tremor (Salazar
four categories: extrapyramidal, cerebellar, autonomic et al., 2000).
and pyramidal. Individuals may display any combina- Several other misconceptions have crept into the
tion of these features and do not necessarily develop neurological mindset regarding the parkinsonian fea-
all of them during the course of their illness. In a study tures of MSA. Recognition that asymmetric onset of
of 203 cases of autopsy-proven MSA drawn from pub- symptoms or signs is more common in Parkinsons
lished literature, Wenning and colleagues (1997) found disease than in the atypical parkinsonian disorders
that parkinsonism was present in 87% of patients dur- (Hughes et al., 1992a) has sometimes been miscon-
ing the course of their illness, autonomic failure in strued to mean that the atypical syndromes, such as
74%, cerebellar signs in 54% and pyramidal signs in MSA, never present asymmetrically. In fact, bilateral
49%. In the same study, parkinsonism was the initial symmetric onset of motor dysfunction actually occurs
motor disturbance in 58% of the patients, cerebellar in only a minority of MSA patients (Colosimo et al.,
ataxia in 29% and a mixture of the two in 9%; pyrami- 1995, 2005a). The idea that MSA is invariably levo-
dal signs were the initial motor feature in only 3% of dopa-unresponsive is also incorrect. Response to levo-
individuals. In an earlier analysis of 100 MSA cases, dopa, occasionally good or even excellent, but often
autonomic symptoms were the initial clinical feature suboptimal and fleeting, is evident in approximately
in 41% of patients, parkinsonism in 46% and cerebel- 3060% of patients (Parati et al., 1993; Boesch et al.,
lar symptoms or signs in only 5% (Wenning et al., 2002; Wenning et al., 2003a; Christine and Aminoff,
1994). Gilman (2002) observes, however, that the pre- 2004). The experience of Tison and colleagues
dominance of parkinsonism in the latter series may (2002), however, stands in some contrast in that they
have reflected sampling bias since the patients were noted a subjective impression of greater than 50%
derived from a movement disorders clinic. improvement in motor function in fewer than 10% of
The proclivity of MSA to present with predomi- their MSA subjects.
nantly parkinsonian or predominantly cerebellar motor Balance impairment with postural instability and fre-
features has led to the convention of classifying MSA quent falling occur earlier and with greater frequency in
as being of either the MSA-P or MSA-C type (Quinn, MSA than in Parkinsons disease. Tison and colleagues
1989a, 2005). The distinction is somewhat artificial, (2002) elicited a history of falling at symptom onset in
however, since most patients eventually acquire both 20% of the 50 MSA patients they studied, compared
extrapyramidal and cerebellar dysfunction. Autonomic with 2% of the 50 individuals with Parkinsons disease;
features eventually develop in both groups. postural instability was present on testing in 10% of the
MSA patients at the initial exam, but in only 2% of the
46.3.1. Extrapyramidal features Parkinsons disease subjects. With disease progression
over 70% of MSA patients develop postural instability
46.3.1.1. Parkinsonism (Tison et al., 2002).
Parkinsonism is by far the most common extrapyra-
midal feature of MSA. Most frequently, it is character-
ized by rigidity and bradykinesia/akinesia. In the 46.3.1.2. Dystonia
literature series of Wenning and colleagues (1997), Dystonia is another facet of extrapyramidal dysfunc-
akinesia was noted to be present in 83% of patients tion that surfaces in MSA. Although reported to be
and rigidity in 63%. It is a common misconception that present relatively infrequently by some investigators
MULTIPLE SYSTEM ATROPHY 311
(Rivest et al., 1990; Wenning et al., 1997) and not at Although most often considered a dystonic phenom-
all by others (Adams and Salam-Adams, 1986), enon, some have suggested that antecollis in MSA
Boesch and colleagues (2002), in a prospective study may sometimes be a manifestation of neck extensor
of 24 individuals with MSA followed for up to 10 myopathy (Askmark et al., 2001). Extreme forward
years, documented the development of dystonia prior flexion of the spine that appears or increases when the
to any levodopa exposure in 46%. In individuals with individual is walking and disappears when supine
MSA who responded to levodopa, the emergence of (camptocormia) and laterally oriented trunk flexion
levodopa-induced dystonic dyskinesia was also fre- (Pisa syndrome) may also develop in the setting
quent. In an accompanying editorial, Riley (2002) of MSA (Slawek et al., 2006). It is also unsettled
made the point that, if actively sought, dystonia whether these phenomena represent axial dystonia or
may actually be one of the most common features of axial myopathy.
MSA. Head and neck are the most common sites for
dystonia development in MSA, in both levodopa-naive 46.3.2. Autonomic dysfunction
and levodopa-induced settings, but limb involvement
also occurs. Several patterns of dystonia are particu- Impairment of autonomic function eventually develops
larly notable in MSA. Dystonia involving orofacial in the vast majority of individuals with MSA. As noted
and platysma muscles may produce a tetanus-like earlier, autonomic symptoms were the presenting clin-
facial distortion that has been christened the risus sar- ical feature in 41% of 100 patients with clinically
donicus of MSA (Wenning et al., 2003b). Two addi- probable MSA and they ultimately developed in 97%
tional phenomena have generated some controversy (Wenning et al., 1994). Similarly, in an analysis of
as to whether they represent dystonic phenomena or 35 subjects with pathologically proven MSA, auto-
not: stridor and disproportionate antecollis. nomic failure had been present in 97% (Wenning
Inspiratory stridor, typically occurring at night, et al., 1995a). Among 75 persons with MSA followed
develops in 1334% of patients with MSA (Wenning at the Mayo Clinic, severe autonomic dysfunction
et al., 1994, 1997). Although most often considered a developed in 97% also (Sandroni et al., 1991). Geni-
later manifestation, one group of investigators noted tourinary, cardiovascular, gastrointestinal (GI) and
stridor to be the initial clinical feature in 4% of 200 thermoregulatory autonomic dysfunction may all
consecutive patients with probable MSA (Uzawa appear in the setting of MSA.
et al., 2005) and in another study 69% of individuals
who developed stridor did so within the first 4 years 46.3.2.1. Genital dysfunction
of disease diagnosis (Yamaguchi et al., 2003). Con-
Erectile dysfunction is almost universal in men with
flicting opinions exist regarding the genesis of stridor
MSA. It ultimately develops in 96% and is the initial
in MSA. Some investigators attribute it to laryngeal
clinical feature in 37% (Beck et al., 1994). It typically
abductor paralysis (Bannister et al., 1981; Iranzo
precedes the development of both urinary dysfunction
et al., 2000, 2004; Uzawa et al., 2005), whereas others
and orthostatic hypotension (Kirchhof et al., 2003).
maintain, and provide electromyographic documenta-
Oertel and colleagues (2003) studied genital sensation
tion, that stridor is a dystonic phenomenon, with sus-
(as a possible equivalent of erectile dysfunction) in 17
tained contraction of vocal cord adductors (Simpson
women with MSA and found reduced genital sensitiv-
et al., 1992; Merlo et al., 2002). Regardless of its etiol-
ity in 47%, compared with only 4% of 27 women with
ogy, the presence of stridor in an individual with MSA
Parkinsons disease and 4% of 26 normal controls. In
should be interpreted as a danger signal. Persons with
most of the women with MSA, the reduced genital
stridor have an increased mortality risk, which has
sensitivity had preceded the onset of motor dysfunc-
led to the suggestion that tracheostomy be considered
tion and in many it had developed subacutely over a
in all individuals in whom stridor develops (Silber
period of several months.
and Levine, 2000).
Unusually prominent neck flexion resulting in a
dropped-head syndrome, or disproportionate antecol- 46.3.2.2. Urinary dysfunction
lis, has been widely recognized as a feature suggestive Disordered bladder function is also common in MSA. It
of, though not unique to, MSA (Quinn, 1989b, may be the initial clinical feature (Sakakibara et al.,
2005). Disproportionate antecollis is uncomfortable 1993) and routinely is one of the earliest autonomic fea-
for patients; it may also interfere with eating and speak- tures to emerge (Mabuchi et al., 2005). Over 90% of
ing and even effectively impair vision (Colosimo et al., individuals with MSA develop micturition disturbances
2005a). As with stridor, the etiology of disproportionate during the course of the disease (Sakakibara et al.,
antecollis in MSA has been the source of controversy. 1993, 2000). Both urinary retention and incontinence
312 R. F. PFEIFFER
may occur. The most frequent urinary symptom in per- autonomic responses normally triggered by increased
sons with MSA is difficulty voiding, which Sakakibara superior mesenteric artery blood flow (Mathias, 2005)
and colleagues (2000) catalogued in 79% of the 121 and is more prominent in MSA than in Parkinsons
patients they studied. In the same study, nocturnal urin- disease (Thomaides et al., 1993).
ary frequency was present in 74%, urgency in 63%,
urge incontinence in 63% and urinary retention in 8%.
46.3.2.4. Gastrointestinal dysfunction
Others have described urinary incontinence in 5571%
and urinary retention in 1827% of individuals with Although considerable ink has flowed concerning
MSA (Wenning et al., 1994, 1997). Urodynamic and electromyographic abnormalities of the anal sphincter
neurophysiological testing also presents a diverse pic- in MSA (discussed below), relatively little has been
ture; both hyperactive and hypoactive bladder dys- written about clinical GI dysfunction. Nevertheless,
function occur. Sakakibara and colleagues (2000) both upper and lower GI disturbances occur frequently
documented detrusor hyperreflexia in 56% of the in MSA. Dysphagia is common in MSA and tends to
MSA patients they studied. In contrast, some other develop earlier than in Parkinsons disease. In one
investigators have found detrusor hyporeflexia to be study, the median latency to the onset of dysphagia
more frequent (Bonnet et al., 1997). Sakakibara and in 15 patients with MSA was 24 months (Muller
colleagues (2000, 2001) also reported the presence of et al., 2001). Higo and colleagues (2003) documented
a low-compliance bladder in 31%, an atonic curve in reduced oropharyngeal and hypopharyngeal swallow-
5% and detrusor-sphincter dyssynergia in 45% of the ing pressures in 29 persons with MSA and incomplete
persons with MSA they studied. Increased postmicturi- relaxation of the upper esophageal sphincter was pre-
tion residual urine (>30 ml) was evident in 74% of sent in 23%. Gastric emptying may also be delayed
patients and residual urine of over 100 ml was noted in individuals with MSA, comparable to that seen in
in 52%. In MSA, urinary disturbances typically deve- Parkinsons disease (Thomaides et al., 2005).
lop earlier and are more severe than those seen in Bowel dysfunction, including both decreased bowel
Parkinsons disease. movement frequency and difficulty with the act of
defecation itself, is another aspect of GI dysfunction
in MSA. In one study, decreased bowel movement fre-
46.3.2.3. Cardiovascular dysfunction quency (fewer than 3/week) was present in 60% of
Cardiovascular dysfunction in the form of orthostatic participating MSA patients, and 67% experienced dif-
hypotension is the autonomic feature that has attracted ficulty with defecation (Sakakibara et al., 2004a). In
the most attention in MSA, but it actually occurs less another study of 16 persons with MSA experiencing
frequently than urogenital dysfunction. Symptoms of GI symptoms, 87% were having fewer than 3 bowel
orthostatic hypotension develop in 4368% of in- movements per week and 69% were experiencing
dividuals with MSA (Wenning et al., 1994, 1997; difficulty with defecation (Stocchi et al., 2000).
Sakakibara et al., 2000). Postural lightheadedness is Decreased bowel movement frequency in MSA is
the most frequently experienced symptom of ortho- due to slowed colon transit time. Sakakibara and
static hypotension, but individuals may instead experi- colleagues (2004a) documented a mean total colon
ence blurred vision, clouded cognition, head and neck transit time of 71.8 hours in 15 individuals with
pain in a coat-hanger distribution, lower back or but- MSA, compared with 39 hours in 10 age-matched con-
tock pain, a sense of weakness or fatigue, tremulous- trols. Disturbances in anorectal function, leading to
ness, or simply an ill-defined feeling of dizziness increased straining and incomplete evacuation during
(Mathias et al., 1999; Bower, 2000; Mathias, defecation, are also present in many patients with
2005). Syncope occurs in 1519% of MSA patients MSA. Manometric and electromyographic recordings
with orthostatic hypotension (Wenning et al., 1994, of anorectal function demonstrate a plethora of abnor-
1997; Sakakibara et al., 2000). Orthostatic hypotension malities that suggests these disturbances are due to
may be present and become symptomatic early in the either paradoxical contraction or insufficient inhibition
course of MSA, a characteristic that provides some of the anal musculature during straining, along with
contrast with Parkinsons disease (Wenning et al., weakness of associated abdominal muscle contraction
1999). A variety of factors can accentuate and magnify (Stocchi et al., 2000; Sakakibara et al., 2004a). Fecal
orthostatic hypotension. Symptoms upon first arising incontinence, especially following laxative ingestion,
in the morning can be prominent, consequent to pro- may also occur in 1231% of patients with MSA
longed recumbency. Postprandial hypotension, espe- (Wenning et al., 1997; Stocchi et al., 2000; Sakakibara
cially following a large, carbohydrate-heavy meal, et al., 2004a). Denervation, with consequent weakness
may also occur, due to impairment of compensatory of the external anal sphincter, is probably responsible.
MULTIPLE SYSTEM ATROPHY 313
46.3.2.5. Thermoregulatory dysfunction pyramidal tract dysfunction on neurological examina-
A number of features indicative of thermoregulatory tion, in contrast, are quite frequently present. In the
and sudomotor dysfunction have been identified in series of 203 MSA patients assembled from the litera-
MSA. Reduced sweating is common and may be severe ture by Wenning and colleagues (1997), hyperreflexia
(Cohen et al., 1987; Kumazawa et al., 1989). Indivi- was present in 46% and Babinski responses in 41%,
duals with MSA demonstrate impaired heat tolerance but spasticity in only 10%. Although the clinical con-
(Colosimo et al., 2005a) and may be at risk for sequences of pyramidal tract dysfunction in MSA are
increased morbidity due to worsened orthostatic hypo- not striking, the presence of unequivocal pyramidal
tension during heat exposure (Pathak et al., 2005). tract signs in an individual with parkinsonism and
Reduced skin temperature may also be present in per- autonomic dysfunction is of diagnostic utility in that
sons with MSA, along with a more profound reduction it points toward a diagnosis of MSA rather than
in skin temperature when exposed to cold (Klein Parkinsons disease.
et al., 1997). These abnormalities may, in turn, account
for the presence of cold, blue or purple hands (and feet) 46.4. Other clinical features
that has been christened as one of the red flags of
MSA (Klein et al., 1997). Raynauds phenomenon, with 46.4.1. Disordered sleep
painful, pale fingers and hands due to vasospasm,
induced by cold or by ergot drugs, may also afflict indi- Recognition is growing that a variety of sleep disor-
viduals with MSA (Kaufmann and Biaggioni, 2003; ders may emerge during the course of MSA. In a ques-
Geser and Wenning, 2005). One further reflection of tionnaire study involving 57 patients with MSA, 70%
impaired thermoregulatory control in MSA is the blunt- voiced complaints of sleep impairment (Ghorayeb
ing of the physiological fall in body temperature that et al., 2002). In the study, 60% were aware of night-
normally occurs at night (Pierangeli et al., 2001). time vocalizations, 52.5% experienced sleep fragmen-
tation, 47.5% described symptoms of rapid-eye
46.3.3. Cerebellar dysfunction movement (REM) sleep behavior disorder, 32.5%
were troubled by early awakening, 20% by insomnia,
In patients evaluated in movement disorders clinics, cer- 19% experienced stridor and 12.5% were kept awake
ebellar dysfunction is only rarely the presenting clinical by symptoms of restless-legs syndrome. Snoring was
feature of MSA, accounting for only 5% in one study also acknowledged by 72.5%. Persons with MSA are
(Wenning et al., 1994). Gilman (2002) points out, how- also more likely to experience excessive daytime slee-
ever, that this may reflect a sampling bias and implies piness than individuals with Parkinsons disease
that the percentage might be greater in a general neurol- (Ghorayeb et al., 2002). Video-polysomnographic
ogy clinic. With disease progression, the percentage of recordings confirm and expand the information gath-
individuals displaying cerebellar dysfunction expands ered from questionnaire studies. In a study of 19
considerably. Gait ataxia eventually becomes evident in MSA patients who were not selected on the basis of
almost 50% of individuals and dysarthria in almost any sleep or respiratory problems, inspiratory noise
everyone (Wenning et al., 1997; Shulman et al., 2004). was documented in every participant, with stridor in
A quintet of abnormalities characterizes cerebellar dys- 42% and snoring in 100% (Vetrugno et al., 2004). In
function in MSA: gait ataxia, limb ataxia, kinetic tremor, fact, subjects in this study spent 50% of their sleep
oculomotor dysfunction and dysarthria (Wenning et al., time either snoring or with stridor. Abnormal sleep
1997, 2004; Shulman et al., 2004; Colosimo et al., structure, with reduced non-REM deep sleep and
2005a). Not all of the features develop in each individual decreased sleep efficiency but normal amounts of
and any combination of them may occur. In one study, REM sleep, were also evident.
gait ataxia eventually became evident in 49% of subjects, REM sleep behavior disorder is now recognized as
limb ataxia in 47%, intention tremor in 24% and nystag- a feature of a-synucleinopathies such as MSA, Parkin-
mus in 23% (Wenning et al., 1997). In addition to ny- sons disease and dementia with Lewy bodies (Boeve
stagmus, other oculomotor abnormalities seen in MSA et al., 2001), but it appears to develop earlier and with
include ocular dysmetria, impaired smooth pursuit and greater severity in individuals with MSA (Iranzo et al.,
square-wave jerks (Bower, 2000). 2005). Manifestations of REM sleep behavior disorder
can be present early in the course of MSA, prior to
46.3.4. Pyramidal signs institution of any antiparkinson medication (Wetter
et al., 2000) and may even antedate the appearance
Clinically apparent pyramidal tract dysfunction, such of motor and autonomic features (Tison et al., 1995;
as spastic paraparesis, is unusual in MSA. Signs of Boeve et al., 2001). In one study, 44% of the MSA
314 R. F. PFEIFFER
patients experienced the symptoms of REM sleep with head turning, distinct from orthostatic light-
behavior disorder more than 1 year before the onset headedness, has been described in individuals with
of motor features (Plazzi et al., 1997). Polysomno- the cerebellar form of MSA (Sakakibara et al., 2004b).
graphic recordings can be very useful in establishing
the presence of REM sleep behavior disorder in 46.5. Consensus criteria
MSA patients and have revealed a considerably higher
frequency of the abnormality than subjective question- The diversity of clinical features of MSA, their vari-
naire studies. In a study of 39 consecutive MSA able mode of presentation and the absence of any
patients reported by Plazzi and colleagues (1997), unequivocal diagnostic test for MSA combine to make
polysomnographic evidence of REM sleep behavior the clinical diagnosis of MSA a difficult, even trea-
disorder was present in 90%. It has been suggested that cherous exercise. In response to this diagnostic quick-
increased or excessive sleep talking may be an early sand, a conference was convened in 1998, in which
manifestation of REM sleep behavior disorder in guidelines for the diagnosis were developed (Gilman
MSA (Tachibana et al., 1997). et al., 1998, 1999). The Consensus Criteria are detailed
in Tables 46.146.3. Three diagnostic categories are
46.4.2. Behavioral disorders employed: possible MSA, probable MSA and definite
MSA (Table 46.1). Pathological confirmation is
Frank dementia is not a feature of MSA and, in fact, is required for a diagnosis of definite MSA. Therefore,
one of the exclusionary criteria for the diagnosis in the clinical setting the only possible diagnoses are
(Gilman et al., 1998, 1999). However, behavioral possible or probable MSA and determination between
changes do occur in the setting of MSA and may actu- these two categories is dependent upon the number
ally be quite common. Executive dysfunction, similar of features within the four clinical domains that are
to but more severe than that seen in Parkinsons dis- met (Table 46.2). The third component of the consen-
ease, has been documented in MSA (Dujardin et al., sus criteria is a list of exclusion criteria; the presence
2003; Lange et al., 2003). Persons with MSA fre- of any feature listed therein prohibits a diagnosis of
quently suffer from depression. In a study of 99 indivi- MSA (Table 46.3).
duals with MSA, Benrud-Larson and colleagues
(2005) documented symptoms of at least mild depres-
sion in 80%, with moderate to severe depression in 46.6. Neuropathology
39%. Emotional blunting or apathy has also been 46.6.1. Macroscopic pathology
described in persons with MSA. In one small study,
this was evident in 92% of 12 individuals evaluated Involvement of both basal ganglia and cerebellum may
(Fetoni et al., 1999). be evident on gross examination of the brain in MSA.

46.4.3. Other features


Table 46.1
A number of other clinical features have been
described in persons with MSA, although they have Diagnostic categories of multiple system atrophy (MSA)
not received extensive scrutiny. Although impairment
I. Possible MSA
of olfaction is often present in patients with MSA, it One criterion plus two features from separate other
is typically mild compared with the deficits character- domains
istic of Parkinsons disease (Nee et al., 1993; Wenning When the criterion is parkinsonism, a poor levodopa
et al., 1995b; Muller et al., 2001). Pramstaller and response qualifies as one feature (hence only one
colleagues (1995) documented electromyographic additional feature is required)
abnormalities consistent with peripheral neuropathy II. Probable MSA
in 40% of the 74 patients with MSA they studied. In Criterion for autonomic failure/urinary dysfunction plus
a single case report, sensory polyneuropathy was the poorly levodopa-responsive parkinsonism or cerebellar
initial neurological abnormality in an individual who dysfunction
later developed MSA (Wu et al., 2004). Pain has been III. Definite MSA
Pathologically confirmed by the presence of a high density
reported to be the initial clinical manifestation of MSA
of glial cytoplasmic inclusions in association with a
in 6% of patients (Pezzoli et al., 2004). Anisocoria was combination of degenerative changes in the nigrostriatal
present in 8% of the 203 cases of MSA described by and olivopontocerebellar pathways
Wenning and colleagues (1997); in 5%, it was asso-
ciated with Horners syndrome. Dizziness or vertigo Adapted from Gilman et al. (1999) with permission.
MULTIPLE SYSTEM ATROPHY 315
Table 46.2 Table 46.3
Clinical domains, features and criteria used in the Exclusion criteria for the diagnosis of multiple system
diagnosis of multiple system atrophy atrophy

Domain I: Autonomic and urinary dysfunction I. History


A. Features: A. Symptomatic onset under 30 years of age
1. Orthostatic hypotension (20 mmHg systolic or 10 mmHg B. Family history of a similar disorder
diastolic) C. Systemic diseases or other identifiable causes for
2. Urinary incontinence or incomplete bladder emptying features listed in Table 2
B. Criterion: D. Hallucinations unrelated to medication
Orthostatic fall in blood pressure (30 mmHg systolic or 15 II. Physical examination
mmHg diastolic) A. DSM criteria for dementia
or B. Prominent slowing of vertical saccades or vertical
Urinary incontinence (persistent, involuntary partial supranuclear gaze palsy
or total bladder emptying, accompanied by erectile C. Evidence of focal cortical dysfunction: aphasia,
dysfunction in men) alien-limb syndrome, parietal dysfunction
or III. Laboratory investigation
Both A. Metabolic, molecular genetic and imaging evidence
Domain II: Parkinsonism of an alternative cause of features listed in Table 46.2
A. Features:
1. Bradykinesia (slowness of voluntary movement with Adapted from Gilman et al. (1999) with permission.
progressive reduction in speed and amplitude during DSM, Diagnostic and Statistical Manual of Mental Disorders.
repetitive actions)
2. Rigidity 46.6.2. Microscopic pathology
3. Postural instability (not caused by primary visual,
vestibular, cerebellar or proprioceptive dysfunction)
On a microscopic level, MSA is characterized by neu-
4. Tremor (postural, resting or both)
B. Criterion:
ronal loss and gliosis involving multiple areas of the
Bradykinesia plus at least one of items 2 through 4 brain, brainstem and spinal cord. The most prominent
Domain III: Cerebellar dysfunction neurodegenerative changes occur in putamen, globus
A. Features: pallidus, substantia nigra, locus ceruleus, cerebellar
1. Gait ataxia (wide-based stance with steps of irregular cortex, inferior olive, pontine nuclei, dorsal motor
length and direction) nucleus of the vagus and in the intermediolateral col-
2. Ataxic dysarthria umns and Onufs nucleus in the spinal cord (Lantos
3. Limb ataxia and Papp, 1994; Wenning et al., 1997; Burn and Jaros,
4. Sustained gaze-evoked nystagmus 2001; Gilman, 2002). Neuronal loss may also
B. Criterion: involve catecholaminergic neurons in the ventrolateral
Gait ataxia plus at least one of items 2 through 4
medulla (Benarroch et al., 1998), neurons in the
Domain IV: Corticospinal tract dysfunction
A. Features:
EdingerWestphal nucleus and in the hypothalamus
1. Extensor plantar responses with hyperreflexia (Shy and Drager, 1960). White-matter pathology, with
B. Criterion: myelin degeneration, is also present in MSA (Matsuo
No corticospinal features are used et al., 1998).
As noted earlier, the discovery by Papp et al.,
Adapted from Gilman et al. (1999) with permission. (1989) of argyrophilic inclusions within the cytoplasm
of oligodendroglia in the brains of MSA patients has
provided the pathological hallmark of MSA
The putamen may be shrunken and display grayish (Fig. 46.1). In fact, MSA is the only neurodegenerative
discoloration due to iron deposition, whereas involve- disease in which oligodendroglial pathology is predo-
ment of the caudate and globus pallidus is less notice- minant (Jellinger et al., 2005). These inclusions, now
able (Dickson et al., 1999b; Colosimo et al., 2005a; labeled GCI, are composed of loosely aggregated,
Jellinger et al., 2005). Loss of pigmentation of the sub- multilayered, tubular filaments that are immunoreac-
stantia nigra pars compacta and locus ceruleus is also tive for ubiquitin and a-synuclein, along with a num-
characteristic. Cerebellar atrophy, along with atrophy ber of additional cytoskeletal proteins (Wakabayashi
of pons, inferior olives and the inferior and middle et al., 1998; Dickson et al., 1999a; Gai et al., 1999,
cerebellar peduncles may also be present (Burn and 2003; Burn and Jaros, 2001). a-Synuclein is normally
Jaros, 2001; Jellinger et al., 2005). expressed in only low levels in oligodendrocytes and
316 R. F. PFEIFFER
of the diagnosis. A band-like rim of hyperintense sig-
nal may be present at the lateral putamenal border on
T2-weighted images. Konagaya and colleagues
(1994) identified this abnormality in 17 of 28 patients
with clinically diagnosed MSA. Some investigators
have suggested that the hyperintensity is due to reac-
tive microgliosis or iron deposition (Schwarz et al.,
1996), whereas others have attributed it to widened
intertissue space produced by putamenal shrinkage
(Konagaya et al., 1998). Hypointensity within the
putamen itself on T2-weighted images may also be
seen in MSA. Hypointensity of the dorsolateral puta-
men coupled with hyperintensity of the putamenal
rim on T2-weighted images is highly specific for
MSA, but its absence does not exclude the diagnosis
Fig. 46.1. Glial cytoplasmic inclusions. a-Synuclein staining.
(Schrag et al., 1998; Kraft et al., 1999). Increased signal
Courtesy of Dr. Dennis Dickson, Mayo Clinic-Jacksonville.
involving the pyramidal tracts from the cortex down
to the cerebral peduncles has recently been described
it is uncertain whether in MSA there is impaired abil- on T2-weighted fluid-attenuated inversion recovery
ity to degrade a-synuclein or whether increased (FLAIR) images, but is not specific for MSA (Limberg
amounts are being formed (Wenning et al., 2003a). et al., 2005). MRI abnormalities within the posterior
Pountney and colleagues (2005a) have recently identi- fossa may also be evident in MSA. Atrophy of the cer-
fied aB-crystallin, a chaperone protein that binds to ebellar vermis, pons, middle cerebellar peduncles and
unfolded proteins and inhibits aggregation, in GCIs the cerebellar hemispheres is often present, primarily
in MSA and have suggested that it may play an impor- in patients presenting with predominantly cerebellar
tant role in the development of the GCIs. They have clinical findings (MSA-C). Degeneration of ponto-
also demonstrated the presence of SUMO-1, a small cerebellar fibers in the pons and middle cerebellar ped-
ubiquitin-like modifier protein, within 1030% of uncles sometimes produces hyperintensity within the
GCIs and speculate that SUMO-1 may be playing a pons on T2-weighted images in a cross-like pattern that
role in the formation of GCIs and reflect dysfunc- has been labeled the hot-cross bun sign (Schrag et al.,
tion of the proteasomal machinery (Pountney et al., 1998). More sophisticated MRI techniques, such as dif-
2005b). GCIs primarily reside in what has been termed fusion-weighted imaging, proton magnetic resonance
a system-bound distribution within the suprasegmen- spectroscopy, magnetization transfer imaging and mag-
tal motor systems (primary and supplementary motor netic resonance volumetry may prove to be of value in
cortex, basal ganglia, corticocerebellar system) and the diagnosis of MSA, but are not used in routine
supraspinal autonomic systems and their targets (Papp clinical care at this time (Seppi et al., 2005).
and Lantos, 1994; Burn and Jaros, 2001). In addition
to GCIs, oligodendroglial nuclear inclusions have been
identified in MSA, as have neuronal cytoplasmic and 46.7.2. Functional neuroimaging
nuclear inclusions (Papp and Lantos, 1992).
Functional neuroimaging procedures, such as positron
46.7. Diagnostic evaluation emission tomography (PET) and SPECT, have been
extensively evaluated for potential utility in the diag-
Although no single diagnostic study is capable of pro- nosis of MSA. However, neither ligands that are presy-
viding incontrovertible proof of the diagnosis of MSA, naptic dopaminergic markers nor dopamine receptor
useful information that may help to include or exclude ligands can consistently and reliably differentiate
the diagnosis can sometimes be derived from a variety MSA from other parkinsonian disorders in individual
of examinations. patients (Quinn, 2005). PET imaging with 18F-de-
oxyglucose may do somewhat better, especially if
46.7.1. Structural neuroimaging computer-assisted interpretation using statistical para-
metric mapping is employed (Eckert et al., 2005;
Although MRI has uncovered no features that are Juh et al., 2005), but is also still not fully adequate
pathognomonic for MSA, several abnormalities have (Quinn, 2005). Moreover, since neither PET nor
been identified that, if present, increase the probability SPECT imaging with these ligands is widely available,
MULTIPLE SYSTEM ATROPHY 317
use of functional imaging in the diagnosis of MSA is heart rate response to deep breathing and the Valsalva
currently confined to the research setting. maneuver, are also abnormal in MSA (Khurana,
Multiple studies employing either 6-[18F]fluorodo- 1994).
pamine PET or 123I-metaiodobenzylguanidine (MIBG) Assessment of bladder function, with measurement
scintigraphy to image the sympathetic innervation of of postvoid residual urine volume, can be performed
the heart have demonstrated marked denervation in by either bladder sonography or catheterization. Cathe-
Parkinsons disease, in contrast to intact innervation terization is simple and requires no special equipment,
in MSA (Goldstein, 2005; Mathias, 2005). A but sonography is non-invasive and can even be per-
meta-analysis of studies involving 245 patients with formed at bedside (Hahn and Ebersbach, 2005).
Parkinsons disease and 45 with MSA who underwent Videourodynamic testing is a more elegant procedure
cardiac MIBG scintigraphy demonstrated a specificity that provides a more expansive picture of bladder
of 94.6% in discriminating between the two conditions function (Sakakibara et al., 2001). Erectile function is
(Braune, 2001). The commercial availability of most often evaluated simply by questionnaire; specia-
MIBG and scintigraphy may encourage more extensive lized objective testing, such as nocturnal penile tumes-
use of this technique in the future in differentiating cence and rigidity assessment, is not usually necessary
MSA from Parkinsons disease. (Montorsi, 2005).
A variety of tests is available for assessment of GI
46.7.3. Autonomic function tests dysfunction in MSA. The modified barium swallow
test is widely employed for evaluation of dysphagia.
Autonomic function testing can be valuable both in the Gastric emptying time can be measured by sciniti-
diagnosis of MSA and in the formulation of treatment graphic means (Thomaides et al., 2005). In eva-
plans. Virtually all aspects of autonomic function are luating bowel function, colon transit time is most
amenable to testing that can illuminate both the char- often assessed by the repetitive ingestion method
acter and severity of any autonomic deficit. Tests of (Sakakibara et al., 2004a), and anorectal manometry
cardiovascular and urinary function are most fre- can be used to examine anorectal function (Stocchi
quently employed in evaluation of the MSA patient, et al., 2000).
but examination of GI and thermoregulatory function Tests of thermoregulatory function that have been
is also available and can be very useful. The mere pre- employed in MSA include the thermoregulatory sweat
sence of abnormalities on autonomic function testing, test and the quantitative sudomotor axon reflex test
however, does not clinch a diagnosis of MSA; similar, (Kihara et al., 1991). However, these tests are not
though typically less severe, abnormalities can also always readily available and have not been extensively
be present in other conditions, especially Parkinsons employed in patients with MSA.
disease.
Tilt-table testing, in which heart rate and blood 46.7.4. Neurophysiological testing
pressure are continuously monitored, is a sensitive
and non-invasive method by which the presence of The ability of external anal and urethral sphincter elec-
orthostatic hypotension can be documented. Ortho- tromyography (EMG) to distinguish MSA from
static hypotension has been defined as a drop of 20 Parkinsons disease and other parkinsonian disorders
mmHg or greater in systolic pressure or a drop of 10 has been the subject of considerable controversy.
mmHg or greater in diastolic pressure within 3 min- Electromyographic evidence of denervation, due to
utes of standing or head-up tilt (Consensus statement anterior horn cell loss in the nucleus of Onufrowicz
on the definition of orthostatic hypotension, pure auto- (Onufs nucleus) in the sacral spinal cord, is present
nomic failure and multiple system atrophy, 1996; in over 80% of individuals with MSA (Wenning
Goldstein et al., 2002). An array of additional tests of et al., 2004). However, similar abnormalities may be
cardiovascular function is available and may be part present in persons with other parkinsonian disorders,
of the testing battery in the autonomic function labora- such as Parkinsons disease and progressive supranuc-
tory. Plasma norepinephrine levels are often normal or lear palsy (Valldeoriola et al., 1995; Colosimo et al.,
near normal when measured in the supine position in 2000). In light of this, the idea has emerged that a
persons with MSA, but levels do not rise appropriately normal sphincter EMG renders a diagnosis of MSA
with standing (Mathias, 2005). Reversal of the circa- very unlikely, especially if disease symptoms have
dian change in blood pressure, assessed by 24-hour been present for more than 5 years, whereas an abnor-
blood pressure and heart rate monitoring, may also mal EMG cannot be considered as confirmation of a
be present in MSA (Mathias, 2005). Tests assessing diagnosis of MSA (Nahm and Freeman, 2003; Paviour
parasympathetic cardiovascular function, such as the et al., 2005).
318 R. F. PFEIFFER
A number of other neurophysiologic studies may controlled trials of targeted treatment modalities. Thus,
demonstrate abnormalities in patients with MSA. the available evidence of efficacy for various agents is
Various varieties of evoked potentials somatosen- largely empiric.
sory, visual and auditory may be abnormal in a min-
ority of persons (Abele et al., 2000b), but these are not 46.8.1. Motor features
of any genuine diagnostic utility. Evidence of periph-
eral neuropathy on nerve conduction velocity testing The parkinsonian features of MSA are treated with the
has been noted in 24% of 42 subjects with MSA same medications that are invoked in the treatment of
(Abele et al., 2000a), but this is also a non-specific Parkinsons disease. A clinically meaningful, though
finding that has no diagnostic usefulness. typically suboptimal, response to levodopa is evident
in 3365% of patients with MSA (Rajput et al.,
46.7.5. Other examinations 1990; Hughes et al., 1992b; Parati et al., 1993; Wen-
ning et al., 1994; Colosimo and Pezzella, 2002; Colo-
Kimber and colleagues (1997) reported that patients with simo et al., 2005b; Gilman et al., 2005). Individuals
MSA do not display a rise in serum growth hormone in with MSA may require larger levodopa doses than
response to administration of the a2-adrenoreceptor individuals with Parkinsons disease to generate a
agonist clonidine, whereas individuals with Parkinsons recognizable response. Therefore, a therapeutic trial
disease do. However, other investigators have found that of levodopa should not be abandoned until a levodopa
the test does not reliably distinguish the two disease dose of at least 1000 mg/day is reached (Gilman et al.,
processes (Tranchant et al., 2000; Pellecchia et al., 1998, 1999), although adverse effects such as ortho-
2001; Strijks et al., 2002). static hypotension may prohibit titration to such levels.
The use of brain parenchyma sonography (transcra- Wenning and colleagues (2005a) recently reported that
nial ultrasound) to differentiate Parkinsons disease 41% of 337 patients with MSA experienced a benefi-
from MSA and other atypical parkinsonian disorders cial response to levodopa at a mean daily dose of
has also been advocated (Benecke, 2002; Walter 686 mg, with the response lasting for an average of 4
et al., 2003). In preliminary studies, hyperechogenicity years. Although the initial improvement diminishes
of the substantia nigra was present in 96% of in- with the passage of time in most individuals, Hughes
dividuals with Parkinsons disease but in only 9% of and colleagues (1992b) reported that over 50% of the
patients with atypical parkinsonian syndromes, MSA patients they studied who initially experienced
whereas lentiform nucleus hyperechogenicity was improvement with levodopa remained partially respon-
evident in 77% of persons with atypical parkinsonism sive until death. Levodopa-induced dyskinesia devel-
and in only 23% of those with Parkinsons disease ops in approximately 50% of responders and, in
(Walter et al., 2003). Investigations that are more contrast to Parkinsons disease, is often dystonic in
recent have reinforced these findings (Behnke et al., character, preferentially involving the face and neck
2005). Although the non-invasive nature of sono- (Boesch et al., 2002).
graphy is appealing, the practical utility of this proce- The experience with other antiparkinsonian agents
dure in individual patients, especially those with has been more discouraging. Although an occasional
suspected MSA, remains to be determined. patient will derive some benefit from a dopamine
agonist, most do not and tolerance of these agents is
46.8. Treatment generally poor. Conflicting reports regarding the effec-
tiveness of amantadine have been published. Rajput
There has been a general perception that MSA is and colleagues (1998) noted improvement on amanta-
rapidly and relentlessly progressive and completely dine in over 60% of the 13 MSA patients they studied,
refractory to any treatment measures. Although it is but in a recent double-blind, placebo-controlled,
certainly correct that no treatment modality has cross-over trial involving 8 subjects, no statistically
demonstrated an ability to alter the progression of the significant improvement with amantadine could be
underlying disease process itself, it is incorrect to con- documented (Wenning, 2005). However, the investiga-
clude that there are no useful symptomatic treatment tors did note a trend toward improvement with aman-
measures that may be marshaled in the management tadine and commented that the small sample size
of MSA. In fact, it is often possible to ameliorate the may have obscured mild amantadine-related benefit.
non-motor features of MSA and even the motor fea- Anticholinergic drugs are generally ineffective in
tures may at least transiently demonstrate some alleviating parkinsonism in individuals with MSA,
response to treatment. However, the relative rarity of but an occasional patient may benefit (Polinsky,
MSA has rendered it exceedingly difficult to mount 1984; Wenning et al., 2005). Botulinum toxin A
MULTIPLE SYSTEM ATROPHY 319
injections may be effective in reducing blepharospasm solifenacin, darifenacin) with a better therapeutic
and limb dystonia, but some investigators caution index holds the promise of reduced toxicity. For indi-
against using it to treat cervical dystonia or antecollis viduals who cannot tolerate anticholinergic drugs,
because of the risk of inducing severe dysphagia (Tho- other treatment approaches are available. Gabapentin
bois et al., 2001; Wenning et al., 2005). Stereotactic and has been reported to improve symptoms of overactive
deep brain stimulation surgery has generally been consid- bladder (Kim et al., 2004), although it has not been
ered ineffective in the treatment of MSA. However, specifically tested in MSA. Desmopressin, admin-
investigators have described sustained improvement in istered at bedtime, can be used to reduce nocturia
4 individuals who underwent bilateral high-frequency (Dasgupta and Haslam, 1999; Fowler, 1999). For
stimulation of the subthalamic nuclei and recommended individuals with an underactive or hypotonic bladder,
a larger prospective study (Visser-Vandewalle et al., anticholinergic drugs are contraindicated. Instead,
2003). In contrast, the same procedure has been reported methods to promote bladder emptying are needed.
to aggravate dysarthria, dysphagia and gait impairment in Unfortunately, the treatment choices for this dysfunc-
MSA (Tarsy et al., 2003). tion are more limited. If an element of prostatic hyper-
There is no proven effective treatment for the trophy is present, a-blocking agents such as tamsulosin
cerebellar dysfunction that develops in MSA. are appropriate, but if the problem is strictly due to a
hypocontractile detrusor muscle, intermittent catheter-
46.8.2. Autonomic features ization is the best treatment option (Fowler, 1999).
For those individuals with detrusor-sphincter dys-
46.8.2.1. Genital dysfunction
synergia, anticholinergic drugs will diminish detrusor
Although erectile dysfunction in MSA responds to overactivity but intermittent catheterization may be
cyclic guanosine monophosphate phosphodiesterase necessary to deal with the failure of the urethral
inhibitors such as sildenafil and its relatives, these sphincter to relax. Botulinum toxin A injections
drugs have also demonstrated a predilection to produce into the urethral sphincter have also been used suc-
plummeting blood pressure with severe symptoms cessfully on an experimental basis in this regard,
of orthostatic hypotension, especially in men with though not in individuals with MSA (Smith et al.,
pre-existent orthostatic hypotension (Hussain et al., 2005).
2001). It thus seems wise to avoid these drugs in
men with MSA. Other treatment modalities for erectile
dysfunction are available. Intracavernosal injections of 46.8.2.3. Cardiovascular dysfunction
alprostadil are effective, but the necessity for and Management of orthostatic hypotension in persons
discomfort with the injections deters many from this with MSA can be very challenging. Both pharmacolo-
treatment modality. Priapism and the formation of gic and non-pharmacologic treatment approaches may
fibrotic nodules within the corpora are other potential be necessary. Non-pharmacologic measures are often
complications (Hatzichristou, 1999). Alprostadil overlooked by physicians, but are simple, inexpensive
can also be administered by intraurethral instillation. and often effective. Avoidance of high environmental
Vacuum devices, used in conjunction with constrictor temperature, large meals, alcohol, prolonged recum-
bands, are also effective, but patient acceptance of bency, straining during voiding or defecation, exces-
the devices is low. sive exercise and sudden changes in position can
reduce the risk of serious postural hypotension
46.8.2.2. Urinary dysfunction (Mathias, 2003, 2005). Sleeping with the head of
Appropriate treatment of bladder dysfunction depends the bed elevated will reduce the supine hypertension
on the specific type of impairment present. The that often occurs in MSA (Mathias, (2005)). The
approach to treatment of an overactive bladder is very mechanism for this nocturnal blood pressure increase
different from that toward an underactive or areflexic is not entirely clear, but may involve baroreflex
bladder. Because both types of bladder dysfunction abnormalities or shifting of fluid into the central vas-
may occur in MSA, use of appropriate urodynamic cular compartment, producing an effective increase in
and neurophysiologic testing is of immense value in blood volume (Goldstein et al., 2003; Mathias,
setting the treatment course. For detrusor overactivity 2005). Elevating the head of the bed may also dimin-
with urinary frequency and urgency, peripherally ish orthostatic hypotension upon getting out of bed in
acting anticholinergic drugs are the treatment of the morning, possibly by reducing nocturnal sodium
choice. Oxybutynin has been the standard-bearer for loss that occurs because of increased renal glomerular
many years, but the recent introduction of several filtration prompted by the effective increase in blood
newer, more selective anticholinergic drugs (trospium, volume (Pechere-Bertschi et al., 1998). Increased salt
320 R. F. PFEIFFER
intake can also diminish orthostatic symptoms by be helpful if patients are experiencing dysphagia with
increasing intravascular volume. The most effective aspiration, but percutaneous endoscopic gastrostomy
non-pharmacologic measure for reducing orthostatic tube placement is sometimes necessary. Domperidone,
symptoms, however, may be the use of individually where available, is effective in accelerating gastric
measured and fitted waist-high support stockings. emptying in persons with Parkinsons disease and,
Unfortunately, the stockings are difficult to don and presumably, would also do so in the setting of MSA,
very uncomfortable to wear, thus patients often resist although no specific information is available. Tega-
wearing them. An abdominal binder also reduces serod has been reported to improve gastric emptying
venous pooling and is often better tolerated than the in normal volunteers and in critically ill patients in
stockings (Mathias, 2003). The initial step in medical intensive care units (Degen et al., 2001; Banh et al.,
management of orthostatic hypotension is usually 2005). Whether it might do so in individuals with
administration of the mineralocorticoid fludrocortisone MSA or Parkinsons disease is unknown. Management
(Mathias, 2005; Wenning et al., 2005). If this is of constipation due to slowed colonic transit in MSA
insufficiently effective, midodrine can be added to can be patterned after that practiced for Parkinsons
the treatment regimen. In a double-blind study of 171 disease (Pfeiffer, 2005a, b). Increased fiber and fluid
patients (40 with MSA), Low and colleagues (1997) should be the first step, along with a stool softener if
reported that midodrine, an a-adrenergic agonist, was necessary. If that does not suffice, lactulose can be
both effective and safe in reducing symptomatic ortho- added to the treatment regimen. The next rung up the
static hypotension. The authors cautioned that mido- treatment ladder is the use of polyethylene glycol elec-
drine should be avoided after 6 p.m. in order to reduce trolyte balanced solutions, which have specifically
the risk of inducing nocturnal supine hypertension. been studied in patients with MSA and found to be
Other drugs that may be directly helpful in alleviating effective (Eichhorn and Oertel, 2001). Finally, enemas
orthostatic hypotension include ephedrine, indometa- are available, if needed. The value of prokinetic agents
cin, yohimbine and L-threo-dihidroxy-phenylserine. in treating colonic dysmotility is uncertain. The other
Octreotide, a somatostatin analog, ameliorates post- component of bowel dysfunction in MSA, defecatory
prandial hypotension by inhibiting release of vasodila- dysfunction due to disturbed function of the anorectal
tory GI peptides (Mathias, 2003). Desmopressin can musculature, presents a much more difficult manage-
diminish orthostatic hypotension in the morning by ment problem. In patients with Parkinsons disease,
reducing nocturnal diuresis (Mathias, 2003). Increas- subcutaneous apomorphine injections and botulinum
ing red cell mass by means of erythropoietin administra- toxin A injections into the external anal sphincter have
tion has also been reported to improve orthostatic been employed with benefit (Mathers et al., 1989;
hypotension (Winkler et al., 2002). Edwards et al., 1993), but there is no experience with
these treatment modalities in MSA.
46.8.2.4. Gastrointestinal dysfunction 46.9. Conclusion
Very little has been written specifically to address
treatment of GI dysfunction in MSA. However, mea- In the almost four decades since Graham and Oppen-
sures that have been successfully employed in the heimer first recognized MSA as a distinct disease pro-
management of GI problems in Parkinsons disease cess, awareness and knowledge concerning the clinical
(Pfeiffer, 2005a) are likely to be useful in MSA also. features and pathophysiology of MSA have soared.
Anticholinergic drugs have traditionally been used to However, progress in discovering effective treatment
alleviate drooling, but adverse effects are sometimes has languished behind the scientific advances. Strides
problematic. An approach that may limit the potential have been made, but there is still a tremendously long
for toxicity while still reducing saliva production is way to travel. Our blindness has been cured and we
the employment of one drop of 1% atropine ophthal- recognize the elephant. The daunting task that still
mic solution sublingually twice daily (Hyson et al., faces us is taming the beast.
2002). Salivary gland injections of both botulinum
References
toxin A and B have also been successfully employed
to reduce saliva production in persons with parkinson-
Abele M, Schulz JB, Burk K et al. (2000a). Nerve conduc-
ism, including some individuals with MSA (Mancini
tion studies in multiple system atrophy. Eur Neurol 43:
et al., 2003; Racette et al., 2003). Although anti- 221223.
parkinson medications sometimes improve dysphagia Abele M, Schulz JB, Burk K et al. (2000b). Evoked poten-
in Parkinsons disease, no similar information exists tials in multiple system atrophy (MSA). Acta Neurol
regarding MSA. Swallowing therapy techniques may Scand 101: 111115.
MULTIPLE SYSTEM ATROPHY 321

Adams RD, Salam-Adams M (1986). Striatonigral degeneration. Bower JH (2000). Multiple system atrophy. In: CH Adler, JE
In: PJ Vinken, GW Bruyn, HL Klawans (Eds.), Extrapyr- Ahlskog (Eds.), Parkinsons Disease and Movement Disor-
amidal Disorders. Handbook of Clinical Neurology, Vol. 5 ders: Diagnosis and Treatment Guidelines for the Practi-
(49). Elsevier, Amsterdam, pp. 205212. cing Physician. Humana Press, Totowa, NJ, pp. 235242.
Adams RD, van Bogaert L, van der Eecken H (1961). Bower JH, Maraganore DM, McDonnell SK et al.
Degenerescence nigro-striees et cerebello-nigro-striee. (1997). Incidence of progressive supranuclear palsy and
Psychiatry Neurol 142: 219259 (Article in French). multiple system atrophy in Olmsted County, Minnesota,
Arndt M (1894). Zur Pathologie des Kleinhirns. Arch 19761990. Neurology 49: 12841288.
Psychiatr Nervenkr 26: 404429 (Article in German). Braune S (2001). The role of cardiac metaiodobenzylguani-
Askmark H, Eeg-Olofsson KE, Johansson A et al. dine uptake in the differential diagnosis of parkinsonian
(2001). Parkinsonism and neck extensor myopathy. A syndromes. Clin Auton Res 11: 351355.
new syndrome or coincidental findings? Arch Neurol 58: Burn DJ, Jaros E (2001). Multiple system atrophy: cellular
232237. and molecular pathology. Mol Pathol 54: 419426.
Bandmann O, Sweeney MG, Daniel SE et al. (1997). Multiple- Cairns NJ, Atkinson PF, Kovacs T et al. (1997). Apolipopro-
system atrophy is genetically distinct from identified inher- tein E e4 allele frequency in patients with multiple system
ited causes of spinocerebellar degeneration. Neurology 49: atrophy. Neurosci Lett 221: 161164.
15981604. Chio A, Magnani C, Schiffer D (1998). Prevalence of Par-
Banh HL, MacLean C, Topp T et al. (2005). The use of tega- kinsons disease in Northwestern Italy: comparison of tra-
serod in critically ill patients with impaired gastric moti- cer methodology and clinical ascertainment of cases. Mov
lity. Clin Pharmacol Ther 77: 583586. Disord 13: 400405.
Bannister R, Gibson W, Michaels L et al. (1981). Laryngeal Christine CW, Aminoff MJ (2004). Clinical differentiation
abductor paralysis in multiple system atrophy. A report on of parkinsonian syndromes: prognostic and therapeutic
three necropsied cases, with observations on the laryngeal relevance. Am J Med 117: 412419.
muscles and the nuclei ambigui. Brain 104: 351368. Chrysostome V, Tison F, Yekhlef F et al. (2004). Epidemiol-
Beck RO, Betts CD, Fowler CJ (1994). Genitourinary dys- ogy of multiple system atrophy: a prevalence and pilot risk
function in multiple system atrophy: clinical features and factor study in Aquitaine, France. Neuroepidemiology 23:
treatment in 62 cases. J Urol 151: 13361341. 201208.
Behnke S, Berg D, Naumann M et al. (2005). Differentiation Cohen J, Low P, Fealey R et al. (1987). Somatic and auto-
of Parkinsons disease and atypical parkinsonian syn- nomic function in progressive autonomic failure and
dromes by transcranial ultrasound. J Neurol Neurosurg multiple system atrophy. Ann Neurol 22: 692699.
Psychiatry 76: 423425. Colosimo C, Pezzella FR (2002). The symptomatic treatment
Benarroch EE, Smithson IL, Low PA et al. (1998). Depletion of multiple system atrophy. Eur J Neurol 9: 195199.
of catecholaminergic neurons of the rostral ventrolateral Colosimo C, Albanese A, Hughes AJ et al. (1995). Some
medulla in multiple systems atrophy with autonomic specific clinical features differentiate multiple system
failure. Ann Neurol 43: 156163. atrophy (striatonigral variety) from Parkinsons disease.
Benecke R (2002). Clinical features and laboratory findings Arch Neurol 52: 294298.
for differentiating parkinsonian syndromes. J Neurol Colosimo C, Inghilleri M, Chaudhuri KR (2000). Parkinsons
249 (Suppl 3): III/6III14. disease misdiagnosed as multiple system atrophy by
Benrud-Larson LM, Sandroni P, Schrag A et al. (2005). Depres- sphincter electro-myography. J Neurol 247: 559561.
sive symptoms and life satisfaction in patients with multiple Colosimo C, Geser F, Wenning GK (2005a). Clinical spec-
system atrophy. Mov Disord 20: 951957. trum and pathological features of multiple system atrophy.
Ben-Shlomo Y, Wenning GK, Tison F et al. (1997). Survival In: M LeDoux, (Ed.), Animal Models of Movement
of patients with pathologically proven multiple system Disorders. Elsevier, Amsterdam, pp. 541570.
atrophy: a meta-analysis. Neurology 48: 384393. Colosimo C, Tiple D, Wenning GK (2005b). Management of
Berciano J, Wenning GK (2005). The Lewis family revisited: multiple system atrophy: state of the art. J Neural Transm
no evidence for autosomal dominant multiple system atro- 112: 16951704.
phy. Parkinsonism Relat Disord 11: 363365. Combarros O, Infante J, Llorca J et al. (2003). Interleukin-
Boesch SM, Wenning GK, Ransmayr G et al. (2002). Dysto- 1A (-889) genetic polymorphism increases the risk of mul-
nia in multiple system atrophy. J Neurol Neurosurg Psy- tiple system atrophy. Mov Disord 18: 13851386.
chiatry 72: 300303. Consensus statement on the definition of orthostatic hypoten-
Boeve BF, Silber MH, Ferman TJ et al. (2001). Association sion, pure autonomic failure, and multiple system atrophy
of REM sleep behavior disorder and neurodegenerative (1996). J Neurol Sci 144: 218219.
disease may reflect an underlying synucleinopathy. Mov Dasgupta P, Haslam C (1999). Treatment of neurogenic
Disord 16: 622630. bladder dysfunction. In: CJ Fowler, (Ed.), Neurology of
Bonnet AM, Pichon J, Vidailhet M et al. (1997). Urinary dis- Bladder, Bowel, and Sexual Dysfunction. Butterworth
turbances in striatonigral degeneration and Parkinsons Heinemann, Boston, pp. 163183.
disease: clinical and urodynamic aspects. Mov Disord Degen L, Matzinger D, Merz M et al. (2001). Tegaserod, a 5-
12: 509513. HT4 receptor partial agonist, accelerates gastric emptying
322 R. F. PFEIFFER

and gastrointestinal transit in healthy male subjects. Gilman S, Low PA, Quinn N et al. (1999). Consensus
Aliment Pharmacol Ther 15: 17451751. statement on the diagnosis of multiple system atrophy.
Dejerine J, Thomas AA (1900). Latrophie olivo-ponto-cere- J Neurol Sci 163: 9498.
belleuse. Nouvelle Iconographie de la Salpetriere 13: Gilman S, May SJ, Shults CW et al. (2005). The North
330370 (Article in French). American Multiple System Atrophy Study Group.
Dickson DW, Liu WK, Hardy J et al. (1999a). Widespread J Neural Transm 112: 16871694.
alterations of asynuclein in multiple system atrophy. Am Goldstein DS (2005). Cardiovascular autonomic dysfunction.
J Pathol 155: 12411251. In: RF Pfeiffer, I BodisWollner (Eds.), Parkinsons Dis-
Dickson DW, Lin W, Liu WK et al. (1999b). Multiple system ease and Nonmotor Dysfunction. Humana Press, Totowa,
atrophy: a sporadic synucleinopathy. Brain Pathol 9: NJ, pp. 149157.
721732. Goldstein DS, Robertson D, Esler M et al. (2002). Dysauto-
Dujardin K, Defebvre L, Krystkowiak P et al. (2003). Exe- nomias: clinical disorders of the autonomic nervous
cutive function differences in multiple system atrophy system. Ann Intern Med 137: 753763.
and Parkinsons disease. Parkinsonism Relat Disord 9: Goldstein DS, Pechnik S, Holmes C et al. (2003). Association
205211. between supine hypertension and orthostatic hypotension
Eckert T, Barnes A, Dhawan V et al. (2005). FDG PET in in autonomic failure. Hypertension 42: 136142.
the differential diagnosis of parkinsonian disorders. Neu- Gouider-Khouja N, Vidailhet M, Bonner AM et al.
roimage 26: 912921. (1995). Pure striatonigral degeneration and Parkinsons
Edwards LL, Quigley EM, Harned RK et al. (1993). Defeca- disease: a comparative clinical study. Mov Disord 10:
tory function in Parkinsons disease: response to apomor- 288294.
phine. Ann Neurol 33: 490493. Graham JG, Oppenheimer DR (1969). Orthostatic hypoten-
Eichhorn TE, Oertel WH (2001). Macrogol 3350/electrolyte sion and nicotine sensitivity in a case of multiple system
improves constipation in Parkinsons disease and multiple atrophy. J Neurol Neurosurg Psychiatry 32: 2834.
system atrophy. Mov Disord 16: 11761177. Hahn K, Ebersbach G (2005). Sonographic assessment of
Fetoni V, Soliveri P, Monza D et al. (1999). Affective symp- urinary retention in multiple system atrophy and idiopathic
toms in multiple system atrophy and Parkinsons disease: Parkinsons disease. Mov Disord 20: 14991502.
response to levodopa therapy. J Neurol Neurosurg Psy- Hanna PA, Jankovic J, Kirkpatrick JB (1999). Multiple sys-
chiatry 66: 541544. tem atrophy: the putative causative role of environmental
Fowler CJ (1999). Neurological disorders of micturition and toxins. Arch Neurol 56: 9094.
their treatment. Brain 122: 12131231. Hatzichristou DG (1999). Treatment of sexual dysfunction
Frumkin H (1998). Multiple system atrophy following and infertility in patients with neurologic diseases. In:
chronic carbon disulfide exposure. Environ Health Per- CJ Fowler, (Ed.), Neurology of Bladder, Bowel, and Sex-
spect 106: 611613. ual Dysfunction. Butterworth Heinemann, Boston, pp.
Gai WP, Power JH, Blumbergs PC et al. (1998). Multiple 209225.
system atrophy: a new alpha-synuclein disease? Lancet Healy DG, Abou-Sleiman PM, Quinn N et al. (2005).
352: 547548. European MSA Study Group. UCHL-1 gene in multiple
Gai WP, Power JH, Blumbergs PC et al. (1999). Alpha-synu- system atrophy: a haplotype tagging approach. Mov Disord
clein immunoisolation of glial inclusions from multiple 20 (10): 13381343.
system atrophy brain tissue reveals multiprotein compo- Higo R, Tayama N, Watanabe T et al. (2003). Videofluoro-
nents. J Neurochem 73: 20932100. scopic and manometric evaluation of swallowing function
Gai WP, Pountney DL, Power JH et al. (2003). alpha-Synu- in patients with multiple system atrophy. Ann Otol Rhinol
clein fibrils constitute the central core of oligodendroglial Laryngol 112: 630636.
inclusion filaments in multiple system atrophy. Exp Neu- Hughes AJ, Ben-Shlomo Y, Daniel SE et al. (1992a). What
rol 181: 6878. features improve the accuracy of clinical diagnosis in Par-
Geser F, Wenning GK (2005). Multiple system atrophy. In: I kinsons disease: a clinicopathologic study. Neurology 42:
Litvan, (Ed.), Atypical Parkinsonian Disorders. Humana 11421146.
Press, Totowa, NJ, pp. 335360. Hughes AJ, Colosimo C, Kleedorfer B et al. (1992b). The
Ghorayeb I, Yekhlef F, Chrysostome V et al. (2002). Sleep dopaminergic response in multiple system atrophy. J Neu-
disorders and their determinants in multiple system atro- rol Neurosurg Psychiatry 55: 10091013.
phy. J Neurol Neurosurg Psychiatry 72: 798800. Hughes AJ, Daniel SE, BenShlomo Y et al. (2002). The
Gilman S (2002). Multiple system atrophy. In: JJ Jankovic, E accuracy of diagnosis of parkinsonian syndromes in a spe-
Tolosa (Eds.), Parkinsons Disease and Movement Disorders. cialist movement disorder service. Brain 125: 861870.
Lippincott Williams & Wilkins, Philadelphia, pp. 170184. Hussain IF, Brady CM, Swinn MJ et al. (2001). Treatment of
Gilman S, Low P, Quinn N et al. (1998). Consensus state- erectile dysfunction with sildenafil citrate (Viagra) in
ment on the diagnosis of multiple system atrophy. Ameri- parkinsonism due to Parkinsons disease or multiple sys-
can Autonomic Society and American Academy of tem atrophy with observations on orthostatic hypotension.
Neurology. Clin Auton Res 8: 359362. J Neurol Neurosurg Psychiatry 71: 371374.
MULTIPLE SYSTEM ATROPHY 323

Hyson HC, Johnson AM, Jog MS (2002). Sublingual Kraft E, Schwarz A, Trenkwalder C et al. (1999). The
atropine for sialorrhea secondary to parkinsonism: a pilot combination of hypointense and hyperintense signal
study. Mov Disord 17: 13181320. changes on T2-weighted magnetic resonance imaging
Infante J, Llorca J, Berciano J et al. (2005). Interleukin-8, sequences: a specific marker of multiple system atrophy?
intercellular adhesion molecule-1 and tumor necrosis Arch Neurol 56: 225228.
factor-a gene polymorphisms and the risk for multiple Kumazawa K, Sobue G, Nakao N et al. (1989). Postganglio-
system atrophy. J Neurol Sci 228: 1113. nic sudomotor function in multiple system atrophy. Rinsho
Iranzo A, Santamaria J, Tolosa E (2000). Continuous posi- Shinkeigaku 29: 13571363 (Article in Japanese).
tive air pressure eliminates nocturnal stridor in multiple Kurisaki H (1999). Prognosis of multiple system atrophy
system atrophy. Barcelona Multiple System Atrophy survival time with or without tracheostomy. Rinsho
Study Group. Lancet 356: 13291330. Shinkeigaku 39: 503507 (Article in Japanese).
Iranzo A, Santamaria J, Tolosa E et al. (2004). Long-term Lange KW, Tucha O, Alders GL et al. (2003). Differentiation
effect of CPAP in the treatment of nocturnal stridor in of parkinsonian syndromes according to differences in
multiple system atrophy. Neurology 63: 930932. executive functions. J Neural Transm 110: 983995.
Iranzo A, Santamaria J, Rye DB et al. (2005). Characteristics Lantos PL, Papp MI (1994). Cellular pathology of multiple
of idiopathic REM sleep behavior disorder and that asso- system atrophy: a review. J Neurol Neurosurg Psychiatry
ciated with MSA and PD. Neurology 65: 247252. 57: 129133.
Jellinger KA, Seppi K, Wenning GK (2005). Grading of neu- Lewis P (1964). Familial orthostatic hypotension. Brain 87:
ropathology in multiple system atrophy: proposal for a 719728.
novel scale. Mov Disord 20 (Suppl 12): S29S36. Limberg N, Jeavons S, Robertson T et al. (2005). Pyramidal
Johnsen JA, Miller VT (1986). Tobacco intolerance in multi- tract imaging in multiple-system atrophy. Mov Disord 20:
ple system atrophy. Neurology 36: 986988. 15271528.
Juh R, Pae CU, Lee CU et al. (2005). Voxel based com- Litvan I, Goetz CG, Jankovic J et al. (1997). What is the
parison of glucose metabolism in the differential diagnosis accuracy of the clinical diagnosis of multiple system
of the multiple system atrophy using statistical parametric atrophy? A clinicopathologic study. Arch Neurol 54:
mapping. Neurosci Res 52: 211219. 937944.
Kaufmann H, Biaggioni I (2003). Autonomic failure in neu- Low PA, Gilden JL, Freeman R et al. (1997), for the Mido-
rodegenerative disorders. Semin Neurol 23: 351363. drine Study Group. Efficacy of midodrine vs. placebo in
Khurana RK (1994). Cholinergic dysfunction in Shy-Drager neurogenic orthostatic hypotension: a randomized, dou-
syndrome: effect of the parasympathomimetic agent, ble-blind multicenter study. JAMA 277: 10461051.
bethanechol. Clin Auton Res 4: 513. Mabuchi N, Hirayama M, Koike Y et al. (2005). Progression
Kihara M, Sugenoya J, Takahashi A (1991). The assessment and prognosis in pure autonomic failure (PAF): compari-
of sudomotor dysfunction in multiple system atrophy. Clin son with multiple system atrophy. J Neurol Neurosurg
Auton Res 1: 297302. Psychiatry 76: 947952.
Kim HS, Lee MS (2003). Frequencies of single nucleotide
Mancini F, Zangaglia R, Cristina S et al. (2003). Double-
polymorphism in alcohol dehydrogenase7 gene in patients
blind, placebo-controlled study to evaluate the efficacy
with multiple system atrophy and controls. Mov Disord
and safety of botulinum toxin type A in the treatment of
18: 10651067.
drooling in parkinsonism. Mov Disord 18: 685688.
Kim YT, Kwon DD, Kim J et al. (2004). Gabapentin for
Mathers SE, Kempster PA, Law PJ et al. (1989). Anal
overactive bladder and nocturia after anticholinergic fail-
sphincter dysfunction in Parkinsons disease. Arch Neurol
ure. Int Braz J Urol 30: 275278.
46: 10611064.
Kimber JR, Watson L, Mathias CJ (1997). Distinction of
idiopathic Parkinsons disease from multiple-system atro- Mathias CJ (2003). Autonomic diseases: management.
phy by stimulation of growth-hormone release with cloni- J Neurol Neurosurg Psychiatry 74 (Suppl 3): iii42iii47.
dine. Lancet 349: 18771881. Mathias CJ (2005). Cardiovascular autonomic dysfunction
Kirchhof K, Apostolidis AN, Mathias CJ et al. (2003). Erec- in Parkinsons disease and parkinsonian syndromes. In:
tile and urinary dysfunction may be the presenting features M Ebadi, RF Pfeiffer (Eds.), Parkinsons Disease. CRC
in patients with multiple system atrophy: a retrospective Press, Boca Raton, pp. 295317.
study. Int J Impot Res 15: 293298. Mathias CJ, Mallipeddi R, Bleasdale-Barr K (1999). Symp-
Klein C, Brown R, Wenning G et al. (1997). The cold hands toms associated with orthostatic hypotension in pure auto-
sign in multiple system atrophy. Mov Disord 12: 514518. nomic failure and multiple system atrophy. J Neurol 246:
Konagaya M, Konagaya Y, Iida M (1994). Clinical and 893898.
magnetic resonance imaging study of extrapyramidal Matsuo A, Akiguchi I, Lee GC et al. (1998). Myelin degen-
symptoms in multiple system atrophy. J Neurol Neurosurg eration in multiple system atrophy detected by unique
Psychiatry 57: 15281531. antibodies. Am J Pathol 153: 735744.
Konagaya M, Sakai M, Matsuoka Y et al. (1998). Patho-MR Merlo IM, Occhini A, Pacchetti C et al. (2002). Not paraly-
imaging study in the putaminal margin in multiple system sis, but dystonia causes stridor in multiple system atrophy.
atrophy. No To Shinkei 50: 383385 (Article in Japanese). Neurology 58: 649652.
324 R. F. PFEIFFER

Montorsi F (2005). Assessment, diagnosis, and investigation Pfeiffer RF (2005b). Intestinal dysfunction. In: RF Pfeiffer, I
of erectile dysfunction. Clin Cornerstone 7: 2935. Bodis-Wollner (Eds.), Parkinsons Disease and Nonmotor
Morris HR, Vaughan JR, Datta SR et al. (2000). Multiple sys- Dysfunction. Humana Press, Totowa, NJ, pp. 115125.
tem atrophy/progressive supranuclear palsy: a-synuclein, Pierangeli G, Provini F, Maltoni P et al. (2001). Nocturnal
synphilin, tau, and APOE. Neurology 55: 19181920. body core temperature falls in Parkinsons disease but
Muller J, Wenning GK, Verny M et al. (2001). Progression not in multiple-system atrophy. Mov Disord 16: 226232.
of dysarthria and dysphagia in postmortem-confirmed Plante-Bordeneuve V, Bandmann O, Wenning G et al.
parkinsonian disorders. Arch Neurol 58: 259264. (1995). CYP2D6-debrisoquine hydroxylase gene poly-
Nahm F, Freeman R (2003). Sphincter electromyography morphism in multiple system atrophy. Mov Disord 10:
and multiple system atrophy. Muscle Nerve 28: 1826. 277278.
Nee LE, Gomez MR, Dambrosia J et al. (1991). Environmen- Plazzi G, Corsini R, Provini F et al. (1997). REM sleep beha-
tal-occupational risk factors and familial associations in vior disorders in multiple system atrophy. Neurology 48:
multiple system atrophy: a preliminary investigation. Clin 10941097.
Auton Res 1: 913. Polinsky RJ (1984). Multiple system atrophy. Clinical
Nee LE, Scott J, Polinsky RJ (1993). Olfactory dysfunction aspects, pathophysiology, and treatment. Neurol Clin 2:
in the Shy-Drager syndrome. Clin Auton Res 3: 281282. 487498.
Nicholl DJ, Bennett P, Hiller L et al. (1999), for the Eur- Pramstaller PP, Wenning GK, Smith SJ et al. (1995). Nerve
opean Study Group on Atypical Parkinsonism. A study conduction studies, skeletal muscle EMG, and sphincter
of five candidate genes in Parkinsons disease and related EMG in multiple system atrophy. J Neurol Neurosurg Psy-
neurodegenerative disorders. Neurology 53: 14151421. chiatry 58: 618621.
Oertel WH, Wachter T, Quinn NP et al. (2003). Redu- Pountney DL, Treweek TM, Chataway T et al. (2005a).
ced genital sensitivity in female patients with multiple sys- Alpha B-crystallin is a major component of glial cytoplas-
tem atrophy of parkinsonian type. Mov Disord 18: 430432. mic inclusions in multiple system atrophy. Neurotox Res
Papp MI, Lantos PL (1992). Accumulation of tubular struc- 7: 7785.
tures in oligodendroglial and neuronal cells as the basic Pountney DL, Chegini F, Shen X et al. (2005b). SUMO-1
alteration in multiple system atrophy. J Neurol Sci 107: marks subdomains within glial cytoplasmic inclusions of
172182. multiple system atrophy. Neurosci Lett 381: 7479.
Papp MI, Lantos PL (1994). The distribution of oligodendro- Quinn N (1989a). Multiple system atrophythe nature of
glial inclusions in multiple system atrophy and its rele- the beast. J Neurol Neurosurg Psychiatry 52 (Suppl): 7889.
vance to clinical symptomatology. Brain 117: 235243. Quinn N (1989b). Disproportionate antecollis in multiple
Papp MI, Kahn JE, Lantos PL (1989). Glial cytoplasmic system atrophy. Lancet 1: 844.
inclusions in the CNS of patients with multiple system atro- Quinn NP (2005). How to diagnose multiple system atrophy.
phy (striatonigral degeneration, olivopontocerebellar atro- Mov Disord 20 (Suppl 12): S5S10.
phy and Shy-Drager syndrome). J Neurol Sci 94: 79100. Racette BA, Good L, Sagitto S et al. (2003). Botulinum toxin
Parati EA, Fetoni V, Geminiani GC et al. (1993). Response B reduces sialorrhea in parkinsonism. Mov Disord 18:
to L-DOPA in multiple system atrophy. Clin Neurophar- 10591061.
macol 16: 139144. Rajput AH, Rozdilsky B, Rajput A et al. (1990). Levodopa
Pathak A, Lapeyre-Mestre M, Montastruc JL et al. efficacy and pathological basis of Parkinson syndrome.
(2005). Heat-related morbidity in patients with orthostatic Clin Neuropharmacol 13: 553558.
hypotension and primary autonomic failure. Mov Disord Rajput AH, Uitti RJ, Fenton ME et al. (1998). Amantadine
20: 12131219. effectiveness in multiple system atrophy and progressive
Paviour DC, Williams D, Fowler CJ et al. (2005). Is sphinc- supranuclear palsy. Parkinsonism Relat Disord 3:
ter electromyography a helpful investigation in the diagno- 211214.
sis of multiple system atrophy? A retrospective study with Riley DE (2002). Dystonia in multiple system atrophy.
pathological diagnosis. Mov Disord 20: 14251430. J Neurol Neurosurg Psychiatry 72: 286.
Pellecchia MT, Salvatore E, Pivonello R et al. (2001). Stimu- Rivest J, Quinn N, Marsden CD (1990). Dystonia in Parkin-
lation of growth hormone release in multiple system atro- sons disease, multiple system atrophy, and progressive
phy, Parkinsons disease, and idiopathic cerebellar ataxia. supranuclear palsy. Neurology 40: 15711578.
Neurol Sci 22: 7980. Saito Y, Matsuoka Y, Takahashi A et al. (1994). Survival of
Pechere-Bertschi A, Nussberger J, Biollaz J et al. (1998). Cir- patients with multiple system atrophy. Intern Med 33:
cadian variations of renal sodium handling in patients with 321325.
orthostatic hypotension. Kidney Int 54: 12761282. Sakakibara R, Hattori T, Tojo M et al. (1993). Micturitional
Pezzoli G, Canesi M, Galli C (2004). An overview of parkin- disturbance in multiple system atrophy. Jpn J Psychiatry
sonian syndromes: data from the literature and from an Neurol 47: 591598.
italian data-base. Sleep Med 5: 181187. Sakakibara R, Hattori T, Uchiyama T et al. (2000). Urinary
Pfeiffer RF (2005a). Gastrointestinal dysfunction in Parkinsons dysfunction and orthostatic hypotension in multiple system
disease. In: M Ebadi, RF Pfeiffer (Eds.), Parkinsons Disease. atrophy: which is the more common and earlier manifesta-
CRC Press, Boca Raton, pp. 259273. tion? J Neurol Neurosurg Psychiatry 68: 6569.
MULTIPLE SYSTEM ATROPHY 325

Sakakibara R, Hattori T, Uchiyama T et al. (2001). Videouro- special emphasis on sleep talk as its early clinical manifes-
dynamic and sphincter motor unit potential analyses in tation. J Neurol Neurosurg Psychiatry 63: 678681.
Parkinsons disease and multiple system atrophy. J Neurol Tarsy D, Apetauerova D, Ryan P et al. (2003). Adverse
Neurosurg Psychiatry 71: 600606. effects of subthalamic nucleus DBS in a patient with mul-
Sakakibara R, Odaka T, Uchiyama T et al. (2004a). Colonic tiple system atrophy. Neurology 61: 247249.
transit time, sphincter EMG, and rectoanal videomanome- Testa D, Monza D, Ferrarini M et al. (2001). Comparison of
try in multiple system atrophy. Mov Disord 19: 924929. natural histories of progressive supranuclear palsy and
Sakakibara R, Hiruma K, Arai K et al. (2004b). Head-turning multiple system atrophy. Neurol Sci 22: 247251.
dizziness in multiple system atrophy. Parkinsonism Relat Thobois S, Broussole E, Toureille L et al. (2001). Severe dys-
Disord 10: 255256. phagia after botulinum toxin injection for cervical dystonia
Salazar G, Valls-Sole J, Marti MJ et al. (2000). Postural and in multiple system atrophy. Mov Disord 16: 764765.
action myoclonus in patients with parkinsonian type multi- Thomaides T, Bleasdale-Barr K, Chaudhuri KR et al. (1993).
ple system atrophy. Mov Disord 15: 7783. Cardiovascular and hormonal responses to liquid food chal-
Schatz IJ (2003). A remarkable patient40 years with prob- lenge in idiopathic Parkinsons disease, multiple system atro-
able multiple system atrophy (MSA). Clin Auton Res 13: phy, and pure autonomic failure. Neurology 43: 900904.
221223. Thomaides T, Karapanayiotides T, Zoukos Y et al. (2005).
Schultze V (1887). Uber einen Fall von Kleinhirnschwund mit Gastric emptying after semi-solid food in multiple system
Degenerationen im verlangerten Marke und im Rucken- atrophy and Parkinson disease. J Neurol 252: 10551059.
marke (wahrscheinlich in Folge von Alkoholismus). Virch- Tison F, Wenning GK, Quinn NP et al. (1995). REM sleep
ows Arch Pathol Anat 108: 331343 (Article in German). behaviour disorder as the presenting symptom of multiple
Schrag A, Kingsley D, Phatouros C et al. (1998). Clinical system atrophy. J Neurol Neurosurg Psychiatry 58: 379380.
usefulness of magnetic resonance imaging in multiple sys- Tison F, Yekhlef F, Chrysostome V et al. (2000). Prevalence
tem atrophy. J Neurol Neurosurg Psychiatry 65: 6571. of multiple system atrophy. Lancet 355: 495496.
Schrag A, Ben-Shlomo Y, Quinn NP (1999). Prevalence of Tison F, Yekhlef F, Chrysostome V et al. (2002). Parkinson-
progressive supranuclear palsy and multiple system atro- ism in multiple system atrophy: natural history, severity
phy: a cross-sectional study. Lancet 354: 17711775. (UPDRS-III), and disability assessment compared with
Schwarz J, Weis S, Kraft E et al. (1996). Signal changes on Parkinsons disease. Mov Disord 17: 701709.
MRI and increases in reactive microgliosis, astrogliosis, Tranchant C, Guiraud-Chaumeil C, EchanizLaguna A et al.
and iron in the putamen of two patients with multiple sys- (2000). Is clonidine growth hormone stimulation a good
tem atrophy. J Neurol Neurosurg Psychiatry 60: 98101. test to differentiate multiple system atrophy from Parkin-
Seppi K, Schocke MF, Wenning GK et al. (2005). How to sons disease? J Neurol 247: 853856.
diagnose MSA early: round table magnetic resonance ima- Trenkwalder C, Schwarz J, Gebhard J et al. (1995). Starnberg
ging. J Neural Transm 112: 16251634. trial on epidemiology of Parkinsonism and hypertension in
Shulman LM, Minagar A, Weiner WJ (2004). Multiple-sys- the elderly. Prevalence of Parkinsons disease and related
tem atrophy. In: RL Watts, WC Koller (Eds.), Movement disorders assessed by a door-to-door survey of inhabitants
Disorders: Neurologic Principles and Practice, second older than 65 years. Arch Neurol 52: 10171022.
edn., McGraw-Hill, New York, pp. 359369. Uzawa A, Sakakibara R, Tamura N et al. (2005). Laryngeal
Shy GM, Drager GA (1960). A neurological syndrome asso- abductor paralysis can be a solitary manifestation of multi-
ciated with orthostatic hypotension. Arch Neurol 2: 511527. ple system atrophy. J Neurol Neurosurg Psychiatry
Silber MH, Levine S (2000). Stridor and death in multiple 76 (12): 17391741.
system atrophy. Mov Disord 15: 699704. Valldeoriola F, Valls-Sole J, Tolosa ES et al. (1995). Striated
Simpson DM, Kaufmann H, Sanders I et al. (1992). Laryn- anal sphincter denervation in patients with progressive
geal dystonia in multiple system atrophy. Muscle Nerve supranuclear palsy. Mov Disord 10: 550555.
15: 12131215. van der Eecken H, Adams RD, van Bogaert L (1960). Strio-
Slawek J, Derejko M, Lass P et al. (2006). Camptocormia or pallidal-nigral degeneration: a hitherto undescribed lesion
Pisa syndrome in multiple system atrophy. Clin Neurol in paralysis agitans. J Neuropathol Exp Neurol 19: 159161.
Neurosurg 108: 699704. Vanacore N, Bonifati V, Fabbrini G et al. (2000). Smoking
Smith CP, Nishiguchi J, OLeary M et al. (2005). Single- habits in multiple system atrophy and progressive supra-
institution experience in 110 patients with botulinum toxin nuclear palsy. European Study Group on Atypical Parkin-
A injection into bladder or urethra. Urology 65: 3741. sonisms. Neurology 54: 114119.
Stocchi F, Badiali D, Vacca L et al. (2000). Anorectal func- Vanacore N, Bonifati V, Fabbrini G et al. for the ESGAP
tion in multiple system atrophy and Parkinsons disease. Consortium (2001). Epidemiology of multiple system
Mov Disord 15: 7176. atrophy. Neurol Sci 22: 9799.
Strijks E, vant Hof M, Sweep F et al. (2002). Stimulation of Vanacore N, Bonifati V, Fabbrini G et al. (2005), the ESGAP
growth-hormone release with clonidine does not distinguish Consortium. Casecontrol study of multiple system atro-
individual cases of idiopathic Parkinsons disease from those phy. Mov Disord 20: 158163.
with striatonigral degeneration. J Neurol 249: 12061210. Vetrugno R, Provini F, Cortelli P et al. (2004). Sleep
Tachibana N, Kimura K, Kitajima K et al. (1997). REM disorders in multiple system atrophy: a correlative video-
sleep motor dysfunction in multiple system atrophy: with polysomnographic study. Sleep Med 5: 2130.
326 R. F. PFEIFFER

Visser-Vandewalle V, Temel Y, Colle H et al. (2003). Bilat- parkinsonian syndromes: a clinicopathological study.
eral high-frequency stimulation of the subthalamic nucleus J Neurol Neurosurg Psychiatry 67: 620623.
in patients with multiple system atrophyparkinsonism. Wenning GK, Geser F, StampferKountchev M et al. (2003a).
Report of four cases. J Neurosurg 98: 882887. Multiple system atrophy: an update. Mov Disord 18 (Suppl
Wakabayashi K, Yoshimoto M, Tsuji S et al. (1998). Alpha- 6): S34S42.
synuclein immunoreactivity in glial cytoplasmic inclusions Wenning GK, Geser F, Poewe W (2003b). The risus sardo-
in multiple system atrophy. Neurosci Lett 249: 180182. nicus of multiple system atrophy. Mov Disord 18: 1211.
Walter U, Niehaus L, Probst T et al. (2003). Brain paren- Wenning GK, Colosimo C, Geser F et al. (2004). Multiple
chyma sonography discriminates Parkinsons disease and system atrophy. Lancet Neurol 3: 93103.
atypical parkinsonian syndromes. Neurology 60: 7477. Wenning GK, Geser F, Poewe W (2005). Therapeutic
Watanabe H, Saito Y, Terao S et al. (2002). Progression and strategies in multiple system atrophy. Mov Disord
prognosis in multiple system atrophy. An analysis of 230 20 (Suppl 12): S67S76.
Japanese patients. Brain 125: 10701083. Wermuth L, Joensen P, Bunger N et al. (1997). High preva-
Wenning GK (2005), on behalf of the Working Group on lence of Parkinsons disease in the Faroe Islands. Neurol-
Atypical Parkinsonism of the Austrian Parkinsons Society. ogy 49: 426432.
Placebo-controlled trial of amantadine in multiple system Wetter TC, Collado-Seidel V, Pollmacher T et al. (2000).
atrophy. Clin Neuropharmacol 28: 225227. Sleep and periodic leg movement patterns in drug-free
Wenning GK, Ben-Shlomo Y, Magalhaes M et al. (1994). patients with Parkinsons disease and multiple system
Clinical features and natural history of multiple system atrophy. Sleep 23: 361367.
atrophy. An analysis of 100 cases. Brain 117: 835845. Winkler AS, Landau S, Watkins P et al. (2002). Observations
Wenning GK, Ben-Shlomo Y, Magalhaes M et al. (1995a). on haematological and cardiovascular effects of erythro-
Clinicopathological study of 35 cases of multiple system poietin treatment in multiple system atrophy with sympa-
atrophy. J Neurol Neurosurg Psychiatry 58: 160166. thetic failure. Clin Auton Res 12: 203206.
Wenning GK, Shephard B, Hawkes C et al. (1995b). Olfactory Wu YR, Chen CM, Ro LS et al. (2004). Sensory neuropathy
function in atypical parkinsonian syndromes. Acta Neurol as the initial manifestation of multiple system atrophy.
Scand 91: 247250. J Formos Med Assoc 103: 727730.
Wenning GK, Tison F, Ben Shlomo Y et al. (1997). Multiple Wullner U, Abele M, SchmitzHuebsch T et al. (2004). Prob-
system atrophy: a review of 203 pathologically proven able multiple system atrophy in a German family. J Neurol
cases. Mov Disord 12: 133147. Neurosurg Psychiatry 75: 924925.
Wenning GK, Scherfler C, Granata R et al. (1999). Time Yamaguchi M, Arai K, Asahina M et al. (2003). Laryngeal
course of symptomatic orthostatic hypotension and urinary stridor in multiple system atrophy. Eur Neurol 49:
incontinence in patients with postmortem confirmed 154159.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 47

Progressive supranuclear palsy

DAVID J. BURN1* AND ANDREW J. LEES2

1
Institute of Ageing and Health, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
2
Reta Lila Weston Institute of Neurological Studies, University College London, London, UK

47.1. Introduction . . . chilled, slow, earthy, fixed old man. A cada-


verous man of measured speech. An old man
In its full-blown form, the clinical picture of progres- who seemed as unable to wink, as if his eyelids
sive supranuclear palsy (PSP, or SteeleRichardson had been nailed to his forehead. An old man
Olszewski syndrome) is both arresting and highly whose eyes two spots of fire had no more
characteristic. The patient has a fixed Mona Lisa motion that [sic] if they had been connected with
stare, with a markedly reduced blink frequency (04 the back of his skull by screws driven through
blinks/minute). The head is retracted and the voice is it and riveted and bolted outside, among his
reduced to a distinctive slurred growl. The sufferer grey hair.
walks clumsily and unsteadily (like a drunken sailor), He had come in and shut the door and he now
with a notable tendency to topple backwards. Motor sat down. He did not bend himself to sit, as other
recklessness is a common early feature, leading to people do, but seemed to sink bolt upright, as if
the highly distinctive rocket sign on rising from a in water, until the chair stopped him.
chair. Clothes are soiled with spilled food, due an
inability to look down at the plate and difficulties
The late 19th century was a productive time for semi-
swallowing (the messy-tie sign). The time taken to
nal descriptions of neurological disorders. Charcot
respond to a question is prolonged, because of slow
encouraged the use of medical photography and draw-
cognitive processing (bradyphrenia).
ings to document the physical signs of the patients under
Recent clinical and molecular genetic studies suggest
his care and from two daguerreotypes published in 1889
that PSP should be considered a relatively common,
in the Nouvelle Iconographie de la Salpetrie`re by Dutil,
discrete neurodegenerative disorder with a number of
one of his interns, a case of PSP may have been
different clinical presentations but a distinctive neuro-
described as a hemiplegic paralysis agitans with unu-
pathological signature. This chapter will include im-
sual postures of the trunk and head (Goetz, 1996). Ret-
portant recent developments in PSP, including the
rocollis and an eye movement disorder were prominent
association of the disease with an H1 haplotype, an altered
components of the clinical picture. Charcots celebrated
tau isoform ratio, evidence for mitochondrial dysfunction
case history and sketches of Bachere, reported as a case
and oxidative stress in the pathogenesis and the discovery
of Parkinsons in extension, may be another missed
of a cluster of a PSP-like syndrome on Guadeloupe.
early example.
A recent review of 2000 studies on Parkinsons dis-
47.2. Historical perspective ease (PD) between 1861 and 1963 found 9 cases with
possible PSP in the pre-encephalitis lethargica (von
The intriguing suggestion has been made that Charles Economos disease) era (18611920) (Brusa et al.,
Dickens may have been first to describe a subject with 2004). The authors felt it could therefore be assumed
classical PSP in 1857 in his novel The Lazy Tour of that PSP is neither a new disease, nor a variant of
Two Idle Apprentices (Larner, 2002): postencephalitic parkinsonism (PEP).

*Correspondence to: Prof. David J. Burn, Regional Neurosciences Centre, Newcastle General Hospital, Westgate Road, Newcas-
tle upon Tyne NE4 6BE, UK. E-mail: d.j.burn@ncl.ac.uk, Tel: 44-(0)-191-256-3425, Fax: 44-(0)-191-256-3534.
328 D. J. BURN AND A. J. LEES
It is of note that, although motor neuron disease the likeliest explanation for the apparent variation. For
and PSP were described within 20 years of each instance, the incidence of 0.4 per 100 000 per year from
other, the former condition, with its accompanying the study of Mastaglia et al. is derived from 8 cases
distinctive pathology, was rapidly accepted by neu- seen in Perth, Western Australia over a 2-year period,
rologists as a discrete morbid entity, whereas a full population approximately one million. Their study
clinicopathological description of PSP had to wait was primarily clinical and electrophysiological, not
almost another century. Misdiagnosis of many cases epidemiological in nature (Mastaglia et al., 1973). In
of PSP as PEP or inappropriate lumping with other contrast, Bower and colleagues (1997) studied the inci-
atypical causes of parkinsonism such as arteriosclero- dence of PSP over a 14-year period in Olmsted County,
tic Parkinsons syndrome may be a partial explana- Minnesota. Sixteen incident cases were identified and
tion for this paradox. none had an age of onset before 50 years of age. The
In 1963, Dr. J. Clifford Richardson described 8 average annual incidence rate for ages 5099 years
patients with a common syndrome of ocular, motor was 5.3 per 100 000. This study was specifically desig-
and mental symptoms at the American Neurological ned to measure incidence and utilized the records link-
Association in Atlantic City. Richardson drew an ana- age system of the Rochester Epidemiology Project.
logy between this seemingly new condition and Von The authors suggested that increased awareness of
Economos disease and Asao Hirano, one of the dis- PSP by physicians could have contributed toward the
cussants, was struck by its similarity to a new disorder higher incidence figure.
being found amongst the indigenous Chamorros on the
Mariana Islands (lytico-bodig). 47.3.2. Prevalence

47.3. Epidemiology PSP comprises 26% of parkinsonian patients seen in


movement disorder clinics in Europe and the USA
47.3.1. Incidence
(Jackson et al., 1983; Katzenschlager et al., 2003).
Extrapolation of population prevalence from such data
Table 47.1 summarizes available incidence data for
is, however, misleading as atypical cases or patients
PSP (Mastaglia et al., 1973; Rajput et al., 1984;
responding poorly to treatment are more likely to be
Radhakrishnan et al., 1988; Bower et al., 1997). The
referred to these clinics. Similarly, the use of so-called
figure from the study of Golbe et al. (1988) was
necroepidemiological data may also be skewed towards
obtained indirectly, using prevalence and survival data.
more atypical parkinsonian cases, including PSP, as
The incidence of PSP ranges from 0.3 to 1.1 per
these patients are more likely to undergo postmortem
100 000 per year, with differences in study design being
(Maraganore et al., 1998).
Only four studies have directly addressed the pre-
Table 47.1
valence of PSP (Golbe et al., 1988; Schrag et al.,
Incidence studies for progressive supranuclear palsy 1999; Nath et al., 2001; Kawashima et al., 2004).
Table 47.2 summarizes these studies, together with
Year Incidence (cases other estimates of the prevalence of PSP. In the latter,
of Geographical per 100 000 per standard diagnostic criteria were not used and the pri-
Author report area covered year)
mary aim of the work was to determine the preva-
Mastaglia 1973 Perth, 0.4* lence of PD.
et al. Australia Golbe et al. (1988), based in New Jersey, USA, first
Rajput et al. 1984 Minnesota, 0.3 addressed the population prevalence of PSP. Neuro-
USA logists were the only physicians approached, so under-
Radhakrishnan 1988 Benghazi, 0.3 ascertainment would have occurred if PSP cases were
et al. Libya being managed by other specialists. The authors had to
Golbe et al. 1988 New Jersey, 0.5y use their own definition of PSP, since standardized
USA
criteria were unavailable at the time. Despite potential
Bower et al. 1997 Minnesota, 1.1
USA
criticisms, this study established that PSP was not as rare
as had previously been supposed. Golbe (1992) later
*Figure derived from number of cases seen in 2 years with ill- suggested that the true prevalence of symptomatic cases
defined population denominator. could be at least twice that of diagnosed cases, since
y
Figure estimated from prevalence and survival data. the average delay from symptom onset to diagnosis is
Modified from Nath and Burn (2000). approximately 50% of the disease course.
PROGRESSIVE SUPRANUCLEAR PALSY 329
Table 47.2
Prevalence studies for progressive supranuclear palsy (PSP)

Year of PSP prevalence Geographical Population Crude prevalence


Author report primary area studied denominator (per 100 000)

Golbe et al. 1988 Yes New Jersey, USA 799 022 1.39
De Rijk 1995 No Rotterdam, Netherlands 6969 14.3*
Wermuth 1997 No Faroe Islands 43 709 4.6
Chio 1998 No North-west Italy 61 830 3.2
Schrag et al. 1999 Yes London and Kent, UK 121 608 4.9
Nath et al. 2001 Yes Newcastle, UK 259 998 6.5
Kawashima et al. 2004 Yes Yonago, Japan 137 420 5.82

*Only persons aged 55 years of age or older were included.


Modified from Nath and Burn (2000).

Seventeen cases of PSP were identified within an improbably early age and incompatible with PSP
the community study (population 259 998) of Nath according to current diagnostic criteria (Kristensen,
et al., yielding an age-adjusted prevalence figure 1985).
of 5.0 (95% confidence interval (CI) 2.57.5) per There is a remarkably consistent median age of dis-
100 000, standardized to the hypothetical European ease onset in the different series, between 60 and 66
population. When the Schrag data are standardized to years. With the exception of Kristensens review, no
the same population, an identical prevalence figure of case has been reported with a disease onset below
5.0 is obtained. The most recent study, undertaken in the age of 40. The upper limit of the age range for
Japan, also found a similar crude prevalence for PSP
of 5.82 (95% CI 1.789.86) per 100 000 (Kawashima Table 47.3
et al., 2004).
Sex ratio and age at disease onset of progressive
There is a high prevalence of PSP in the French
supranuclear palsy from published series
Antilles, with a minimum prevalence of 14 per 100 000
on the island of Guadeloupe (Caparros-Lefebvre Median age at
et al., 2002). Of 220 consecutive patients with Year of No. of Sex ratio disease onset
Parkinsons syndrome examined, 58 had probable Author report cases (M:F) (range) years
PSP, a further 96 had undetermined parkinsonism
(many of whom closely resembled the incomplete Kristensen* 1985 325y 1.5:1 60.0 (1280)
or a typical bradykinetic presentation of PSP), 50 Maher and 1986 52 0.7:1 63.5 (5077)
Lees
had PD and 15 had an amyotrophic lateral sclero-
Golbe et al. 1988 50 1.4:1 62.9 (4475)
sisparkinsonian syndrome. Pathological confirma-
Frasca et al. 1991 26 1:1 63.1 (4376)
tion of PSP, with a major doublet of pathological De Bruin 1994 90{ 1.5:1 62.0 (4778)
tau at 64 and 69 kDa in brain tissue homogenates and
(see below), has been found in all three of the prob- Lees*
able PSP cases coming to postmortem. Collins 1995 12{ 3:1 66.0 (4877)
et al.
Litvan et al. 1996c 24{ 1.6:1 63.0 (4573)
47.3.3. Age of onset and sex ratio Verny et al. 1996 21{ 0.4:1 62.0 (4370)
Santacruz 1998 437} 1.1:1 66.0 (4187)
Table 47.3 summarizes the sex ratio and age of onset et al.
of PSP in published series to date (Kristensen, 1985;
Frasca et al., 1991; De Bruin and Lees, 1994; Collins *Review of published cases.
y
et al., 1995; Litvan et al., 1996c; Verny et al., 1996; Sex and disease onset specified in 302 and 202 cases, respectively.
{
All cases pathologically confirmed.
Santacruz et al., 1998). One must be cautious in the }
Admixture of living and deceased patients, surveyed by postal ques-
interpretation of cases where no pathological confir- tionnaire. Median age at disease onset is approximated from data
mation is available. In one review, the lower end of given in paper.
the range for age at onset was reported as 12 years Modified from Nath and Burn (2000).
330 D. J. BURN AND A. J. LEES
PSP must also be viewed cautiously, as clinicopatho- Bradykinesia affects nearly half the patients by the
logical series are prone to bias and older cases are time of diagnosis and up to 95% of PSP patients
less likely to undergo postmortem (Maraganore et al., during the course of their illness (Maher and Lees,
1998). 1986; Verny et al., 1996). Bilateral bradykinesia was
Men may be diagnosed later than women follow- reported in 88% of the NINDS series (Litvan et al.,
ing symptom onset (33.4 versus 24.1 months), but 1996c). Axial and nuchal rigidity, in particular, may
die earlier following the diagnosis (37.0 versus 47.6 also be marked but disproportionate retrocollis, al-
months) (Santacruz et al., 1998). Regarding the sex though thought to be characteristic of PSP, is usually
ratio of PSP, six series have found a male predomi- a late and relatively infrequent sign (Litvan, 2001).
nance, two a female predominance and one an even Generalized hyperreflexia is common, but of limited
ratio (Table 47.3). The most parsimonious explana- diagnostic help. Tremor may be reported in up to
tion is that there is no significant difference in the 20% of cases; this is usually postural in type (Nath
sex ratio. et al., 2003). A classic pill-rolling tremor is atypical
for PSP (Quinn, 1997). The profoundly reduced blink
frequency, facial dystonia and gaze abnormalities
47.4. Classical progressive supranuclear palsy
produce an unnervingly impassive face (so-called rep-
(Richardsons syndrome)
tilian stare). Dystonic vertical wrinkles in the glabel-
47.4.1. Clinical features lar region and bridge of the nose have been termed
the procerus sign (Romano and Colosimo, 2001),
Poor mobility and slowness of movement are the most although the use of this term has been questioned by
commonly occurring symptoms at disease onset, others (Lepore and Moudgil, 2002). Glabellar and pal-
occurring in nearly 70% of cases, followed by cogni- momental reflexes are present in 92% and 25% of
tive problems (15%) and bulbar dysfunction (14%) PSP, respectively, but do not correlate with cognition,
(Nath et al., 2003). Falls occur in the majority of as assessed using the Mini-Mental State Examination
patients and are typically backwards (Maher and Lees, (MMSE) (Brodsky et al., 2004).
1986). In the National Institute of Neurological Dis- Early bulbar dysfunction is notable, with a peculiar
orders (NINDS) study, 83% of 24 PSP patients had a growling dysarthrophonia (Litvan et al., 1996c).
history of falls (Litvan et al., 1996c), whereas 88% Swallowing difficulties, reported in 60% of patients,
reported this problem out of 187 cases studied by Nath may be severe enough to warrant insertion of a per-
and coworkers (2003). cutaneous endoscopic gastrostomy feeding system in
The supranuclear gaze paresis characteristic of PSP up to 10% (Nath et al., 2003).
is in the vertical plane, with non-targeted sac- Vague changes in personality may be amongst the
cadic movements first affected. As the clinical picture earliest signs of PSP, at a time when the diagnosis can-
evolves, slow pursuit eye movements are involved not be made with any degree of certainty. These
and eventually there may be complete vertical and hor- changes include forgetfulness, anhedonia, irritability,
izontal gaze ophthalmoparesis. Electro-oculographic depressed mood and impaired concentration. The cog-
studies confirm early and persistent saccadic slowing nitive problems may worsen with disease progression
in PSP compared to other parkinsonian syndromes and ultimately conform to a pattern of subcorticofron-
whereas, unlike corticobasal degeneration (CBD), sac- tal dementia, characterized by a slowing of thought
cadic latency is normal (Vidailhet et al., 1994; Rottach processes and impaired executive functions with recall
et al., 1996; Rivaud-Pechoux et al., 2000). The per- deficit improved by cueing, recognition and preserva-
centage of errors in an antisaccade task, an index of tion of instrumental activities (Grafman et al., 1995;
prefrontal dysfunction, is markedly increased in PSP Aarsland et al., 2003).
(Vidailhet et al., 1994). Square-wave jerks are com- Patients with PSP may also show behavioral
mon in PSP. Vertical optokinetic nystagmus in PSP changes, exhibiting apathy and disinhibition, together
has an impaired slow-phase response and slowed quick with prehension, initiation and utilization behaviors
phases, combined with frequent small, paired, hori- (Pillon et al., 1991). These behavioral and cognitive
zontal saccadic intrusions (Garbutt et al., 2004). Eyelid problems are a source of considerable distress to the
and corneal problems are common, including vague grit- patients and their carers. Depression has not been well
tiness, corneal ulceration, photophobia and apraxia of studied in PSP but may occur in up to 20% of patients
eyelid opening. The latter, occurring in 43% of 49 PSP (Aarsland et al., 2003). Sometimes it may be difficult
patients undergoing a standardized clinical assessment, to discriminate mood disorder from apathy. Rapid-eye
may be severe enough to cause functional blindness movement (REM) sleep behavior disorder is rare in
(Nath et al., 2003). PSP, when compared with synucleinopathic disorders
PROGRESSIVE SUPRANUCLEAR PALSY 331
(PD, multiple system atrophy (MSA) and dementia with An analysis of International Classification of Dis-
Lewy bodies) (Boeve et al., 2003). eases, version 9 (ICD-9)-coded deaths obtained through
The MMSE is relatively insensitive to frontal the UK Office of National Statistics over an 8-year per-
dysfunction and either the Dementia Rating Scale or iod yielded a crude annual mortality rate for PSP of 1.77
the Addenbrookes Cognitive Examination-Revised (95% CI 1.641.90) cases per million (Nath et al.,
(ACE-R) may be more useful for assessing global 2005). In this study, the annual mortality rate increased
cognition in PSP (Aarsland et al., 2003). The Frontal over time, possibly as a result of increased incidence or
Assessment Battery (FAB) is a short bedside cognitive increased awareness of the disorder. The commonest
and behavioral battery, comprising six subtests explor- proximate cause of death in PSP is pneumonia, as cited
ing the following: conceptualization, mental flexibil- in 4565% of death certificates (Maher and Lees, 1986;
ity, motor programming, sensitivity to interference, Nath et al., 2005).
inhibitory control and environmental autonomy
(Dubois et al., 2000). The FAB is sensitive to the fron- 47.5. Misdiagnosis and diagnostic criteria
tal lobe dysfunction of PSP, takes about 10 minutes to
administer and has good discriminant validity. Motor Primary care diagnoses on hospital referral are protean
perseveration is also detected by showing, then asking, and include PD (30%), balance disorders (20%),
the patient to clap three times only. PSP sufferers com- stroke (10%) and depression (7%) (Nath et al., 2003).
monly continue to clap in excess of three times (the Combining the prevalence studies of Schrag et al.
signe dapplause). (1999) and the methodologically similar community-
The combination of subcorticofrontal disturbance based component of the Nath et al. (2001) study, a
and postural instability may lead to the so-called total of 23 PSP cases were identified. Of these, 10
rocket sign, whereby the patient jumps up impulsively patients (43%) carried a primary referral diagnosis of
from a seated position, only to fall back immediately PSP. In the remainder, PD and cerebrovascular disease
into the chair. Sitting en bloc is also a common sign, accounted for all but one of the misdiagnosed cases.
with both feet coming up off the floor as the patient In a clinicopathological study, based in a specialist
falls backwards into their chair when asked to sit down movement disorders service, 19 of 143 cases of parkin-
(Litvan, 2001). sonism were pathologically confirmed as PSP (Hughes
et al., 2002). Antemortem clinical diagnosis was correct
47.4.2. Natural history in 16 of these 19 cases: MSA, PD and parkinsonism
undetermined constituted the three misdiagnoses
Classic PSP is a rapidly progressive neurodegenerative (Hughes et al., 2002). Utilizing the Society for PSP brain
illness. The median duration from disease onset to bank, Josephs and Dickson (2003) found that, of 180
death is 5.85.9 years (Maher and Lees, 1986). Mean cases referred with a clinical diagnosis of PSP, 137 had
interval from symptom onset to diagnosis ranges from this diagnosis confirmed pathologically. CBD, MSA
3.6 to 4.9 years, indicating that many patients with and dementia with Lewy bodies accounted for 70% of
PSP may remain misdiagnosed for much of their dis- the 43 misdiagnosed cases. A history of tremor, psycho-
ease course (Golbe et al., 1988). Onset of falls (hazard sis, dementia and asymmetric findings was more frequent
ratio (HR) 3.28, 95% CI 1.179.13), speech problems in misdiagnosed cases. Using the Queen Square Brain
(HR 4.74, 95% CI 1.1020.4) or diplopia (HR 4.23, Bank for Neurological Disorders, Osaki and colleagues
95% CI 1.2314.6) within 1 year and swallowing pro- (2003) found that the clinical diagnosis of PSP was con-
blems within 2 years (HR 3.91, 95% CI 1.3911.0) firmed pathologically in 78% of 60 cases, a remarkably
were associated with a worse prognosis in one study similar positive predictive value to that obtained by
of 187 PSP cases (Nath et al., 2003). Goetz and collea- Josephs and Dickson. False-positive diagnoses inclu-
gues (2003) approached the natural history in a differ- ded PD with significant cortical Lewy body (n 3) or
ent way, defining key motor impairments in PSP as Alzheimer (n 1) pathology and MSA (n 4). At the
unintelligible speech, no independent walking, inabil- first consultation, only 17% of the 47 true PSP cases,
ity to stand unassisted, wheelchair-bound or recom- 6 of which had been seen by consultant neurologists,
mendation for feeding tube placement. Median time were diagnosed as PSP.
from disease onset to the first key motor impairment Postural instability, leading to falls (typically back-
in a sample of 50 subjects was 48 months, 24 months wards) within the first year of disease onset, coupled
after the first consultation. The three gait items with a vertical supranuclear gaze paresis have good
occurred temporally closely together and, when con- discriminatory diagnostic value when PSP is com-
sidered as a single milestone, occurred at a median pared with other degenerative parkinsonian syndromes
disease duration of only 57 months. (Litvan et al., 1997).
332 D. J. BURN AND A. J. LEES
47.5.1. Diagnostic criteria cal Disorders and Stroke and Society for Progressive
Supranuclear Palsy (NINDS-SPSP) diagnostic criteria
Seven different sets of diagnostic criteria have been (Table 47.5) (Litvan et al., 1996a, 2003). When applied
proposed for PSP (Table 47.4) (Lees, 1987; Blin et al., retrospectively to a case mix comprising various parkin-
1990; Duvoisin, 1992; Golbe, 1993; Tolosa et al., 1994; sonian syndromes, the NINDS-SPSP criteria have high
Collins et al., 1995; Litvan et al., 1996a). In the majority, diagnostic sensitivity and specificity (Litvan et al.,
the criteria were not derived in a systematic fashion, but 2003). These parameters have not, however, yet been
were mainly compiled from the extensive clinical experi- determined for prospective series with pathological cor-
ence of the authors. There is considerable overlap between relation, nor have they been applied retrospectively to
the different sets of criteria, with disease onset over the age an independent clinicopathological series. The accuracy
of 40 or 45 and supranuclear gaze palsy common to all. of the NINDS-SPSP clinical diagnostic criteria has also
The most rigorous approach to date led to the been evaluated along with existing criteria for three
formulation of the National Institute of Neurologi- other dementing disorders by a different group of raters
in an independent sample of pathologically confirmed
Table 47.4
cases (Lopez et al., 1999). This study confirmed that
Published diagnostic criteria for progressive supranuclear both probable and possible NINDS-SPSP diagnostic
palsy categories for PSP had excellent specificity.
One area where the NINDS-SPSP criteria would be
Author (year) Derivation and use
predicted to have lower sensitivity is when the devel-
Lees (1987) Not explicitly stated; defined as progressive opment of core diagnostic features is delayed. Appli-
non-familial disorder beginning in cation of the NINDS-SPSP criteria marginally
middle or old age with SNO and  2 of 5 improved the accuracy of initial clinical diagnosis in
further cardinal features one retrospective clinicopathological series (Osaki
Blin et al. Not explicitly stated; defined as probable et al., 2003), but did not improve accuracy of the final
(1990) if all of 9 criteria met or possible if 7 clinical diagnosis. Diagnostic sensitivity, using the
out of 9 criteria fulfilled possible PSP category of these criteria, was still only
Duvoisin Not explicitly stated; criteria divided into
21%, compared with 17% for initial clinical diagnosis.
(1992) four sections essential for diagnosis,
confirmatory manifestations,
manifestations consistent with but not 47.6. Clinical heterogeneity
diagnostic of PSP and features
inconsistent with PSP Atypical phenotypic variants of PSP add to the
Golbe (1993) Not explicitly stated; defined as onset after difficulty of accurate diagnosis. Pathologically
age 40, progressive course bradykinesia confirmed cases of PSP have been reported in which
and SNO, plus  3 of 6 further features, there was pure akinesia, whereas others have
plus absence of 3 inconsistent clinical documented early and severe dementia (Davis et al.,
features 1985; Matsuo et al., 1991). Additional reports have
Tolosa et al. Not explicitly stated; defined as a non- described features that would conventionally be
(1994) familial disorder of onset after age 40,
considered unusual for PSP, including unilateral limb
progressive course and SNO, plus  3 of
dystonia or apraxia, prominent tremor, palatal myoclo-
5 further features for probable and 2 of
5 for possible, plus absence of 5 nus and cricopharyngeal dysfunction (Schleider and
inconsistent clinical features Nagurney, 1977; Masucci and Kurtzke, 1989; Barclay
Collins et al. Retrospectively from review of 12 and Lang, 1997; Pharr et al., 1999). Conversely, cases
(1995) pathologically confirmed cases; of frontotemporal lobar degeneration with ubiquitin-
algorithm-based, including prerequisites only-immunoreactive neuronal changes (including
and exclusionary criteria; SNO and/or the frontotemporal lobar degeneration with motor
prominent postural instability, plus a neuron disease (FTLD-MND) variant) (Paviour et al.,
number of other specified signs 2004), frontotemporal dementia linked to chromosome
Litvan et al. Systematic literature review, logistic 17 (tau exon 10 16 mutation) (Morris et al., 2003),
(1996a) regression and CART analysis; validated
Whipples disease (Averbuch-Heller et al., 1999),
using data from postmortem confirmed
neurosyphilis (Murialdo et al., 2000), cerebral
cases; definite, probable and
possible categories described autosomal dominant arteriopathy with subcortical
(see Table 47.5 and text) infarcts and leukoencephalopathy (CADASIL) (Van
Gerpen et al., 2003), primary antiphospholipid anti-
SNO, supranuclear ophthalmoparesis; CART, classification and body syndrome (Reitblat et al., 2003), Creutzfeldt
regression tree analysis. Jakob disease (Josephs et al., 2004), clebopride
PROGRESSIVE SUPRANUCLEAR PALSY 333
Table 47.5
NINDS-SPSP diagnostic criteria for progressive supranuclear palsy (PSP)

Mandatory exclusion
PSP Mandatory inclusion criteria criteria Supportive criteria

Possible Gradually progressive disorder Recent history of Symmetric akinesia or rigidity, proximal
encephalitis more than distal
Onset age 40 or later Alien-limb syndrome, Abnormal neck posture, especially
cortical sensory deficits, retrocollis. Poor or absent response of
focal frontal or parkinsonism to levodopa. Early
temporoparietal atrophy dysphagia and dysarthria. Early onset
Either vertical supranuclear palsy or Hallucinations or delusions of cognitive impairment including
both slowing of vertical saccades unrelated to > 2 of: apathy, impairment in abstract
and postural instability with falls dopaminergic therapy thought, decreased verbal fluency,
< 1 year after disease onset utilization or imitation behavior or
No evidence of other diseases that Cortical dementia of frontal release signs
could explain the foregoing features, Alzheimer type
as indicated by exclusion criteria Prominent, early cerebellar
symptoms or unexplained
autonomic dysautonomia
Probable Gradually progressive disorder
Onset age 40 or later
Vertical supranuclear palsy and
prominent postural instability with
falls < 1 year after disease onset
No evidence of other diseases that
could explain the foregoing features,
as indicated by exclusion criteria
Definite Clinically probable or possible PSP
and histopathological evidence
of typical PSP

NINDS-SPSP, National Institute of Neurological Disorders and Stroke and Society for Progressive Supranuclear Palsy.
Modified from Litvan et al. (1996a).

exposure (Campdelacreu et al., 2004) and postsurgical was, at 9.2 years, significantly longer than the classic
repair of ascending aortic dissection or aneurysm (Mokri cases (5.9 years). It was suggested that the first group
et al., 2004) have been reported with a PSP phenotype. of cases might be named Richardsons syndrome
Morris and colleagues (2002a) have drawn attention and the second group PSP-parkinsonism (PSP-P)
to a clinically atypical form of PSP, in which a classi- (Williams et al., 2004).
cal distribution of PSP pathology occurs, but with the
deposition of Alzheimer disease-type tau protein rather 47.7. Investigations
than PSP-type tau (see below). Four of these 15 atypi-
cal cases had PD-like presentations with an asymme- The diagnosis of PSP still rests on the clinical history
trical onset and good levodopa response and 3 of the and examination. Attempts have been made, however,
4 were documented to have normal eye movements. to improve diagnostic accuracy through cerebrospinal
Another case had asymmetrical dystonia and apraxia, fluid (CSF) analysis and protein biomarkers, structu-
suggestive of CBD. ral and functional imaging and neurophysiological
Using factor analysis, Williams and colleagues techniques.
(2004) identified two distinct groups amongst 107
consecutive cases of pathologically confirmed PSP. 47.7.1. Neuropsychological assessment
Fifty-three percent of cases had features compatible
with the classic clinical description outlined above. Neuropsychological assessment in the early stages may
A second group of 32% cases, however, were charac- assist the accurate clinical diagnosis of a parkinsonian dis-
terized by asymmetric onset, tremor and response to order, as may the time course and pattern of progression of
levodopa. Mean disease duration in this group cognitive and behavioral decline (Soliveri et al., 2000).
334 D. J. BURN AND A. J. LEES
Patients with PSP show a greater decline in attention, 47.7.3. Magnetic resonance imaging (MRI)
set-shifting and categorization abilities, compared with
PD and MSA. Patients with PSP also show greater Over 70% of patients with clinically diagnosed PSP
impairment in both phonemic and semantic fluency may be correctly classified on the basis of 0.5 T
than patients with MSA or PD. Using discriminant or 1.5 T MRI brain scanning (Schrag et al., 2000).
function analysis, variables derived from four verbal Criteria used for the diagnosis of PSP include midbrain
fluency tasks (simple and alternate semantic and phone- diameter on axial scans of less than 17 mm, signal
mic fluency) were able to classify correctly over 90% increase in the midbrain, atrophy or signal increase
of PSP patients (Lange et al., 2003). The Katz Adjust- of the red nucleus and signal increase in the globus
ment Scale-Relatives (KAS-R) may be sensitive to pallidus. Other studies have also suggested that
changes in apathy, social withdrawal and independence reduced midbrain diameter on routine MRI may be
in PSP (Millar et al., 2006). Comparative studies are of value in discriminating PSP from PD and MSA-P,
required, however, to assess the sensitivity and although values may overlap and do not clearly corre-
specificity of this profile in differentiating PSP from late with disease duration or severity (Warmuth-Metz
other parkinsonian conditions. et al., 2001; Righini et al., 2004). Atrophy or abnormal
signal of the superior cerebellar peduncle on proton
47.7.2. Cerebrospinal fluid analysis density-weighted MRI, postulated to represent demye-
lination and gliosis, may also be of help in differentiat-
Studies have attempted to identify biomarkers in the ing PSP from PD (Oka et al., 2001). Pathological data
CSF to achieve early and accurate diagnosis, as well indicate that atrophy of this structure is a relatively
as monitoring response to treatment. CSF amyloid early feature of PSP and correlates with disease dura-
A-b42 levels are normal in PSP and do not discrimi- tion (Tsuboi et al., 2003). Tegmental T2 hyperintensity
nate this condition from PD (Holmberg et al., 2003). has poor sensitivity for the diagnosis of PSP (Righini
Tau has potential as a candidate protein, although it et al., 2004). In all radiological studies, clinically
may lack specificity unless isoform analysis can also typical PSP cases were selected, rendering the discri-
be performed (see below). Significantly higher tau minatory value of MRI something of a tautology. The
protein levels in CSF have been reported in CBD, value of routine MRI scanning in longitudinal clinico-
compared with PSP, yielding sensitivities and speci- pathological studies, particularly of early indeterminate
ficities of 100% and 87.5%, respectively (Urakami cases, has not been investigated.
et al., 2001). Other proteins in the CSF, including MRI-based volumetry (MRV) has also been used to
neurofilament protein (NFL) and glial fibrillary acidic differentiate PSP from other parkinsonian syndromes,
protein (GFAP), have also been studied. Whereas no by examining atrophy of the caudate nucleus, puta-
difference was found in CSF GFAP levels between men, brainstem and cerebellum (Schulz et al., 1999).
PD, MSA and PSP, high NFL concentrations seemed Although significant group differences were found in
to differentiate typical from atypical parkinsonian dis- mean striatal and brainstem volumes, on an individual
orders (Holmberg et al., 1998). The overlap in ranges, patient basis this technique could still not reliably
however, limits the sensitivity of this technique and it separate PSP from MSA-P. Voxel-based morphome-
was not possible to differentiate MSA from PSP cases. try, an observer-independent MRV technique, has
More recently, Holmberg and colleagues (2001) sug- demonstrated atrophy in PSP, predominantly involving
gested that the concomitant use of a levodopa test in mesiofrontal targets of striatal projections (Brenneis
combination with CSF NFL assay could improve diag- et al., 2004). This study did not compare PSP with
nostic accuracy for atypical parkinsonism to 90%. other parkinsonian syndromes, however, so the diag-
Major products of lipid peroxidation are selectively nostic value of these observations is unknown. More
increased in PSP midbrain tissue, suggesting that a elaborate mathematical modeling has also been
CSF assay for these products could provide a specific applied in an attempt to discriminate PSP from CBD
biomarker. using MRV (Groschel et al., 2004). As for MRI, the
Since the clinical phenotype of Whipples disease potential utility of MRV remains to be established in
may resemble PSP, investigators have examined CSF prospective studies, including subjects with early
from PSP patients for Tropheryma whippelii DNA indeterminate parkinsonism.
(Pezzella et al., 2004). Polymerase chain reaction Proton magnetic resonance spectroscopy may pro-
for T. whippelii was negative in all samples tested, vide an indirect measure of neuronal loss in vivo.
indicating that this pathogen is not involved in the There have been a number of reports of the use of
etiopathogenesis of PSP, nor is it a useful biomarker this technique in PSP, concentrating mainly upon
for the disease. spectral changes in the lentiform nucleus. In general,
PROGRESSIVE SUPRANUCLEAR PALSY 335
although lentiform N-acetylaspartate/choline and/or N- and statistical parametric mapping highlights hypome-
acetylaspartate/creatine ratios may be reduced in PSP, tabolism in cingulate gyrus, caudate nucleus, thalamus
the discriminatory value of this technique on an indi- and midbrain in PSP (Juh et al., 2004), compared to nor-
vidual basis remains questionable. A systematic mal controls, PD and MSA patients, whereas midbrain
review of proton magnetic resonance spectroscopy in hypometabolism may be an early diagnostic marker
parkinsonian syndromes concluded that the heteroge- for PSP (Mishina et al., 2004). Both findings require
neity of the results to date precludes the use of any validation in independent, prospective series.
of these findings in differential diagnosis at the Evaluation of the nigrostriatal dopaminergic system
present time (Clarke and Lowry, 2001). using cocaine analogs and functional imaging (PET or
Apparent diffusion coefficient measurements using SPECT) reliably shows presynaptic dopaminergic
diffusion-weighted MRI (DWI) may discriminate degeneration in PSP and can also demonstrate pro-
PSP from PD with a sensitivity of 90% and a positive gression of this degeneration. The pattern of abnormal-
predictive value of 100%; significant increases in ity is, however, non-specific and cannot differentiate
regional apparent diffusion coefficients have been PSP from other parkinsonian syndromes (Pirker
noted in striatum and globus pallidus in PSP cases et al., 2002), even when used in combination with
(Seppi et al., 2003). Importantly, however, DWI could a dopamine D2-receptor ligand (Kim et al., 2002;
not discriminate PSP from MSA-C in this study. Mag- Plotkin et al., 2005). A newly developed PET ligand,
netization transfer imaging, a measure that correlates the dopamine D2-receptor antagonist, [18F]-desmethoxy-
with myelination and axonal density, has revealed fallypride, can discriminate between typical and atypi-
changes in magnetization transfer ratios corresponding cal parkinsonism with high specificity (100%) and
with known sites of pathological predilection in PSP, sensitivity (76%), but cannot reliably differentiate
MSA and PD (Eckert et al., 2004). The same study PSP from MSA, limiting its use in clinical practice
showed fairly good discrimination of PD from (Schreckenberger et al., 2004).
control subjects and of MSA from PSP. The lack of specificity in functional imaging of the
dopaminergic system has led to the study of other neu-
47.7.4. Brain parenchyma sonography (BPS) rotransmitter systems in PSP. Thus, significant reduc-
tions in both caudate and putamen opioid receptor
BPS is a new ultrasound technique capable of display- binding were seen in PSP using [11C]-diprenorphine
ing tissue echogenicity of the brain through an intact PET, compared with PD, MSA and controls (Burn
skull. Information may be obtained from key brain et al., 1995). [11C]-flumazenil and PET, used to image
structures that may be of value in differentiating PSP benzodiazepine receptors, demonstrated a modest
from other parkinsonian syndromes. BPS can be useful reduction in binding in the anterior cingulate gyrus
in discriminating typical from atypical parkinsonism when compared with normal controls, but no other
(MSA and PSP) (Walter et al., 2003). Marked hypere- regional abnormality (Foster et al., 2000). Although
chogenicity of the substantia nigra has been reported benzodiazepine binding is reduced in the globus palli-
in CBD but not in PSP, whereas dilatation of the third dus of autopsy tissue from PSP patients and preserved
ventricle is common in PSP, but not in CBD. The pre- in the striatum, the low level of the receptors in the
sence of at least one of the BPS findings, marked pallidum and the proximity of the putamen indicate
nigral hyperechogenicity and third ventricle width functional imaging of the benzodiazepine receptor is
<10 mm, indicated CBD with sensitivity of 100%, a too insensitive to detect these differences.
specificity of 83% and a positive predictive value of Ligands for the cholinergic system may offer more
80% in one small cross-sectional study (Walter et al., hope in improving diagnostic accuracy for PSP. Ana-
2004). logs of vesamicol, an inhibitor of the acetylcholine vesi-
cular transporter, demonstrate loss of intrinsic striatal
47.7.5. Functional imaging cholinergic neurons (Eidelberg and Dhawan, 2002).
N-methyl-4-[11C]piperidyl acetate and PET reflect ace-
Blood flow and oxygen or glucose metabolism positron tylcholinesterase activity and indicate preferential loss
emission tomography (PET) studies in PSP subjects of cholinergic innervation to the thalamus in PSP,
have demonstrated relative frontal lobe hypometa- compared with PD and controls (Shinotoh et al., 1999).
bolism, although bifrontal hypoperfusion using In vivo imaging of activated microglia, using [11C]
more widely available 99mTc-HMPAO (99mTc-hexa- PK11195 and PET, revealed increased binding in the
methylpropylene amine oxime) single-photon emission lentiform nucleus and pons, in particular, although dor-
computed tomography technique (SPECT) is not a solateral prefrontal cortex, caudate, substantia nigra
robust finding in PSP. [18F]-Fluorodeoxyglucose PET and thalamus were also involved (Gerhard et al., 2001).
336 D. J. BURN AND A. J. LEES
The significance of this preliminary finding (in only 2 optimal criteria for PSP, according to the NINDS-SPSP
PSP patients) is uncertain. criteria, implying early postural instability and falls
within the first year of disease onset. The sensitivity
47.7.6. Neurophysiological techniques and specificity of this and several of the other tech-
niques described above in prospectively followed
A variety of neurophysiological techniques have been indeterminate parkinsonian cases would be of interest.
used to study PSP, both with the aim of improving diag- Sleep architecture studies in PSP patients are char-
nostic accuracy and also improving understanding of acterized by almost complete absence of REM sleep,
the underlying pathophysiological process. A longitud- reduction of total sleep time and sleep spindles, atonic
inal oculomotor study of patients with PSP and other slow-wave sleep and fragmentation with disrup-
parkinsonian syndromes has suggested that electro- ted sleep pattern (Montplaisir et al., 1997; Brotini
oculography may help diagnose PSP earlier. PSP and Gigli, 2004). Cell loss in the pedunculopontine
patients are characterized by decreased saccadic velo- tegmentum may be the pathophysiological basis for
city throughout their disease course, as well as having the absence of REM sleep.
less specific findings of frequent square-wave jerks
and increased error rate on antisaccade tasks (Rivaud- 47.8. Pathology
Pechoux et al., 2000). Significant orthostatic hypoten-
sion is rare in PSP, in contrast to MSA, with only minor The majority of PSP cases have little or no visible atro-
and inconsistent abnormalities on formal assessment of phy macroscopically, distinguishing it from other tauo-
sympathetic and parasympathetic functions. Further- pathies (Schofield et al., 2004). PSP is characterized by
more, there is a normal rise in growth hormone follow- the destruction of a number of subcortical structures,
ing clonidine administration in PSP patients (Kimber including the substantia nigra, globus pallidus, sub-
et al., 2000). Sphincter electromyography may differ- thalamic nucleus and midbrain and pontine reticular
entiate atypical akinetic-rigid syndromes from PD, but formation (Jellinger and Blancher, 1992). Deeper corti-
fails to discriminate reliably between PSP and MSA cal layers, especially around the precentral gyrus, may
(Vodusek, 2001). Neuronal loss in Onufs nucleus of also be affected to a lesser degree. Large numbers of
the sacral spinal cord in PSP could explain this lack of neurofibrillary tangles (NFTs, made up of ultramicro-
specificity (Scaravilli et al., 2000). The auditory startle scopic straight filaments), neuropil threads and tufted
response is delayed or absent in PSP and without habi- astrocytes are also found in these brain regions. These
tuation, although there is overlap with values obtained distinctive histopathological inclusions are made up of
from other akinetic-rigid syndromes. A combination of insoluble aggregates of tau phosphoprotein. Coiled
abnormalities on acoustic blink reflex, acoustic startle bodies are small round cells of oligodendrocytic origin
reflex and electro-oculography may have high specifi- found in white matter, underlining the widespread nat-
city (95%) and sensitivity (100%) for an early diagnosis ure of the pathology in PSP. Tau-positive glial inclu-
of PSP, according to one prospective study of 41 atypi- sions are also positively stained with antibodies
cal parkinsonian patients with mean 26-month clinical against ubiquitin, DJ-1 (Neumann et al., 2004) and heat
follow-up (Gironell et al., 2003). Abnormalities in shock protein (HSP) ab-crystallin (but not HSP70)
visual event-related potentials (P300 amplitude and (Richter-Landsberg and Bauer, 2004). In contrast to
reaction times to rare target stimuli), somatosensory CBD, ballooned neurons are sparse in PSP and limited
evoked potentials (enlarged cortical somatosen- to the limbic system (Wakabayashi and Takahashi,
sory evoked potentials) and pattern of facial reflexes 2004).
(electromyogram activity in mentalis but not orbicularis
oculi muscles following electrical stimulation of the 47.8.1. Neuropathological diagnostic criteria and
median nerve at the wrist) have all been reported in differential diagnosis
PSP patients. Transcranial magnetic stimulation reveals
disinhibition of the motor cortex in PSP, with enlarged There are a number of clinical and pathological similari-
response amplitudes at rest, reduced intracortical inhibi- ties between PSP, PEP and the parkinsonismdementia
tion and prolonged ipsi- and contralateral silent periods complex of Guam (PDCG) (Geddes et al., 1993).
(Kuhn et al., 2004; Wolters et al., 2004). The clinical When no clinical information is given, the neuro-
utility of these investigations remains uncertain, as does pathologist may have difficulty in differentiating PSP
their pathophysiological significance. Computerized from PEP and PDCG. The sensitivity and reliability
posturography testing may differentiate early PSP from of the diagnosis of PEP improve significantly when
early PD and age-matched controls (Ondo et al., 2000). clinical data are available (Litvan et al., 1996b). NFTs
Seventy-five percent of the 20 PSP patients met all in subcortical and cortical areas are found in all three
PROGRESSIVE SUPRANUCLEAR PALSY 337
conditions. PSP may also be difficult to discriminate fibrillar aggregates that are resistant to proteolysis.
from CBD, where NFTs also occur and where the A number of posttranslational processes may be in-
basophilic inclusions of CBD are not always readily volved in the aggregation of tau in PSP, but hyperpho-
distinguished from the tangle pathology. sphorylation, glycation and transglutamination have
Standardized neuropathological criteria have been been principally implicated. Glycation leads to the for-
developed for PSP. An early version divided the neu- mation of advanced glycation end-products (AGEs),
ropathology of PSP into typical, atypical and com- detected histochemically in NFTs (Sasaki et al.,
bined types, although the value of the atypical 1998). The tau in these aggregates is abnormally
category was controversial (Hauw et al., 1994). hyperphosphorylated and in turn this may lead to
Revised criteria subsequently omitted this category, tau release from microtubules. Tissue transglut-
leaving typical and combined PSP, with the latter asso- aminase (TGase) is a calcium-activated enzyme which
ciated with infarcts in the brainstem, basal ganglia, or cross-links substrate proteins into insoluble, protease-
both sites (Table 47.6) (Litvan et al., 1996b). A clini- resistant complexes, potentially initiating NFT forma-
cal history compatible with PSP is mandatory when tion. By altering the conformation of tau, TGase may
defining typical PSP, according to these criteria. render digestion sites inaccessible to proteases. In sup-
port of a pathogenic role for TGase, high levels of
47.8.2. Tau aggregation and cell death epsilon-(gamma-glutamyl) lysine cross-linked tau,
together with increased TGase and mRNA levels for
Tau is critically important for the dynamic behavior TGase 1 and 2 have been found in the pallidum and
and stabilization of microtubules in the cytoskeleton. pons in PSP (Zemaitaitis et al., 2003). Antibodies cap-
In normal neurons, tau is soluble, unfolded and binds able of detecting nitrated tau have also shown labeling
reversibly to microtubules, enhanced by chaperone in the glial and neuronal tau of PSP, in addition to
activity of HSPs. In PSP the protein loses its affinity other tauopathies, implying that nitrative injury might
for microtubules and excess free tau accumulates into also be involved (Horiguchi et al., 2003). In addition
to pathogenic mechanisms leading to tau hyperpho-
Table 47.6
sphorylation, proteasomal inhibition may also be
necessary to produce the stable fibrillary deposits of
NINDS-SPSP neuropathological criteria for the diagnosis tau found in PSP, as indicated by results from an
of typical progressive supranuclear palsy (PSP) in vitro oligodendroglial cell model (OLN-t40) (Gold-
baum et al., 2003). Indirect evidence for proteasome
Inclusion criteria Exclusion criteria
inhibition in PSP is derived from immunohistochem-
Typical A high density of Large or numerous ical and immunoblotting studies, demonstrating an
PSP neurofibrillary infarcts; marked aberrant ubiquitin protein (UBB1) in human pons tis-
tangles and neuropil diffuse or focal sue (Hope et al., 2004). UBB1 is regarded as a repor-
threads in > 3 of: atrophy; Lewy ter for proteasomal dysfunction. Interestingly, the
pallidum, bodies; changes presence of UBB1 was not associated with an HSP
subthalamic nucleus, diagnostic of response. Since HSPs are linked to normal tau func-
substantia nigra or Alzheimers disease;
tion and also act as a rescue response to UBB1
pons and a low-to- oligodendroglial
expression, this could explain both tau dysfunction
high density of argyrophilic
neurofibrillary inclusions, Pick and tau accumulation in PSP (Hope et al., 2004).
tangles or neuropil bodies; diffuse The mechanism leading to cell death in PSP is
threads in > 3 of: spongiosis; prion unknown but is likely to be multifactorial, with both
striatum, oculomotor P-positive amyloid environmental (toxic) and genetic influences playing
complex, medulla, or plaques a role. Microglial activation is greater in PSP than in
dentate nucleus* and control brains and correlates with tau burden in most
clinical history areas. There is also accumulating evidence for oxida-
compatible with PSP tive stress and mitochondrial dysfunction in PSP
(Albers and Augood, 2001). Complex I activity is sig-
*Tau-positive astrocyte processes or astrocyte cell bodies in the nificantly reduced compared with controls in transmi-
areas of neurofibrillary tangles and neuropil threads confirm the
tochondrial cytoplasmic hybrid (cybrid) cell lines,
diagnosis; other lesions include various degrees of neuronal loss
and gliosis in affected areas. expressing mitochondrial genes from subjects with
NINDS-SPSP, National Institute of Neurological Disorders and PSP. The mitochondrial dysfunction may be of toxic
Stroke and Society for Progressive Supranuclear Palsy. or genetic origin. In addition, tau-positive astrocytes
Modified from Litvan et al. (1996b). may exert neurotoxicity through the overproduction
338 D. J. BURN AND A. J. LEES
of nitric oxide, in excess of the detoxification capacity showed significant reduction in cortical and striatal
of superoxide dismutase. The formation of AGE-tau, glucose metabolism.
detectable in NFTs, is also associated with the genera- The relative rarity of familial PSP cases may be due
tion of oxygen free radicals and the induction of oxida- to a failure to recognize atypical cases, other dia-
tive stress. Phosphorylation of tau in PSP and Picks gnostic problems or death of the carriers before the
disease may be a direct consequence of the oxidative appearance of clinical symptoms. The use of neuro-
stress-induced activation of mitogen-activated protein physiological and/or imaging techniques to detect pre-
kinases, including the p38 pathway (phosphor-MKK6 symptomatic cases could assist in linkage analysis of
and phosph-p38) (Hartzler et al., 2002). potential PSP families, with a view to identifying cau-
The consumption of tropical plants and herbal teas sative genes. Conversely, families with phenotypically
has been linked with an abnormally high frequency of characteristic PSP may have been reported where
a levodopa-resistant form of parkinsonism, clinically molecular pathology would be indicative of another
and pathologically resembling PSP, in Guadeloupe neurodegenerative condition. Frontotemporal dementia-
(French West Indies) (Caparros-Lefebvre et al., 1999). parkinsonism (FTDP-17), for example, is clearly linked
Stabilization or even improvement in some symp- to mutations in the tau gene (see below) and may mimic
toms has been reported after cessation of consumption PSP clinically.
of these fruits and infusions. Moreover, when mesence-
phalic dopaminergic neurons are exposed in culture to 47.8.4. Molecular pathology
corexime and reticuline, the most abundant subfractions
of Annona muricata (corossol, soursop), apoptotic cell The human tau gene (MAPT) is located on chro-
death occurs (Lannuzel et al., 2002). Cell death in these mosome 17q21 and contains 16 exons. Six different
cultures seems independent of excitotoxic mechanisms, isoforms of tau are found in the human brain, gener-
although energy depletion has been implicated. Low- ated by alternate splicing of exons 2, 3 and 10. These
molecular weight benzylisoquinoline derives of the Ann- isoforms can be divided into two groups of three, differ-
onaceae family (reticuline and N-methylcoculaurine) ing in the presence of three or four repeated microtubule-
are toxic to SH-SY5Y neuroblastoma cells and can binding domains (three-repeat or four-repeat tau). The
inhibit mitochondrial complex I in vitro (Kotake et al., isoform is determined by whether the exon 10 coding
2004). for one of the four microtubule-binding repeat domains,
a 31-amino-acid repeat located in the C-terminal part, is
47.8.3. Familial progressive supranuclear palsy spliced in or out of the final tau protein product. In
normal brain, there is a slight preponderance of three-
Although PSP is considered to be a late-onset, sporadic repeat tau, whereas in PSP the ratio is at least 3:1 in favor
neurodegenerative disease, a number of families with of four-repeat tau. This isoform ratio contrasts with
sometimes heterogeneous clinical presentation have Alzheimers disease, in which the paired helical fila-
been described. Postmortem confirmation of diagnosis ments contain both three- and four-repeat tau, and also
in one or more cases has been obtained in many of with Picks disease where three-repeat tau is the major
these reports. In a report of 12 pedigrees, the presence component (van Slegtenhorst et al., 2000). Bodig
of affected members in at least two generations in (PDCG) has tau inclusions with an isoform composition
eight of the families and the absence of consangui- similar to Alzheimers disease but very few studies have
nity suggested autosomal-dominant transmission with been reported so far. Tau isoform composition varies
incomplete penetrance (Rojo et al., 1999). markedly within PSP, without a simple relationship
A clinical study yielded the intriguing observation between isoform composition and the pattern of insolu-
that 39% of 23 asymptomatic first-degree relatives of ble tau before dephosphorylation (Gibb et al., 2004).
patients with PSP scored abnormally on a PD test bat- Some of the clinically atypical PSP cases reported by
tery, compared with none of 23 age-matched normal Morris and colleagues (2002a) were also found to have
controls (Baker and Montgomery, 2001). The authors a triplet band, raising the possibility that bodig may not
suggested that the test battery could have detected an be a disorder restricted to a few geographic isolates.
asymptomatic carrier state or risk for PSP, or a sub- These differences may help the neuropathologist to cate-
clinical effect of a shared environmental exposure. gorize and distinguish the tauopathies, since electrophor-
Further evidence for subclinical cases comes from esis reveals two main protein bands of 64 and 68kDa in
PET studies using 18F-dopa and 18fluorodeoxyglu- PSP and CBD, whereas Picks disease has two lighter
cose (Piccini et al., 2001). Four of 15 asymptomatic bands of 55 and 64 kDa. Furthermore, PSP and CBD may
relatives scanned from two families with PSP had be distinguishable on immunoblots of sarkosyl-insoluble
abnormal striatal 18F-dopa uptake and a fifth subject brain extracts by different patterns of amino-terminally
PROGRESSIVE SUPRANUCLEAR PALSY 339
cleaved tau fragments, with a 33-kDa band predo- mutations. However, a sequence analysis of MAPT
minating in PSP, compared with two closely related exons 913 in two small families with PSP and 7 clini-
bands of approximately 37 kDa in CBD (Arai et al., cally typical and atypical sporadic PSP cases with
2004). pathological confirmation of diagnosis has not identi-
The discovery of mutations in MAPT in some fied coding or splice site mutations, suggesting that
families with FTDP-17 confirmed that tau dysfunction PSP or typical PSP-like syndromes are not due to muta-
can lead to neurodegeneration. In some of these tions in MAPT (Morris et al., 2002b). A recent study
families, the three- to four-repeat tau ratio is similar concluded that screening for MAPT gene mutations in
to that found in PSP. A number of different MAPT sporadic cases is unlikely to identify pathogenic muta-
mutations have now been found in FTDP-17 families, tions (Stanford et al., 2004). However, based on asso-
in and near the 50 splice site, downstream of exon 10 ciation studies, it is possible that pathogenic variation
(Hutton, 2000). Through disruption of a stem-loop in non-coding regions (e.g. introns and promoter and
structure formed in pre-mRNA, 50 splice site mutations 30 or 50 UTR) could be involved in influencing expres-
increase recognition of exon 10 by U1 snRNP splicing sion levels of mRNA.
factor, increasing the proportion of exon 10 mRNA Conrad and colleagues (1997) first reported a poly-
and thus four-repeat tau. Analysis of FTDP-17 families morphic dinucleotide repeat sequence in intron 9
would support this stem-loop hypothesis and its (between exons 9 and 10) of MAPT in which the A0
pathogenicity, in that mutations with the greatest effect allele (TG repeat number of 11) and in particular the
on splicing in vitro cause an earlier age of disease A0/A0 genotype were overrepresented in PSP cases
onset. Disruption of the stem-loop structure need not, compared with controls in the white population. These
of course be only genetic in origin and toxic causes data were later extended to a haplotype, H1, including
could also be involved. several polymorphisms in linkage disequilibrium with
There is some evidence from analysis of tau mRNA A0, spanning MAPT (Baker et al., 1999). During evo-
in affected brain regions in PSP that selective four- lution of the two human tau haplotypes, H1 and H2,
repeat tau deposition in PSP may also involve disrup- almost no recombination has occurred between the
tion of exon 10 alternative splicing. Furthermore, there two alleles. Although the H1 allele has a frequency
have now been two reported families with a clinical approaching 100% in pathologically confirmed
syndrome resembling PSP where mutations in MAPT patients with PSP, it is also found in about 70% of
have been detected (Stanford et al., 2000; Pastor controls. It is not known at the present time whether
et al., 2001). In the first, Australian kindred, a silent there is a rarer mutation on the H1 haplotype predis-
mutation (S305S) was identified in the stem-loop posing to PSP, or whether it is the haplotype itself.
structure. Although not producing an amino acid sub- The presence of an H1 haplotype or H1/H1 genotype
stitution (hence silent), functional exon-trapping ex- may therefore be regarded as no more than a modest
periments suggested that the mutation caused up to genetic predisposition towards developing PSP.
a fivefold increase in splicing of exon 10, resulting Furthermore, almost all normal Japanese people carry
in overexpression of four-repeat tau. In the second, the H1/H1 genotype. The H1 haplotype seems to have
Spanish kindred, two brothers born from a third-degree no effect upon the tau or amyloid burden in the lenti-
consanguineous marriage were both affected by clini- form nucleus of PSP cases. Furthermore, the H1/H1
cally atypical PSP. Both cases had an age of disease genotype does not influence age at disease onset,
onset below the age of 40, a history of cocaine-abuse severity or survival of patients with PSP. Recently,
asymmetric parkinsonism and reduced saccadic speed, the Saitohin gene (STH) Q7R polymorphism, nested
whereas neither had an ophthalmoparesis. In 1 of the 2 within intron 9 of MAPT, has been shown to be in com-
cases, a homozygous deletion at codon 296 (delN296) plete linkage disequilibrium with the extended H1/H2
was identified, lying within the sequence corresponding haplotype (de Silva et al., 2003; Ezquerra et al., 2004).
to the second tubulin repeat of tau protein. The clinical This implicates the Q allele of this non-silent STH poly-
phenotype of these siblings closely resembled that morphism as a potentially important candidate patho-
described in a familial tauopathy with a N279K muta- genic variant in PSP. STH codes for a protein of
tion, where cases developed parkinsonism, supranuc- unknown homologies and function that may play an
lear gaze paresis and dementia in their fifth decade important role in tau regulation.
(Delisle et al., 1999). The nosology of these familial Further studies using 19 single nucleotide poly-
PSP mutations remains a matter of debate and at this morphisms (SNPs) have identified specific subha-
point it may be better to classify them as familial tauo- plotypes in 17q21 that modify risk for PSP and CBD
pathies. It is likely that further sporadic cases clinically (Pastor et al., 2004). A subhaplotype, H10 A, was
resembling young-onset PSP will be found to have tau present in 16% of PSP patients but not observed in
340 D. J. BURN AND A. J. LEES
controls, whereas the H2E0 A subhaplotype was rarely walking aids, such as weighted walkers and exercises
present in the disease group, suggesting that it might (Sosner et al., 1993). Severe postural instability, coupled
have a protective role. Again, using SNPs, linkage dis- with the inability of the patient to recognize the balance
equilibrium in the regions flanking MAPT was mapped problem because of frontal lobe dysfunction, may mean
to establish the maximum extent of the haplotype that a wheelchair is the safest option when falling
block on chromosome 17q21.31 as a region covering becomes a frequent occurrence. Expert nursing support
approximately 2 Mb (Pittman et al., 2004). The entire and lay associations are invaluable for carers and
fully extended H1 haplotype was associated with PSP, families. Such support can also provide important advice
implicating several other genes in addition to MAPT as with regard to future care planning, legal options of
candidate pathogenic loci. power of attorney, trusteeship, advance directives and
Additional intriguing findings are of significant asso- consideration for brain donation (Rohs, 1996).
ciations between the A0 polymorphism of tau and both Table 47.7, modified from a review by Litvan
PD and CBD and between the extended tau gene haplo- (2001), summarizes palliative treatments that may be
type H1 and CBD and clinically defined non-demented considered for PSP.
PD cases (Houlden et al., 2001; Maraganore et al.,
2001). For PD, it has been postulated that the H1 haplo-
47.9.2. Neurotransmitter replacement strategies
type might interact with a-synuclein, thereby influencing
the propensity of a-synuclein to aggregate. This would
No drug tested to date has had a major symptomatic ben-
imply potential pathogenic synergism between synuclei-
efit in PSP. Minor benefits could have been missed, since
nopathies and tauopathies. A study of pathologically
small trial sizes may have introduced a type II error.
confirmed PD cases did not, however, confirm any asso-
ciation with the H1 haplotype (de Silva et al., 2002).
However, in the genetically more homogeneous Norwe-
gian population, Farrer and colleagues (2002) showed a Table 47.7
significant association of tau with sporadic PD. At present,
the association of MAPT with PD therefore remains Palliative treatments for progressive supranuclear palsy
controversial and requires further study. (PSP)
To date, no linkage has been found between PSP and
Feature Palliative approach
the candidate genes for PD synuclein, synphilin or par-
kin, nor the ApoE4 allele, a risk factor for late-onset Alz- Gait instability Weighted walkers; physiotherapy
heimers disease. In addition, no association with Dysphagia Thickeners; dietician, speech and
polymorphisms in the CYP2D6 (which encodes for deb- language input; percutaneous
risoquine 4-hydroxylase cytochrome P450), CYP1A1, endoscopic gastrostomy
N-acetyltransferase 2, dopamine transporter and glu- Dysarthria Speech therapy; communication
tathione S-transferase M1 genes has been found for PSP. aids
Decreased rate of eye Artificial tears (to avoid
blink exposure keratitis); dark
47.9. Management glasses (to reduce photophobia)
Vertical Prism glasses; talking books
47.9.1. General considerations ophthalmoplegia
Blepharospasm and Eyelid crutches; botulinum toxin
A multidisciplinary approach is essential for the man- apraxia of eyelid
agement of PSP. Speech and language therapists and opening
dieticians can advise to improve dysphagia and com- Depression Antidepressants; supportive
munication difficulties, although severe dysphagia therapy
may warrant insertion of a percutaneous gastrostomy Emotional incontinence Tricyclic antidepressants
feeding system. Mirror prism spectacles can make it Drooling Anticholinergics (topical or oral)
use cautiously
possible for patients with severe down-gaze limitation
Patient and family Social services; two lay
to read and feed themselves. Eyelid crutches, sometimes support associations:
combined with botulinum toxin therapy, may be useful PSP (Europe) Association
for apraxia of eyelid opening (Krack and Marion, (http://www.pspeur.org)
1994). Botulinum toxin injections may also be helpful Society for PSP
in improving blepharospasm, retrocollis and orofacial (http://www.psp.org)
dystonia (Polo and Jabbari, 1994; Piccione et al., 1997;
Muller et al., 2002). A physiotherapist can advise on Modified from Litvan (2001).
PROGRESSIVE SUPRANUCLEAR PALSY 341
The response to a variety of medications, usually admi- 47.9.2.2. Cholinergic agents
nistered in a non-systematic and unblinded fashion and PSP is characterized by a loss of cholinergic neurons,
without the use of standardized assessment scales has particularly in the striatum, thalamus, pedunculopon-
been documented (Jackson et al., 1983; Jankovic, 1984; tine nucleus and nucleus basalis of Meynert. Choliner-
Maher and Lees, 1986; Nieforth and Golbe, 1993; gic blockade using scopolamine worsens memory and
Kompoliti et al., 1998; Birdi et al., 2002). gait in PSP (Litvan et al., 1994). Cholinomimetic
drugs have thus been explored for their therapeutic
47.9.2.1. Dopaminergic agents potential in PSP.
Physostigmine, a centrally acting cholinesterase
Intravenous levodopa fails to elicit the increases in
inhibitor, improved long-term memory and visuo-
cerebral blood flow in PSP observed in PD (Kobari
spatial function in 8 PSP patients in a double-blind,
et al., 1992), whereas PSP patients rarely show a clini-
placebo-controlled cross-over study, but no benefits
cally significant response to apomorphine testing
were reported for extraocular, parkinsonian or pseudo-
(DCosta et al., 1991; Bonuccelli et al., 1992). Thera-
bulbar features (Litvan et al., 1989; Kertzman et al.,
peutic trials of levodopa noted benefit in 51%
1990). The same drug had no effect upon swallow-
(Jankovic, 1984), 54% (Golbe et al., 1990) and 38%
ing or oral motor function in a 10-day cross-over
(Nieforth and Golbe, 1993) of patients. In a more
placebo-controlled double-blind trial (Frattali et al.,
recent study, only 4 out of 12 postmortem confirmed
1999). Direct stimulation of postsynaptic cholinergic
PSP cases showed a modest improvement whilst
receptors with the non-selective M1 and M2 muscari-
receiving levodopa that was not sustained (Kompoliti
nic agonist, RS-86, did not produce any beneficial
et al., 1998). Adverse effects occurred in over half of
effects in motor function, eye movements or cognitive
the patients. Birdi and colleagues (2002) reported that
performance in 10 PSP patients in a 9-week double-
in 15 of their postmortem-confirmed patients receiving
blind, placebo-controlled cross-over trial, despite elec-
levodopa either alone or in combination with an anti-
troencephalographic evidence of enhanced central
cholinergic, dopamine agonist, selegiline or amanta-
cholinergic activity upon sleep architecture (Foster
dine, 9 had shown some benefit. Consistent with
et al., 1989).
the recent observations by Williams and colleagues
The effects of the cholinesterase inhibitor donepezil
(2004), none of the 7 cases with supranuclear ophthal-
upon cognitive function, motor skills and activities of
moplegia responded to levodopa, whereas 5 of 8
daily living (ADL) were assessed in 6 PSP patients
patients without ophthalmoplegia improved (although
at baseline and after 3 months of treatment. There
the magnitude and duration of the improvement were
was no change from baseline in any of the parameters
not documented).
measured (Fabbrini et al., 2001). Litvan used the same
Bromocriptine is generally ineffective in doses up
dose of donepezil (10 mg) in 21 patients in a double-
to 70 mg/day (Williams et al., 1979). A controlled
blind, placebo-controlled, randomized, cross-over trial
study of lisuride, a relatively selective dopamine D2-
(Litvan et al., 2001). There was a modest improvement
receptor agonist, in daily doses of up to 5 mg in 7
in one of the memory tests but a significant worsening
patients did not yield any positive results whereas 3
in motor and ADL scores.
patients developed hallucinations (Neophytides et al.,
1982). Pergolide at a daily dose of 4 mg improved par-
47.9.2.3. Adrenergic agents
kinsonian and/or pseudobulbar signs in 2 out of 3 PSP
patients (Jackson et al., 1983). Pramipexole, a non- A placebo-controlled, double-blind, cross-over study
ergot dopamine agonist with high affinity for the dopa- of idazoxan in 9 PSP patients showed a significant
mine D3-receptor, failed to show any benefit in 6 PSP decrease in the Unified Parkinsons Disease Rating
patients, in a daily dose of up to 4.5 mg (Weiner et al., Scale (UPDRS), corresponding to an improvement in
1999). Two patients reported visual hallucinations and mobility, balance and dexterity (Ghika et al., 1991).
1 a worsening of motor symptoms. In contrast, the potent a2-antagonist efaroxan did not
Selegiline, a monoamine oxidase type B inhibitor, significantly improve motor impairments in 14 PSP
has not shown any significant therapeutic effect in patients using a double-blind, placebo-controlled,
PSP (Golbe et al., 1990). Amantadine, a drug with cross-over study design (Rascol et al., 1998).
multiple actions, including increasing the synthesis,
release and reuptake of dopamine, as well as potent 47.9.2.4. Other drugs
N-methyl-D-aspartate antagonist properties, can pro- Nortriptyline may be useful for the management of
vide a transient therapeutic benefit in up to 15% of depression in PSP (Tamai and Almeida, 1997). Ami-
cases (Litvan and Chase, 1992). triptyline has been produced with variable results.
342 D. J. BURN AND A. J. LEES
Three of 4 patients showed moderate to definite was associated with a number of transient postopera-
improvement in one study, whereas postural instability tive complications (Koller et al., 1989; Ward-Smith
worsened in the fourth (Newman, 1985). A retrospec- and Berry, 1990). A small number of PSP patients
tive analysis of 28 PSP patients, treated with doses of have also undergone pallidotomy without significant
amitriptyline ranging from 50 to 200 mg, indicated benefit (Litvan, 2001).
an improvement in gait or dysphagia in 9 patients,
but moderate to severe side-effects, particularly antic- 47.9.4. Future directions
holinergic-related, occurred in 14. Other case reports
have demonstrated moderate improvement in motor More radical advances in PSP therapeutics will come
scores at low doses (2080 mg/day), with cognitive from a greater understanding of the pathophysiology
and behavioral problems only emerging at higher of the condition and the development of animal mod-
doses (Engel, 1996). els in which to test new disease-modifying treatments
A double-blind, placebo-controlled, cross-over (Golbe, 2000). Regarding the latter, transgenic mice,
study of the GABAergic drug zolpidem in 10 PSP Drosophila and zebrafish models have biological simi-
patients revealed significant improvements in motor larities with a number of tauopathies, including PSP
function and saccadic eye movements (Daniele et al., (Lewis et al., 2000; Wittman et al., 2001; Tomasiewicz
1999). Adverse effects were drowsiness and increased et al., 2002). One example of the use of such models
postural instability. A further case report highlighted a comes from a report of the inhibition of glycogen
non-sustained improvement in gaze palsy and parkin- synthase 3 kinase (GSK-3b), an enzyme believed to
sonism in a patient taking 5 mg zolpidem at night, with be important in tau phosphorylation, leading to func-
benefit lasting 4 weeks (Mayr et al., 2002). tional improvement in transgenic Drosophila (Mudher
Methysergide was associated with clinical impro- et al., 2004). Lithium is an inhibitor of GSK-3b and
vement in 1 of 3 PSP patients and with deterioration trials are already planned for Alzheimers disease
of symptoms in another (Kompoliti et al., 1998). One (Mudher et al., 2004), although preliminary studies
of 2 patients experienced modest improvement with using human tissue have suggested that GSK-3b levels
5-hydroxytryptophan. Fluoxetine, mianserin and biper- are unchanged in PSP, compared with age-matched
iden hydrochloride have all been used in single controls (Borghi et al., 2004).
patients with no observed improvement (Kompoliti Other approaches to disease modification in PSP
et al., 1998). could involve the manipulation of splicing regulation
by RNA stem-loops, with the aim of reducing four-
47.9.3. Non-pharmacological interventions repeat tau production (Golbe, 2000). This could be
achieved, for example, by the use of RNA interference
Barclay et al. (1996) reported 5 patients with PSP who to inhibit the translation of E10 messenger RNA
each received 9 electroconvulsive therapy (ECT) treat- (Litvan, 2001; Davidson and Paulson, 2004).
ments, over 3 weeks. One patient showed a dramatic A number of posttranslational processes may be
improvement in mobility, although no long-term data involved in the aggregation of tau in PSP, but glyca-
were available, whereas 2 experienced a mild benefit tion and transglutamination have been principally
and a further 2 did not change. There were transient implicated (see above). Targeted inhibition of TGase
side-effects in all patients, including confusion and might be capable of reducing tau aggregation. The for-
bulbar dysfunction. An apomorphine challenge carried mation of AGE-tau is also associated with the genera-
out pre- and post-ECT did not show any change in tion of oxygen free radicals and the induction of
motor improvement, suggesting that ECT did not oxidative stress. Phosphorylation of tau in PSP may
induce dopamine receptor sensitization. ECT may be a direct consequence of oxidative stress-induced
improve severe depression in PSP without beneficial activation of mitogen-activated protein kinases,
or deleterious effects on motor function (Netzel and including the p38 pathway, thus opening up further
Sutor, 2001), although adverse effects of ECT on therapeutic potential through manipulation of this
motor function have also been reported (Hauser and pathway (Hartzler et al., 2002).
Trehan, 1994). Microglial activation is greater in PSP than in con-
Other non-drug treatments include transcranial trol brains and microglial activation correlates with tau
alternate current pulsed electromagnetic fields, which burden in most areas. The use of anti-inflammatory
apparently reduced the frequency of freezing by 50% agents could therefore be considered, although from
and falls by 90% in 1 patient (Sandyk, 1998). Auto- trials of these drugs in Alzheimers disease and the
logous adrenal medullary transplantation into the cau- size of effect, the feasibility of similar studies for
date nucleus of 3 PSP patients had limited efficacy and PSP must be questionable. Finally, the use of trophic
PROGRESSIVE SUPRANUCLEAR PALSY 343
factors could be a promising therapeutic approach for Baker M, Litvan I, Houlden H et al. (1999). Association of
PSP, although there would be considerable technical an extended haplotype in the tau gene with progressive
issues to overcome (Litvan, 2001). supranuclear palsy. Hum Mol Genet 8: 711715.
Barclay CL, Lang AE (1997). Dystonia in progressive supra-
47.10. Conclusions nuclear palsy. J Neurol Neurosurg Psychiatry 62:
352356.
Despite greater awareness in recent years, many patients Barclay CL, Duff J, Sandor P et al. (1996). Limited useful-
ness of electroconvulsive therapy in progressive supranuc-
with PSP remain undiagnosed or misdiagnosed for much
lear palsy. Neurology 46: 12841286.
of their disease duration. The role of tau protein in the
Birdi S, Rajput AH, Fenton M et al. (2002). Progressive
pathophysiological process, together with the establish- supranuclear palsy diagnosis and confounding features:
ment of a modest genetic predisposition has stimul- report on 16 autopsied cases. Mov Disord 17: 12551264.
ated research. At the same time, studies on a cluster of Blin J, Baron JC, Dubois B et al. (1990). Positron emission
a PSP-like condition in Guadeloupe have produced new tomography study in progressive supranuclear palsy: brain
clues for potential environmental toxins. If the funda- hypometabolic pattern and clinicometabolic correlations.
mental pathophysiological process in PSP turns out to Arch Neurol 47: 747752.
be an overproduction of four-repeat tau, it will be crucial Boeve BF, Silber MH, Parisi JE et al. (2003). Synucleinopa-
to determine how the function of the RNA stem-loop thy pathology and REM sleep behavior disorder plus
structure, a key regulator in the alternate splicing process, dementia or parkinsonism. Neurology 61: 4045.
Bonuccelli U, Piccini P, Del Dotto P et al. (1992). Apomor-
may be affected by various genetic influences and toxins.
phine test in de novo Parkinsons disease. Funct Neurol 7:
As disease-modifying therapies emerge it will be
295298.
vital to intervene as early in the pathological process Borghi R, Piccini A, Delacourte A et al. (2004). Protein
as possible. There is therefore a need for a greater levels of glycogen synthase 3 kinase are normal in pro-
awareness of PSP and the development of robust diag- gressive supranuclear palsy. Neurosci Lett 366: 6770.
nostic clinical and investigational markers that predict Bower JH, Maraganore DM, McDonnell SK et al.
whether an individual with suspicious, but not typical (1997). Incidence of progressive supranuclear palsy and
classical features, will go on to develop PSP. Physi- multiple system atrophy in Olmsted County, Minnesota,
cians should consider PSP when rigidity and bradyki- 1976 to 1990. Neurology 49: 12841288.
nesia coexist with early falls. The patient should be Brenneis C, Seppi K, Schocke M et al. (2004). Voxel based
examined carefully for slowing of downward saccadic morphometry reveals a distinct pattern of frontal atrophy
in progressive supranuclear palsy. J Neurol Neurosurg
eye movements and the presence of square-wave jerks.
Psychiatry 75: 246249.
The presence of frank vertical ophthalmoparesis is of
Brodsky H, Dat Vuong K, Thomas M et al. (2004). Glabellar
significant diagnostic help but it should also be borne and palmomental reflexes in Parkinsonian disorders. Neu-
in mind that this physical sign might take several years rology 63: 10961098.
to develop in some patients. Brotini S, Gigli GL (2004). Epidemiology and clinical fea-
tures of sleep disorders in extrapyramidal disease. Sleep
References Med 5: 169179.
Brusa A, Stoehr R, Pramstaller PP (2004). Progressive supra-
Aarsland D, Litvan I, Salmon D et al. (2003). Performance nuclear palsy: new disease or variant of postencephalitic
on the dementia rating scale in Parkinsons disease with parkinsonism? Mov Disord 19: 247252.
dementia and dementia with Lewy bodies: comparison Burn DJ, Rinne JO, Quinn NP et al. (1995). Striatal opioid
with progressive supranuclear palsy and Alzheimers dis- receptor binding in Parkinsons disease, striatonigral
ease. J Neurol Neurosurg Psychiatry 74: 12151220. degeneration and Steele-Richardson-Olszewski syndrome.
Albers DS, Augood SJ (2001). New insights into progressive A [11C]diprenorphine PET study. Brain 118: 951958.
supranuclear palsy. Trends Neurosci 24: 347353. Campdelacreu J, Kumru H, Tolosa E et al. (2004). Progres-
Arai T, Ikeda K, Akiyama H et al. (2004). Identification of sive supranuclear palsy syndrome induced by clebopride.
amino-terminally cleaved tau fragments that distinguish Mov Disord 19: 482484.
progressive supranuclear palsy from corticobasal degen- Caparros-Lefebvre D, Elbaz A (1999). Caribbean Parkinson-
eration. Ann Neurol 55: 7279. ism Study Group. Possible relation of atypical parkinson-
Averbuch-Heller L, Paulson GW, Daroff RB et al. ism in the French West Indies with consumption of
(1999). Whipples disease mimicking progressive supra- tropical plants: a case-control study. Lancet 354:
nuclear palsy: the diagnostic value of eye movement 281286.
recording. J Neurol Neurosurg Psychiatry 66: 532535. Caparros-Lefebvre D, Sargent N, Lees AJ et al.
Baker KB, Montgomery EB (2001). Performance on the (2002). Guadeloupean parkinsonism: a cluster of progres-
PD test battery by relatives of patients with progressive sive supranuclear palsy-like tauopathy. Brain 125:
supranuclear palsy. Neurology 56: 2530. 801811.
344 D. J. BURN AND A. J. LEES

Chio A, Magnani C, Schiffer D (1998). Prevalence of Engel PA (1996). Treatment of progressive supranuclear
Parkinsons disease in Northwestern Italy: comparison of palsy with amitriptyline: therapeutic and toxic effects.
tracer methodology and clinical ascertainment of cases. J Am Geriatr Soc 44: 10721074.
Mov Disord 13: 400405. Ezquerra M, Campdelacreu J, Munoz E, Oliva R et al.
Clarke CE, Lowry M (2001). Systematic review of proton (2004). Sequence analysis of tau 30 untranslated region
magnetic resonance spectroscopy of the striatum in par- and saitohin gene in sporadic progressive supranuclear
kinsonian syndromes. Eur J Neurol 8: 573577. palsy. J Neurol Neurosurg Psychiatry 75: 155157.
Collins SJ, Ahlskog JE, Parisi JE et al. (1995). Progressive Fabbrini G, Barbanti P, Bonifati V et al. (2001). Donepezil in
supranuclear palsy: neuropathologically based diagnostic the treatment of progressive supranuclear palsy. Acta Neu-
clinical criteria. J Neurol Neurosurg Psychiatry 58: rol Scand 103: 123125.
167173. Farrer M, Skipper L, Berg M et al. (2002). The tau H1 hap-
Conrad C, Andreadis A, Trojanowski JQ et al. (1997). Gene- lotype is associated with Parkinsons disease in the Nor-
tic evidence for the involvement of tau in progressive wegian population. Neurosci Lett 322: 8386.
supranuclear palsy. Ann Neurol 41: 277281. Foster NL, Aldrich MS, Bluemlein L et al. (1989). Failure of
Daniele A, Moro E, Bentivoglio AR (1999). Zolpidem in cholinergic agonist RS-86 to improve cognition and move-
progressive supranuclear palsy. N Engl J Med 341: ment in PSP despite effects on sleep. Neurology 39:
543544. 257261.
Davidson BL, Paulson HL (2004). Molecular medicine for Foster NL, Minoshima S, Johanns J et al. (2000). PET mea-
the brain: silencing of disease genes with RNA interfer- sures of benzodiazepine receptors in progressive supranuc-
ence. Lancet Neurol 3: 145149. lear palsy. Neurology 54: 17681773.
Davis PH, Bergeron C, McLachlan DR (1985). Atypical pre- Frasca J, Blumbergs PC, Henschke P et al. (1991). A clinical
sentation of progressive supranuclear palsy. Ann Neurol and pathological study of progressive supranuclear palsy.
17: 337343. Clin Exp Neurol 28: 7989.
DCosta DF, Abbott RJ, Pye IF et al. (1991). The apomor- Frattali CM, Sonies BC, Chi-Fishman G et al. (1999). Effects
phine test in Parkinsonian syndromes. J Neurol Neurosurg of physostigmine on swallowing and oral motor functions
Psychiatry 54: 870872. in patients with progressive supranuclear palsy: a pilot
De Bruin VM, Lees AJ (1994). Subcortical neurofibrillary study. Dysphagia 14: 165168.
degeneration presenting as Steele-Richardson-Olszewski Garbutt S, Riley DE, Kumar AN et al. (2004). Abnormalities
and other related syndromes: a review of 90 pathologically of optokinetic nystagmus in progressive supranuclear
verified cases. Mov Disord 9: 381389. palsy. J Neurol Neurosurg Psychiatry 75: 13861394.
de Rijk MC, Breteler MMB, Graveland GA et al. (1995). Geddes JF, Hughes AJ, Lees AJ et al. (1993). Pathological
Prevalence of Parkinsons disease in the elderly: the overlap in cases of parkinsonism associated with neurofi-
Rotterdam study. Neurology 45: 21432146. brillary tangles. A study of recent cases of postencephalitic
de Silva R, Hardy J, Crook J et al. (2002). The tau locus parkinsonism and comparison with progressive supra-
is not significantly associated with pathologically con- nuclear palsy and Guamanian parkinsonismdementia
firmed sporadic Parkinsons disease. Neurosci Lett 330: complex. Brain 116: 281302.
201203. Gerhard A, Banati RB, Cagnin A et al. (2001). In vivo ima-
de Silva R, Hope A, Pittman A et al. (2003). Strong associa- ging of activated microglia with [11C] PK11195 positron
tion of the Saitohin gene Q7 variant with progressive emission tomography (PET) in idiopathic and atypical
supranuclear palsy. Neurology 61: 407409. Parkinsons disease. Neurology 56: A270.
Delisle MB, Murrell JR, Richardson R et al. (1999). A muta- Ghika J, Tennis M, Hoffman E et al. (1991). Idazoxan treat-
tion at codon 279 (N279K) in exon 10 of the Tau gene ment in progressive supranuclear palsy. Neurology 41:
causes a tauopathy with dementia and supranuclear palsy. 986991.
Acta Neuropathol (Berl) 98: 6277. Gibb GM, de Silva R, Revesz T et al. (2004). Differential
Dubois B, Slachevsky A, Litvan I et al. (2000). The FAB: involvement and heterogeneous phosphorylation of tau
a frontal assessment battery. Neurology 55: 16211626. isoforms in progressive supranuclear palsy. Brain Res
Duvoisin RC (1992). Clinical diagnosis. In: I Litvan, Mol Brain Res 121: 95101.
Y Agid (Eds.), Progressive Supranuclear Palsy: Clin- Gironell A, Kulisevsky J, Roig C et al. (2003). Diagnostic
ical and Research Approaches.Oxford University potential of acoustic startle reflex, acoustic blink reflex,
Press, New York, pp. 1533. and electro-oculography in progressive supranuclear
Eckert T, Sailer M, Kaufmann J et al. (2004). Differentiation palsy: a prospective study. Mov Disord 18: 12731279.
of idiopathic Parkinsons disease, multiple system atrophy, Goetz CG (1996). An early photographic case of prob-
progressive supranuclear palsy, and healthy controls using able progressive supranuclear palsy. Mov Disord 11:
magnetization transfer imaging. Neuroimage 21: 229235. 617618.
Eidelberg D, Dhawan V (2002). Can imaging distinguish Goetz CG, Leurgans S, Lang AE et al. (2003). Progression of
PSP from other neurodegenerative disorders? Neurology gait, speech and swallowing deficits in progressive supra-
58: 997998. nuclear palsy. Neurology 60: 917922.
PROGRESSIVE SUPRANUCLEAR PALSY 345

Golbe LI (1992). Epidemiology. In: I Litvan, Y Agid (Eds.), Houlden H, Baker M, Morris HR et al. (2001). Corticobasal
Progressive Supranuclear Palsy: Clinical and Research degeneration and progressive supranuclear palsy share a
Approaches.Oxford University Press, Oxford, pp. 3443. common tau haplotype. Neurology 56: 17021706.
Golbe LI (1993). Progressive supranuclear palsy. In: J Jankovic, Hughes AJ, Daniel SE, Ben-Shlomo Y et al. (2002). The
E Tolosa (Eds.), Parkinsons Disease and Move- accuracy of diagnosis of parkinsonian syndromes in a spe-
ment Disorders.Williams & Wilkins, Baltimore, pp. cialist movement disorder service. Brain 125: 861870.
145161. Hutton M (2000). Missing tau mutation identified. Ann
Golbe LI (2000). Progressive supranuclear palsy in the mole- Neurol 47: 417418.
cular age. Lancet 356: 870871. Jackson JA, Jankovic J, Ford J (1983). Progressive supranuc-
Golbe LI, Davis PH, Schoenberg BS et al. (1988). Prevalence lear palsy: clinical features and response to treatment in
and natural history of progressive supranuclear palsy. sixteen patients. Ann Neurol 13: 273278.
Neurology 38: 10311034. Jankovic J (1984). Progressive supranuclear palsy: clinical
Golbe LI, Sage JI, Duvoisin RC (1990). Drug treatment of 83 and pharmacological update. Neurol Clin 2: 473486.
patients with progressive supranuclear palsy. Neurology Jellinger KA, Blancher C (1992). Neuropathology. In: I Lit-
40: 438. van, Y Agid (Eds.), Progressive Supranuclear Palsy: Clin-
Goldbaum O, Oppermann M, Handschuh M et al. (2003). ical and Research Approaches.Oxford University Press,
Proteasome inhibition stabilizes tau inclusions in oligo- Oxford, pp. 4488.
dendroglial cells that occur after treatment with okadaic Josephs KA, Dickson DW (2003). Diagnostic accuracy of
acid. J Neurosci 23: 88728880. progressive supranuclear palsy in the Society for Progres-
Grafman J, Litvan I, Stark M (1995). Neuropsychological sive Supranuclear Palsy brain bank. Mov Disord 18:
features of progressive supranuclear palsy. Brain Cogn 10181026.
28: 311320. Josephs KA, Tsuboi Y, Dickson DW (2004). Creutzfeldt-
Groschel K, Hauser TK, Luft A et al. (2004). Magnetic reso- Jakob disease presenting as progressive supranuclear
nance imaging-based volumetry differentiates progressive palsy. Eur J Neurol 11: 343346.
supranuclear palsy from corticobasal degeneration. Neuro- Juh R, Kim J, Moon D et al. (2004). Different metabolic pat-
image 21: 714724. terns analysis of Parkinsonism on the 18F-FDG PET. Eur
Hartzler AW, Zhu X, Siedlak SL et al. (2002). The p38 pathway J Radiol 51: 223233.
is activated in Pick disease and progressive supranuclear Katzenschlager R, Cardozo A, Cobo MRA et al.
palsy: a mechanistic link between mitogenic pathways, oxi- (2003). Unclassifiable parkinsonism in two European ter-
dative stress, and tau. Neurobiol Aging 23: 855859. tiary referral centres for movement disorders. Mov Disord
Hauser RA, Trehan R (1994). Initial experience with electro- 18: 11231131.
convulsive therapy for progressive supranuclear palsy. Kawashima M, Miyake M, Kusumi M et al. (2004). Preva-
Mov Disord 9: 467469. lence of progressive supranuclear palsy in Yonago, Japan.
Hauw JJ, Daniel SE, Dickson D et al. (1994). Preliminary Mov Disord 19: 12391240.
NINDS neuropathologic criteria for Steele-Richardson- Kertzman C, Robinson DL, Litvan I (1990). Effects of phy-
Olszewski syndrome (progressive supranuclear palsy). sostigmine on spatial attention in patients with progressive
Neurology 44: 20152019. supranuclear palsy. Arch Neurol 47: 13461350.
Holmberg B, Rosengren L, Karlsson JE et al. (1998). Kim YJ, Ichise M, Ballinger JR et al. (2002). Combination of
Increased cerebrospinal fluid levels of neurofilament pro- dopamine transporter and D2 receptor SPECT in the diag-
tein in progressive supranuclear palsy and multiple-system nostic evaluation of PD, MSA, and PSP. Mov Disord 17:
atrophy compared with Parkinsons disease. Mov Disord 303312.
13: 7077. Kimber J, Mathias CJ, Lees AJ et al. (2000). Physiological,
Holmberg B, Johnels B, Ingvarsson P et al. (2001). CSF- pharmacological and neurohormonal assessment of auto-
neurofilament and levodopa tests combined with discrimi- nomic function in progressive supranuclear palsy. Brain
nant analysis may contribute to the differential diagnosis 123: 14221430.
of Parkinsonian syndromes. Parkinsonism Relat Disord Kobari M, Fukuuchi Y, Shinohara T et al. (1992). Local cer-
8: 2331. ebral blood flow and its response to intravenous levodopa
Holmberg B, Johnels B, Blennow K et al. (2003). Cerebrosp- in progressive supranuclear palsy. Comparison with Par-
inal fluid Abeta42 is reduced in multiple system atrophy kinsons disease. Arch Neurol 49: 725730.
but normal in Parkinsons disease and progressive supra- Koller WC, Morantz R, Vetere-Overfield B et al. (1989).
nuclear palsy. Mov Disord 18: 186190. Autologous adrenal medullary transplant in progressive
Hope AD, Lashley T, Lees AJ et al. (2004). Failure in heat- supranuclear palsy. Neurology 39: 10661068.
shock protein expression in response to UBB1 protein in Kompoliti K, Goetz CG, Litvan I et al. (1998). Pharmacologi-
progressive supranuclear palsy in humans. Neurosci Lett cal therapy in progressive supranuclear palsy. Arch Neurol
359: 9498. 55: 10991102.
Horiguchi T, Uryu K, Giasson BI et al. (2003). Nitration of Kotake Y, Okuda K, Kamizono M et al. (2004). Detection
tau protein is linked to degeneration in tauopathies. Am and determination of reticuline and N-methylcoculaurine
J Pathol 163: 10211031. in the Annonaceae family using liquid chromatography-
346 D. J. BURN AND A. J. LEES

tandem mass spectrometry. J Chromatogr B Analyt Tech- related disorders? A clinicopathological study. Brain
nol Biomed Life Sci 806: 7578. 120: 6574.
Krack P, Marion MH (1994). Apraxia of lid opening, a Litvan I, Phipps M, Pharr VL et al. (2001). Randomized pla-
focal eyelid dystonia: clinical study of 32 patients. Mov cebo-controlled trial of donepezil in patients with progres-
Disord 9: 610615. sive supranuclear palsy. Neurology 57: 467473.
Kristensen MO (1985). Progressive supranuclear palsy20 Litvan I, Bhatia KP, Burn DJ et al. (2003). SIC task force
years later. Acta Neurol Scand 71: 177189. appraisal of clinical diagnostic criteria for parkinsonian
Kuhn AA, Grosse P, Holtz K et al. (2004). Patterns of abnor- disorders. Mov Disord 18: 467486.
mal motor cortex excitability in atypical parkinsonian syn- Lopez OL, Litvan I, Catt KE et al. (1999). Accuracy of four
dromes. Clin Neurophysiol 115: 17861795. clinical diagnostic criteria for the diagnosis of neurode-
Lange KW, Tucha O, Alders GL et al. (2003). Differentiation generative dementias. Neurology 53: 12921299.
of parkinsonian syndromes according to differences in Maher ER, Lees AJ (1986). The clinical features and natural
executive functions. J Neural Transm 110: 983995. history of the Steele-Richardson-Olszewski syndrome
Lannuzel A, Michel PP, Caparros-Lefebvre D et al. (progressive supranuclear palsy). Neurology 36:
(2002). Toxicity of Annonaceae for dopaminergic neu- 10051008.
rons: potential role in atypical parkinsonism in Guade- Maraganore DM, Anderson DW, Bower JH et al.
loupe. Mov Disord 17: 8490. (1998). Potential selection bias in autopsy series of Par-
Larner AJ (2002). Did Charles Dickens describe progres- kinsons disease and related disorders. Neurology 50:
sive supranuclear palsy in 1857? Mov Disord 17: A98A99.
832833. Maraganore DM, Hernandez DG, Singleton AB et al.
Lees AJ (1987). The Steele-Richardson-Olszewski syndrome (2001). Case-control study of the extended tau gene
(progressive supranuclear palsy). In: CD Marsden, S Fahn haplotype in Parkinsons disease. Ann Neurol 50:
(Eds.), Movement Disorders 2, vol. 7. Butterworths, Lon- 658661.
don, 272287. Mastaglia FL, Grainger K, Kee F et al. (1973). Progressive
Lepore FE, Moudgil SS (2002). Procerus sign in progressive supranuclear palsy (the Steele-Richardson-Olszewski syn-
supranuclear palsy. Neurology 58: 18661867. drome): clinical and electrophysiological observations in
Lewis J, McGowan E, Rockwood J et al. (2000). Neurofibril- eleven cases. Proc Aust Assoc Neurol 10: 3544.
lary tangles, amyotrophy and progressive motor distur- Masucci EF, Kurtzke JF (1989). Tremor in progressive
bance in mice expressing mutant (P301L) tau protein. supranuclear palsy. Acta Neurol Scand 80: 296300.
Nat Genet 25: 402405. Matsuo H, Takashima H, Kishikawa M et al. (1991). Pure
Litvan I (2001). Diagnosis and management of progressive akinesia: an atypical manifestation of progressive supra-
supranuclear palsy. Semin Neurol 21: 4148. nuclear palsy. J Neurol Neurosurg Psychiatry 54:
Litvan I, Chase TN (1992). Traditional and experimental 397400.
therapeutic approaches. In: I Litvan, (Ed.), Progressive Mayr BJ, Bonelli RM, Niederwieser G et al. (2002). Zolpi-
Supranuclear Palsy: Clinical and Research Approaches. dem in progressive supranuclear palsy. Eur J Neurol 9:
Oxford University Press, New York, pp. 254269. 184185.
Litvan I, Gomez C, Atack JR et al. (1989). Physostigmine Millar D, Griffiths P, Zermansky AJ et al. (2006). Character-
treatment of progressive supranuclear palsy. Ann Neurol izing behavioral and cognitive dysexecutive changes in
26: 404407. progressive supranuclear palsy. Mov Disord 21: 199207.
Litvan I, Blesa R, Clark K et al. (1994). Pharmacological Mishina M, Ishii K, Mitani K et al. (2004). Midbrain hypo-
evaluation of the cholinergic system in progressive supra- metabolism as early diagnostic sign for progressive supra-
nuclear palsy. Ann Neurol 36: 5561. nuclear palsy. Acta Neurol Scand 110: 128135.
Litvan I, Agid Y, Calne D et al. (1996a). Clinical research Mokri B, Ahlskog JE, Fulgham JR et al. (2004). Syndrome
criteria for the diagnosis of progressive supranuclear palsy resembling PSP after surgical repair of ascending aorta
(Steele-Richardson-Olszewski syndrome): report of the dissection or aneurysm. Neurology 62: 971973.
NINDS-SPSP international workshop. Neurology 47: 19. Montplaisir J, Petit D, Decary A et al. (1997). Sleep and
Litvan I, Hauw JJ, Bartko JJ et al. (1996b). Validity and quantitative EEG in patients with progressive supranuclear
reliability of the preliminary NINDS neuropathologic cri- palsy. Neurology 49: 9991003.
teria for progressive supranuclear palsy and related disor- Morris HR, Gibb G, Katzenschlager R et al. (2002a). Patho-
ders. J Neuropathol Exp Neurol 55: 97105. logical, clinical and genetic heterogeneity in progressive
Litvan I, Mangone CA, McKee A et al. (1996c). Natural his- supranuclear palsy. Brain 125: 969975.
tory of progressive supranuclear palsy (Steele-Richardson- Morris HR, Katzenschlager R, Janssen JC et al. (2002b).
Olszewski syndrome) and clinical predictors of survival: Sequence analysis of tau in familial and sporadic progres-
a clinicopathological study. J Neurol Neurosurg Psychiatry sive supranuclear palsy. J Neurol Neurosurg Psychiatry
60: 615620. 72: 388390.
Litvan I, Campbell G, Mangone CA et al. (1997). Which Morris HR, Osaki Y, Holton J et al. (2003). Tau exon 1016
clinical features differentiate progressive supranuclear mutation FTDP-17 presenting clinically as sporadic young
palsy (Steele-Richardson-Olszewski syndrome) from onset PSP. Neurology 61: 102104.
PROGRESSIVE SUPRANUCLEAR PALSY 347

Mudher A, Shepherd D, Newman TA, et al. (2004). GSK- Pastor P, Ezquerra M, Perez JC et al. (2004). Novel haplo-
3beta inhibition reverses axonal transport defects and types in 17q21 are associated with progressive supranuc-
behavioural phenotypes in Drosophila. Mol Psychiatry 9: lear palsy. Ann Neurol 56: 249258.
522530. Paviour DC, Lees AJ, Josephs KA et al. (2004). Frontotem-
Muller J, Wenning GK, Wissel J et al. (2002). Botulinum poral lobar degeneration with ubiquitin-only-immunoreac-
toxin treatment in atypical parkinsonian disorders asso- tive neuronal changes: broadening the clinical picture to
ciated with disabling focal dystonia. J Neurol 249: include progressive supranuclear palsy. Brain 127:
300304. 24412451.
Murialdo A, Marchese R, Abbruzzese G et al. (2000). Neu- Pezzella FR, Paglia MG, Colosimo C (2004). Cerebrospinal
rosyphilis presenting as progressive supranuclear palsy. fluid analysis for Whipples disease in patients with pro-
Mov Disord 15: 730731. gressive supranuclear palsy. Mov Disord 19: 220222.
Nath U, Burn DJ (2000). The epidemiology of progressive Pharr V, Litvan I, Brat DJ et al. (1999). Ideomotor apraxia in
supranuclear palsy. Parkinsonism Relat Disord 6: 145153. progressive supranuclear palsy: a case study. Mov Disord
Nath U, Ben-Shlomo Y, Thomson R et al. (2001). The 14: 162166.
prevalence of progressive supranuclear palsy (Steele- Piccini P, de Yebenez J, Lees AJ et al. (2001). Familial pro-
Richardson-Olszewski syndrome) in the UK. Brain 124: gressive supranuclear palsy: detection of subclinical cases
14381449. using 18F-dopa and 18Fluorodeoxyglucose positron emis-
Nath U, Ben-Shlomo Y, Thomson RG et al. (2003). Clinical sion tomography. Arch Neurol 58: 18461851.
features and natural history of progressive supranuclear Piccione F, Mancini E, Tonin P et al. (1997). Botulinum
palsy: a clinical cohort study. Neurology 60: 910916. toxin treatment of apraxia of eyelid opening in progressive
Nath U, Thomson RG, Wood R et al. (2005). Population- supranuclear palsy: report of two cases. Arch Phys Med
based mortality rates and quality of death certification Rehabil 78: 525529.
in progressive supranuclear palsy (Steele-Richardson- Pillon B, Dubois B, Ploska A et al. (1991). Severity and spe-
Olszewski Syndrome). J Neurol Neurosurg Psychiatry cificity of cognitive impairment in Alzheimers, Hunting-
78: 498502. tons, and Parkinsons diseases and progressive
Neophytides A, Lieberman AN, Goldstein M et al. supranuclear palsy. Neurology 41: 634643.
(1982). The use of lisuride, a potent dopamine and seroto- Pirker W, Djamshidian S, Asenbaum S et al. (2002). Progres-
nin agonist, in the treatment of progressive supranuclear sion of dopaminergic degeneration in Parkinsons disease
palsy. J Neurol Neurosurg Psychiatry 45: 261263. and atypical parkinsonism: a longitudinal beta-CIT
Netzel PJ, Sutor B (2001). Electroconvulsive therapy-respon- SPECT study. Mov Disord 17: 4553.
sive depression in a patient with progressive supranuclear Pittman AM, Myers AJ, Duckworth J et al. (2004). The
palsy. J ECT 17: 6870. structure of the tau haplotype in controls and in pro-
Neumann M, Muller V, Gorner K et al. (2004). Pathological gressive supranuclear palsy. Hum Mol Genet 13:
properties of the Parkinsons disease-associated DJ-1 in 12671274.
alpha-synucleinopathies and tauopathies: relevance for Plotkin M, Amthauer H, Klaffke S et al. (2005). Combined
multiple system atrophy and Picks disease. Acta Neuro- (123)I-FP-CIT and (123)I-IBZM SPECT for the diagnosis
pathol (Berl) 107: 489496. of parkinsonian syndromes: study on 72 patients. J Neural
Newman GC (1985). Treatment of progressive supranuclear Transm 112: 677692.
palsy with tricyclic antidepressants. Neurology 35: Polo KB, Jabbari B (1994). Botulinum toxin-A improves the
11891193. rigidity of progressive supranuclear palsy. Ann Neurol 35:
Nieforth KA, Golbe LI (1993). Retrospective study of drug 237239.
response in 87 patients with progressive supranuclear Quinn NP (1997). Parkinsons disease: clinical features. In:
palsy. Clin Neuropharmacol 16: 338346. NP Quinn, (Ed.), Parkinsonism, vol. 6. Balliere Tindall,
Oka M, Katayama S, Imon Y et al. (2001). Abnormal signals London, pp. 113.
on proton density-weighted MRI of the superior cerebellar Radhakrishnan K, Thacker AK, Maloo JC et al. (1988).
peduncle in progressive supranuclear palsy. Acta Neurol Descriptive epidemiology of some rare neurological
Scand 104: 15. diseases in Benghazi, Libya. Neuroepidemiology 7:
Ondo W, Warrior D, Overby A et al. (2000). Computerized 159164.
posturography analysis of progressive supranuclear palsy: Rajput AH, Offord KP, Beard CM et al. (1984). Epidemiol-
a case-control comparison with Parkinsons disease and ogy of parkinsonism: incidence, classification and mortal-
healthy controls. Arch Neurol 57: 14641469. ity. Ann Neurol 16: 278282.
Osaki Y, Ben-Shlomo Y, Lees AJ et al. (2003). Accuracy of Rascol O, Sieradzan K, Peyro-Saint-Paul H et al.
clinical diagnosis of progressive supranuclear palsy. Mov (1998). Efaroxan, an alpha-2 antagonist, in the treatment
Disord 19: 181189. of progressive supranuclear palsy. Mov Disord 13:
Pastor P, Pastor E, Carnero C et al. (2001). Familial atypical 673676.
progressive supranuclear palsy associated with homozig- Reitblat T, Polishchuk I, Dorodnikov E et al. (2003). Primary
osity for the delN296 mutation in the tau gene. Ann Neu- antiphospholipid antibody syndrome masquerading as pro-
rol 49: 263267. gressive supranuclear palsy. Lupus 12: 6769.
348 D. J. BURN AND A. J. LEES

Richter-Landsberg C, Bauer NG (2004). Tau-inclusion body Seppi K, Schocke MFH, Esterhammer R et al. (2003). Diffu-
formation in oligodendroglia: the role of stress proteins and sion-weighted imaging discriminates progressive supra-
proteasome inhibition. Int J Dev Neurosci 22: 443451. nuclear palsy from PD, but not from the parkinson
Righini A, Antonini A, De Notaris R et al. (2004). MR ima- variant of multiple system atrophy. Neurology 60:
ging of the superior profile of the midbrain: differential 922927.
diagnosis between progressive supranuclear palsy and Par- Shinotoh H, Namba H, Yamaguchi M et al. (1999). Positron
kinsons disease. Am J Neuroradiol 25: 927932. emission tomographic measurement of acetylcholinester-
Rivaud-Pechoux S, Vidailhet M, Gallouedec G et al. ase activity reveals differential loss of ascending choliner-
(2000). Longitudinal ocular motor study in corticobasal gic systems in Parkinsons disease and progressive
degeneration and progressive supranuclear palsy. Neurol- supranuclear palsy. Ann Neurol 46: 6269.
ogy 54: 10291032. Soliveri P, Monza D, Paridi D et al. (2000). Neuropsycholo-
Rohs G (1996). Progressive supranuclear palsy: managing gical follow up in patients with Parkinsons disease, stria-
the disabilities and providing nursing support. Axone 17: tonigral degeneration-type multiple system atrophy, and
6065. progressive supranuclear palsy. J Neurol Neurosurg Psy-
Rojo A, Pernaute RS, Fontan A et al. (1999). Clinical genet- chiatry 69: 313318.
ics of familial progressive supranuclear palsy. Brain 122: Sosner J, Wall GC, Sznajder J (1993). Progressive supranuc-
12331245. lear palsy: clinical presentation and rehabilitation of two
Romano S, Colosimo C (2001). Procerus sign in progressive patients. Arch Phys Med Rehabil 74: 537539.
supranuclear palsy. Neurology 57: 19281929. Stanford PM, Halliday GM, Brooks WS et al. (2000).
Rottach KG, Riley DE, DiScenna AO et al. (1996). Dynamic Progressive supranuclear palsy pathology caused by a
properties of horizontal and vertical eye movements in novel silent mutation in exon 10 of the tau gene: expan-
parkinsonian syndromes. Ann Neurol 39: 368377. sion of the disease phenotype caused by tau gene muta-
Sandyk R (1998). Transcranial AC pulsed applications of tions. Brain 123: 880893.
weak electromagnetic fields reduces freezing and falling Stanford PM, Brooks WS, Teber ET et al. (2004). Frequency of
in progressive supranuclear palsy: a case report. Int J Neu- tau mutations in familial and sporadic frontotemporal
rosci 94: 4154. dementia and other tauopathies. J Neurol 251: 10981104.
Santacruz P, Uttl B, Litvan I et al. (1998). Progressive supra- Tamai S, Almeida OP (1997). Nortriptyline for the treatment
nuclear palsy: a survey of the disease course. Neurology of depression in progressive supranuclear palsy. J Am
50: 16371647. Geriatr Soc 45: 10331034.
Sasaki N, Fukatsu R, Tsuzuki K et al. (1998). Advanced gly- Tolosa E, Valldeoriola F, Marti MJ (1994). Clinical diagno-
cation end products in Alzheimers disease and other neu- sis and diagnostic criteria of progressive supranuclear
rodegenerative diseases. Am J Pathol 153: 11491155. palsy (Steele-Richardson-Olszewski syndrome). J Neural
Scaravilli T, Pramstaller PP, Salerno A et al. (2000). Neuro- Transm Suppl 42: 1531.
nal loss in Onufs nucleus in three patients with progres- Tomasiewicz HG, Flaherty DB, Soria JP et al. (2002). Trans-
sive supranuclear palsy. Ann Neurol 48: 97101. genic zebrafish model of neurodegeneration. J Neurosci
Schleider MA, Nagurney JT (1977). Progressive supranuclear Res 70: 734745.
ophthalmoplegia. Association with cricopharyngeal dys- Tsuboi Y, Slowinski J, Josephs KA et al. (2003). Atrophy of
function and recurrent pneumonia. JAMA 237: 994995. the superior cerebellar peduncle in progressive supranuc-
Schofield EC, Caine D, Kril JJ et al. (2005). Staging disease lear palsy. Neurology 60: 17661769.
severity in movement disorder tauopathies: brain atrophy Urakami K, Wada K, Arai H et al. (2001). Diagnostic signif-
separates progressive supranuclear palsy from corticobasal icance of tau protein in cerebrospinal fluid from patients
degeneration. Mov Disord 20: 3439. with corticobasal degeneration or progressive supranuclear
Schrag A, Ben-Shlomo Y, Quinn NP (1999). Prevalence of palsy. J Neurol Sci 183: 9598.
progressive supranuclear palsy and multiple system atro- Van Gerpen JA, Ahlskog JE, Petty GW (2003). Progressive
phy: a cross-sectional study. Lancet 354: 17711775. supranuclear palsy phenotype secondary to CADASIL.
Schrag A, Good CD, Miszkiel K et al. (2000). Differentiation Parkinsonism Relat Disord 9: 367369.
of atypical Parkinsonian syndromes with routine MRI. van Slegtenhorst M, Lewis J, Hutton M (2000). The mole-
Neurology 54: 697702. cular genetics of the tauopathies. Exp Gerontol 35:
Schreckenberger M, Hagele S, Seissmeier T et al. 461471.
(2004). The dopamine D2 receptor ligand 18F-desmeth- Verny M, Jellinger KA, Hauw JJ et al. (1996). Progressive
oxyfallypride: an appropriate fluorinated PET tracer for supranuclear palsy: a clinicopathological study of 21
the differential diagnosis of parkinsonism. Eur J Nucl cases. Acta Neuropathol (Berl) 91: 427431.
Med Mol Imaging 31: 11281135. Vidailhet M, Rivaud S, Gouidier-Khouja N et al. (1994). Eye
Schulz JB, Skalej M, Wedekind D et al. (1999). Magnetic movements in parkinsonian syndromes. Ann Neurol 35:
resonance imaging-based volumetry differentiates idio- 420426.
pathic Parkinsons syndrome from multiple system atro- Vodusek DB (2001). Sphincter EMG and differential dia-
phy and progressive supranuclear palsy. Ann Neurol 45: gnosis of multiple system atrophy. Mov Disord 16:
6574. 600607.
PROGRESSIVE SUPRANUCLEAR PALSY 349

Wakabayashi K, Takahashi H (2004). Pathological heteroge- Williams AC, Nutt J, Lake CR et al. (1979). Actions of bromo-
neity in progressive supranuclear palsy and corticobasal criptine in the Shy-Drager and Steele-Richardson-Olszewksi
degeneration. Neuropathology 24: 7986. syndromes. In: K Fuxe, DB Calne (Eds.), Dopaminergic
Walter U, Niehaus L, Probst T et al. (2003). Brain parench- Ergot Derivatives and Motor Function.Pergamon Press,
yma sonography discriminates Parkinsons disease and Oxford, pp. 271283.
atypical parkinsonian syndromes. Neurology 60: 7477. Williams DR, Paviour DC, Watt HC et al. (2004). Chara-
Walter U, Dressler D, Wolters A et al. (2004). Sonographic cteristics of two distinct clinical phenotypes observed in
discrimination of corticobasal degeneration vs progressive pathologically proven progressive supranuclear palsy:
supranuclear palsy. Neurology 63: 504509. Richardsons syndrome and PSP-parkinsonism. Mov Disord
Ward-Smith PA, Berry P (1990). Autologous transplantation 19: S328S329.
as a treatment for progressive supranuclear palsy. J Neu- Wittman CW, Wszolek MF, Shulman JM et al. (2001). Tauo-
rosci Nurs 22: 100103. pathy in Drosophila: neurodegeneration without neurofi-
Warmuth-Metz M, Naumann M, Csoti I et al. (2001). Measure- brillary tangles. Science 293: 711714.
ment of the midbrain diameter on routine magnetic reso- Wolters A, Classen J, Kunesch E et al. (2004). Measure-
nance imaging: a simple and accurate method of ments of transcallosally mediated cortical inhibition for
differentiating between Parkinson disease and progressive differentiating parkinsonian syndromes. Mov Disord 19:
supranuclear palsy. Arch Neurol 58: 10761079. 518528.
Weiner WJ, Minagar A, Shulman LM (1999). Pramipexole in Zemaitaitis MO, Kim SY, Halverson RA et al. (2003). Trans-
progressive supranuclear palsy. Neurology 52: 873874. glutaminase activity, protein, and mRNA expression are
Wermuth L, Joensen P, Bunger N et al. (1997). High preva- increased in progressive supranuclear palsy. J Neuropathol
lence of Parkinsons disease in the Faroe Islands. Neurol- Exp Neurol 62: 173184.
ogy 49: 426432.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 48

Corticobasal degeneration

NATIVIDAD P. STOVER*, HARRISON C. WALKER AND RAY L. WATTS

University of Alabama at Birmingham, Birmingham, AL, USA

48.1. Historical aspects et al., 1964; Litvan et al., 1996), amyotrophic lateral
sclerosisparkinsonismdementia complex and argyro-
Corticobasal degeneration (CBD) is a neurodegenera- philic grain disease (AGD) (Tolnay and Clavaguera,
tive disorder that has gained the interest of clinicians 2004). In parallel, it has been discovered that abnorm-
and scientists, especially over the last two decades. alities of the vesicle-associated protein a-synuclein
The typical clinical presentation of CBD is one of an underlie the pathology of Parkinsons disease (PD),
atypical parkinsonian syndrome with cerebral cortical Lewy body dementia (LBD) and multiple system atro-
signs such as apraxia or cortical sensory loss, similar phy (MSA) (Wenning et al., 1994) and thus they com-
to the 3 cases described originally (Rebeiz et al., prise the synucleinopathies (Dickson, 1999b; Arai
1967). The disorder was initially called corticodentato- et al., 2001b). Cases of CBD have clinical features that
nigral degeneration with neuronal achromasia, based also overlap with the synucleinopathies.
on the pathological findings in the first cases. Other It is still debated whether CBD is a distinctive
names used include corticonigral degeneration with nosological entity or a syndrome. Currently, we rec-
neuronal achromasia, corticobasal ganglionic degen- ognize two clinical phenotypes of CBD: the first is a
eration and myoclonic dystonia. The term cortico- classic motor presentation with asymmetric parkinso-
basal degeneration was initially used in 1989 (Gibb nian symptoms and apraxia and/or cortical sensory
et al., 1989) and more recently the term corticobasal loss and the second is a presentation of dementia often
syndrome or complex has also been proposed to char- with focal cortical signs with or without parkinsonian
acterize better the heterogeneity and overlap of the features (Hachinski, 1997; Kertesz and Munoz, 2004).
clinical features and neuropathologic findings in cases
of CBD (Kertesz et al., 2000). 48.2. Description of the initial cases
The observation that the aggregation of tau, a
microtubule-associated protein, is involved in the James Parkinson described PD in 1817 and Jean-Martin
pathology of several neurologic disorders has revolu- Charcot and his disciples drew emphasis to the cardinal
tionized our understanding of CBD and its relationship features of the disease in the last years of the 19th
to other neurodegenerative diseases that are collec- century. They also described a number of parkinsonian
tively called tauopathies (Litvan, 1999; Goedert variants with atypical jerking movements and abnormal
et al., 2000; Tolnay and Probst, 2003; Cairns et al., posture in the limbs, described originally as hemiple-
2004). The identification of mutations in the tau gene gic parkinsonism (Charcot and Vulpian, 18611862;
on chromosome 17 in familial cases of frontotemporal Charcot, 18871888). It is possible that some of these
dementia with parkinsonism (FTDP-17) (Verin et al., patients had CBD.
1997) and the overlapping clinical and pathological In 1925 J. Lhermitte described a clinical case sug-
features of CBD with other tauopathies emphasized gestive of CBD to the French Neurological Society:
the importance of tau proteins in this group of disor- A carpenter retired at age 67 because of progressive
ders: Picks disease, primary progressive aphasia right hand clumsiness. At age 72, he could no longer
(PPA), progressive supranuclear palsy (PSP) (Steele walk independently and ambulated with a wide-based,

* Correspondence to: Natividad P. Stover, Department of Neurology, University of Alabama at Birmingham, 360 Sparks
Center, 1720 7th Avenue South, Birmingham, AL 35294-0017, USA. E-mail: npstover@uab.edu, Tel: 1-205-934-0683,
Fax: 1-205-996-4039.
352 N. P. STOVER ET AL.
shuffling gait with the right arm flexed. In addition, regions corresponding with areas of cortical atrophy
his right arm moved involuntarily like a foreign showed considerable demyelination and fibrous glio-
body. In spite of normal primary sensation, he could sis. The hippocampus, inferior and medial temporal
not recognize objects placed in his right hand (Ballan cortex and occipital regions were spared in the original
and Tison, 1997). cases. Considerable loss of pigmented neurons in the
The initial three cases described by Rebeiz et al. at substantia nigra (SN) was observed in 2 of the cases
the Massachusetts General Hospital (1967, 1968) pre- and was less marked in the third, mainly in the lateral
sented a unique pattern of progressive neurological two-thirds. The subthalamic nucleus contained a few
disorder with motor impairment, affecting mainly the swollen neurons and gliosis and in 2 of the cases there
left side of the body and characterized by stiffness, was significant loss and swelling of nerve cells in the
slowness, clumsiness, numbness or deadness and central nuclei of the cerebellum with retrogade atrophy
awkward movements of the left arm and leg as initial of the superior cerebellar peduncles, as well as gliosis
symptoms. In 2 patients the leg was affected first with of the red nuclei and the ventrolateral portion of the
progressive gait impairment. As the disease pro- thalamus. Those 2 cases also showed degeneration of
gressed, involuntary movements became prominent the corticospinal tracts in the brainstem and spinal
with elevation and abduction of the limbs that came cord, most likely secondary to neuronal loss in the cer-
on during attempted motor activity and when the ebral cortex. The characteristic pathological findings
affected arm was being used the opposite limb exhib- seen in other neurodegenerative diseases, such as
ited involuntary synkinesias. Each of the 3 cases had Lewy bodies, Pick bodies and amyloid plaques, were
some tremor, even if it was not the prominent feature. not observed in the initial cases.
There was progressive impairment in the use of A case described initially as myoclonic dystonia
affected limbs without motor weakness except late in was reported by Obeso et al. in 1983; the patient
the course of the disease. In 1 case, there was impaired was followed until death and the pathology was char-
position sense, tactile localization, two-point discrimi- acteristic for CBD. This patient had clinical features
nation and ability to recognize objects by touch and resembling PSP but pathological findings similar to
another patient had impaired position sense. In all cases Picks disease. The patient had focal dystonia and
there were increased tendon reflexes and increased myoclonus of an arm, alien-hand phenomenon and
resistance to passive movements of the affected limbs an akinetic-rigid syndrome and he developed supra-
and severe finger contractures. With time the 3 pati- nuclear gaze palsy, parkinsonism and mild cerebellar
ents developed impairment of speech and dysphagia. signs. The pathology showed frontoparietal atrophy
Cognitive dysfunction was not a remarkable feature in with cortical cell loss, Picks disease cells with glio-
the clinical picture until later in the disease. The symp- sis in the basal ganglia, midbrain tegmentun, SN and
toms in the 3 patients started in late middle age and they locus ceruleus. The pathological examination also
died 68 years after diagnosis. showed inclusions in the SN that were called cortico-
Radiographic contrast studies showed cerebral atro- basal inclusions, similar to the globose neurofibril-
phy in the 3 patients, more pronounced on the right than lary tangles (NFT) of PSP. Some of the nigral
on the left. None of the patients had seizures or myoclo- inclusions were similar to those seen in Picks dis-
nus. Electroencephalograms (EEG) showed slow- and ease (Obeso et al., 1985).
sharp-wave activity without specific changes. The rest Watts et al. (1985) described a case with pathologic
of the laboratory investigations were normal. correlation of CBD. The patient presented with stiff-
Postmortem examination of the brains revealed ness of the right arm and leg followed by progression
asymmetric frontoparietal cortical atrophy. On micro- of the symptoms to the left side. There was a rapid,
scopic examination the most severe changes were in irregular action tremor and impairment of balance that
the superomedial frontoparietal and Rolandic regions progressed to inability to walk without assistance.
with severe neuronal loss and disappearance of the During the third year of disease, he developed dysar-
cells in the outer three cortical layers with astrocytic thria and poor semantic content in his language. He
gliosis and little microglial activation. Many neurons had hypomimia, blepharospasm and saccadic break-
in the deep cortical layers showed swelling of the cell down of smooth pursuit eye movements. The right
bodies frequently accompanied by vacuolization. The arm and leg had severely increased tone and dystonic
swollen neurons often had eccentric nuclei without posturing of the fingers. The patient lost the ability
Nissl substance in the cytoplasm that prompted the to perform motor tasks previously learned like using
terms achromatic and ballooned as descriptors and a toothbrush and saluting with the right arm. Fascicu-
these histological abnormalities colocalized with the lations were seen in several muscle groups of the
distribution of the neuronal loss. The white-matter upper and lower limbs. There was no family history of
CORTICOBASAL DEGENERATION 353
neurologic disease. Electrophysiologic and laboratory described cases are Caucasian. Most of the patients
evaluations were normal. Imaging studies showed reported have been sporadic, but some studies do sug-
peri-Rolandic cortical atrophy and signal attenuation gest a familial tendency. One of the original cases
of the white matter beneath the left central sulcus. described had a history of similar neurologic disorders
Neuropathological examination revealed asymmetrical in several family members, possibly representing a
cerebral cortical atrophy, worse on the left with neuro- case of FTDP-17. There is no evidence at present that
nal loss and reactive gliosis in all layers of the premo- any CBD cases are related to a toxic exposure or to an
tor, anterior cingulate, inferior parietal, insula and infectious agent (Di Maria et al., 2000; DePold Hohler
anterior temporal regions. White-matter changes con- et al., 2003).
sisted of extensive loss of myelinated axons and glio-
sis. Occasional neuritic plaques were noted, but there 48.4. Diagnosis
were no NFT. In areas not so severely affected there
were swollen neurons with achromasia. There was neu- There have been several attempts to establish diagnos-
ronal loss in the SN bilaterally, but no Lewy bodies were tic criteria for CBD, but a formal validation has not
seen. No abnormalities were observed in the striatum, been performed. Most of the cases diagnosed with
globus pallidus, substantia innominata, amygdala, CBD in movement disorder clinics focus primarily
hypothalamus, cerebellum, brainstem or spinal cord. on motor symptoms. Signs of cortical impairment such
The peripheral nerves showed demyelination and as apraxia or cortical sensory loss usually develop
there were denervation changes in skeletal muscles. within 13 years of onset, but in some series dementia
Therapeutic trials with multiple agents were ineffective. was the most frequent initial symptom in patients with
Several other authors have contributed clinical and pathologically confirmed CBD (Bergeron et al., 1998;
pathological descriptions of cases consistent with Litvan et al., 2003).
CBD and the use of modern laboratory techniques in Riley and Lang (1993) proposed a set of clinical
recent years has helped to characterize the disease characteristics of CBD based on the review of 12
and its relationship with the other neurodegenerative cases, 9 with pathological confirmation of CBD. The
disorders better. clinical manifestations were divided as related to basal
In recent years a number of patients with atypical ganglia dysfunction (bradykinesia/akinesia, rigidity,
parkinsonian syndromes and a predominance of demen- dystonia, postural instability, falls, athetosis and oro-
tia have turned out to have the classical pathologic lingual dyskinesias), cerebral cortical signs (cortical
picture of CBD (Grimes et al., 1999; Graham et al., sensory loss, apraxia, alien-limb phenomenon, demen-
2003a; Kurz, 2005). There are also reports of cases pre- tia, frontal-release signs and dysphasia) and other
senting with clinical features of CBD that lack specific manifestations (postural/action tremor, hyperreflexia,
pathological features (Lerner et al., 1992; Maraganore impaired ocular motility, dysarthria, focal reflex
et al., 1992; Giannakopoulos et al., 1995; Brown myoclonus, impaired eyelid motion and dysphagia).
et al., 1998). The authors proposed inclusion and exclusion features
for the diagnosis of CBD to establish research criteria.
48.3. Epidemiology The inclusion criteria were defined as the presence of
rigidity plus one cortical sign (apraxia, cortical sensory
Epidemiologic studies in CBD are difficult because of loss or alien limb) or asymmetric rigidity, dystonia and
clinical diagnostic uncertainties and the lack of a bio- focal reflex myoclonus. Other authors (Kumar et al.,
logical marker. The incidence and prevalence of 1998) also included as inclusion criteria an asym-
CBD increase if the different clinical and pathological metric, progressive course, presence of higher cortical
phenotypes are considered as part of a syndrome or dysfunction and a movement disorder consisting of an
complex. The incidence of non-Alzheimer and non- akinetic-rigid syndrome, levodopa resistance, promi-
vascular dementia cases is around 10% in cases from nent limb dystonia and reflex or focal myoclonus.
brain banks (DLB, Picks disease and FTDP-17) and Exclusion criteria were defined as early dementia,
this percentage may increase to 20% if CBD and early vertical gaze palsy, rest tremor, severe auto-
PSP are included (Tanner, 1996). nomic dysfunction, sustained responsiveness to levo-
The age of onset of CBD is usually in later adult dopa and lesions or imaging studies suggesting a
life with a mean onset of symptoms at 63 (7.7) years. different diagnosis.
The youngest case with pathological confirmation was Watts et al. (1994) proposed major and minor
34 years old. Some authors have suggested a predomi- criteria to be used in the clinical diagnosis of CBD.
nance in women but both sexes are affected. There The major criteria were defined as akinesia, rigidity,
is no clear ethnic preponderance and most of the postural/gait disturbance, action/postural tremor,
354 N. P. STOVER ET AL.
alien-limb phenomenon, dystonia, myoclonus and cor- dysarthria and postural instability often develop over
tical signs. Minor criteria were described as chor- the ensuing years (Wenning et al., 1998).
eoathetosis, dementia, cerebellar signs, supranuclear
gaze abnormalities, frontal-release signs and blephar- 48.4.1. Motor symptoms
ospasm. A strong degree of asymmetry was also
necessary for the diagnosis of CBD. The so-called typical cases of motor presentation in
Another study presented the description of 105 CBD consist of limb clumsiness or difficulty using
patients with different neurodegenerative diseases and the limb, with or without rigidity and difficulty coordi-
known pathologic correlation to a group of movement nating small, precise movements. An asymmetric, insi-
disorders specialists, to investigate the accuracy of the diously progressive parkinsonism, usually of the
clinical diagnosis of CBD. The study evaluated 10 bradykinetic or akinetic-rigid type, with or without tre-
autopsy-proven cases of CBD from a movement disor- mor, is typical (Schneider et al., 1997). The symptoms
der center and found that specificity of the diagnosis respond poorly to levodopa treatment, although one-
of CBD cases using the clinical features was high but third of patients may have some initial response, mak-
the sensitivity was low, particularly in the first 3 years ing the diagnosis challenging before other signs and
of the disease. This finding suggests that CBD is most symptoms appear. Akinesia, rigidity and apraxia are
likely underdiagnosed. In this study, the best initial the most common motor findings during the course
clinical predictors for the diagnosis of CBD included of the disease, occurring in over 90% of motor cases
limb dystonia, asymmetric parkinsonism, apraxia and within the first 3 years of illness. Postural instability,
absence of balance or gait disturbances. There were gait disorder and speech disorder early in the course
some cases with pathology consistent with PSP that of CBD are also frequent clinical features (Lang
had limb dystonia as the predominant presenting sign et al., 1994a). Rigidity in one arm is probably the most
and most of the cases with an initial gait disorder common parkinsonian feature documented in motor
actually had pathology indicative of PSP. Other CBD cases and this symptom alone can be severe
clinical motor features commonly seen in CBD are and very debilitating (Lang, 2000). There are also
alien-limb phenomenon, choreoathetoid movements, cases reported with symptoms affecting the lower
blepharospasm, eye movement abnormalities, myo- limbs initially (Lalive et al., 2000).
clonus and speech and swallowing problems. This The tremor in CBD differs from the typical resting
study also mentioned cognitive and neuropsychiatric or postural tremor seen in PD. It is a faster (6 8 Hz)
symptoms among the frequent manifestations (Litvan tremor with action and postural components and it
et al., 1997). has a more irregular, jerky quality. Myoclonic move-
The statistical analyses in the study with 24 patients ments are most frequently reported in the upper extre-
pathologically diagnosed with PSP and 27 cases with mities and they are usually superimposed on the
CBD identified two predictor models for CBD tremor.
patients. Patients with CBD presented with lateralized Myoclonus develops in approximately half of
motor and cognitive signs, whereas PSP patients often patients with CBD during the course of the illness
had severe postural instability at onset, symmetric par- (Chen et al., 1992; Thompson et al., 1994). Myoclonus
kinsonism, vertical supranuclear gaze palsy and speech may be preceded by a jerky tremor and it manifests
problems and CBD patients with a non-motor phe- itself in the most affected limb early in the disease.
notype had early frontal dementia and eventually Myoclonus is most evident in distal muscles and it
bilateral parkinsonism (Litvan et al., 1999). may coexist with dystonia in the most affected limbs
Rinne et al. (1994) reported that the most common as well (Brunt et al., 1995; Mima et al., 1998). The
motor presentation in a series of 36 patients with myoclonus in CBD is best seen during action or main-
pathologically proven or clinically probable diagnosis tenance of a posture; however, it can also be elicited
of CBD was a useless arm due to any combination of by other stimuli, including cutaneous or auditory sti-
rigidity, akinesia, dystonia, apraxia or alien-limb phe- muli. The myoclonus may be masked by the presence
nomena, with or without myoclonus. Of those patients, of increased muscle tone (Thompson and Shibasaki,
20 presented with symptoms beginning in the upper 2000; Lleo et al., 2002; Caviness, 2003).
extremities and 10 in the lower extremities. Electrophysiologic studies show that the myoclonus
Other less common presentations of CBD include is mainly an action or reflex myoclonus, preceded by a
combined arm and leg involvement with motor dysfunc- short-duration muscle discharge with simultaneous
tion, unilateral painful paresthesias and orofacial dyski- activation of antagonistic muscles (Carella et al.,
nesias. The gradual extension of the symptoms to 1991, 1997). Brief trains of high-frequency myoclonic
the contralateral limbs, facial hypomimia, dysphagia, discharges happen in clusters of 23 Hz and in action
CORTICOBASAL DEGENERATION 355
myoclonus this activity occurs throughout the period of increased latency of horizontal saccades bilaterally
voluntary muscle activation and in a pattern consistent compared with controls and patients with PSP. Vertical
with corticospinal activation. The intracortical compo- saccades may be slightly impaired in patients with CBD
nent of the long latency in reflex myoclonus in CBD is compared with PSP patients (except for upgaze in
shorter than the intracortical processing of sensory input elderly patients) (Rottach et al., 1996). In early stages
in typical cortical reflex myoclonus. Cortical hyperex- of CBD smooth pursuit may be slow and exhibit sacca-
citability was demonstrated in some cases of CBD with dic breakdown, but the range of movements is generally
myoclonus, consistent with impaired cortical inhibitory full (Hamilton, 2000; Rivaud-Pechoux et al., 2000;
systems. Although myoclonus is commonly seen in Vidailhet and Rivaud-Pechoux, 2000). As the illness
CBD, it is also described in later stages of Alzheimers progresses, patients with CBD gradually lose the ability
disease and in CreutzfeldtJakob disease (CJD), MSA, to initiate rapid saccades in response to verbal com-
Huntingtons disease and cerebellar degeneration mands; however, they retain both spontaneous saccades
(Shibasaki, 1995; Thompson, 1995). and optokinetic nystagmus. Asymmetry of saccade
Dystonia is frequently observed in patients with latency is also a helpful and persistent abnormality char-
CBD and it has not been possible to differentiate his- acteristic of CBD cases. Consecutive evaluation of eye
torically the onset of dystonia from the onset of other movements separated by several months may therefore
features of CBD (Vanek and Jankovic, 2000). Dysto- help to differentiate among CBD, PSP and other atypi-
nia was described in the initial cases and other studies cal parkinsonian syndromes. Visuospatial dysfunction
reported the presence of dystonia in 4070% of cases has been described as the presenting symptom in CBD
(with pathologic confirmation of CBD in a small num- (Tang-Wai et al., 2003).
ber of these cases). Initial presentation with dystonia in Upper motor neuron syndromes and corticospinal
one arm has been reported in a majority of motor cases signs may be present in CBD patients and were
of CBD. The dystonia is usually asymmetric and the described in the early cases by several authors.
arm is the most frequently affected region. Usually Speech problems and dysphagia are frequently
the hand and forearm are flexed and the arm is reported in patients with CBD, especially in later phases
adducted at the shoulder area. The fingers are typically of the disease as cortical and subcortical pathways
flexed at the metacarpophalangeal joints, extended or become involved (Frattali and Sonies, 2000). Motor
flexed at the proximal and distal interphalangeal joints speech problems can range from monotonous voice,
and show variable degrees of fixed postures with or dysphonia, echolalia or pallilalia to dysarthria or even
without associated contractures. Head, neck, leg, knee, anarthria (Lang, 1992). Dysphagia usually appears at
foot (mainly inversion), trunk and toe (flexed or later stages of the disease (Muller et al., 2001).
extended) dystonia are less frequently observed (Oide
et al., 2002). Some patients have thumb, index and 48.4.2. Cortical symptoms
middle fingers extended and fourth and fifth fingers
flexed. Axial dystonia (head, neck, trunk) is less fre- The most frequent cortical symptoms are apraxia,
quent and may include retrocollis, anterocollis or torti- cortical sensory abnormalities, aphasia, alien-limb
collis (Vanek and Jankovic, 2001). Oromandibular phenomenon and frontal-release reflexes.
dystonia, orofacial dyskinesias and blepharospasm Apraxia is considered to be a hallmark of CBD
have been reported. Pain accompanying dystonia is early in the disease and raises the possibility of a diag-
described in over 40% of cases; it can be very intense nosis if seen as part of the motor presentation. Differ-
and is usually associated with contractures (Stover ent types of apraxia can be seen in patients with CBD
and Watts, 2001). Rigidity and bradykinesia are more depending not only on the area of cortex affected initi-
evident in the dystonic limbs. Spontaneous onset of ally but also on the timeframe of disease progression
choreoathetoid movements in patients with and with- in which the apraxia is evaluated (Jacobs et al., 1994;
out dystonia involving the limbs and facial muscles Okuda and Tachibana, 1994; Soliveri et al., 2003,
may be present, usually associated with the dystonic 2005; Zadikoff and Lang, 2005).
extremity (Riley and Lang, 2000). Ideomotor apraxia has been the most frequent form
Eye movement abnormalities in CBD are distinctive of apraxia in patients with CBD in several studies.
from other movement disorders frequently misdiag- Ideomotor apraxia is manifested by impairment of tim-
nosed as CBD, such as PSP, and ocular findings may ing, sequencing, spatial organization and mimicking of
therefore help to improve diagnostic accuracy in the movements. Patients with ideomotor apraxia commit
early stages of the disease (Pierrot-Deseilligny et al., mainly temporal (irregular speed and sequencing) and
1989; Pierrot-Deseilligny, 1994; Vidailhet et al., spatial errors (abnormal amplitude, orientation of
1994). In patients with CBD there is a significantly objects and movements and abnormal use of body
356 N. P. STOVER ET AL.
parts as objects). Ideomotor apraxia is associated with way, a focal sensory complaint in the setting of atypical
damage to the parietal association areas, premotor cor- parkinsonism should raise the question of CBD.
tex and interhemispheric white-matter bundles that The alien-limb phenomenon is a failure to recog-
connect them, as well as basal ganglia and thalamus nize ownership of an extremity in the absence of visual
(Jacobs et al., 1999; Leiguarda et al., 2000). cues, accompanied by observable involuntary motor
Patients with CBD may also demonstrate ideational activity. It is associated with autonomous activity of
and limb kinetic apraxia (Otsuki et al., 1997; Okuma the affected extremity, which may be perceived by
et al., 2000). Fingers and hand movements are more the subject as outside his or her control (Goldberg
commonly affected and the movements are awkward, and Bloom, 1990; Fisher, 2000). Up to half of patients
amorphous and contaminated with movements unne- with CBD may develop the alien-limb phenomenon,
cessary to perform a task. This form of apraxia is often with coexisting dystonia (Banks et al., 1989;
mainly seen with lesions of the premotor cortex with Ball et al., 1993), myoclonus, apraxia, athetosis,
or without associated parietal cortex or basal ganglia pseudoathetosis and signs of cortical sensory loss
involvement. Ideomotor and limb kinetic apraxia may dysfunction such as sensory neglect, astereognosis
coexist in the same patient. Limb kinetic apraxia is and agraphesthesia (Doody and Jankovic, 1992). If
most frequently described in patients with mild dysto- the dystonia or rigidity is severe enough to cause
nia or rigidity (Merians et al., 1999; Ishiwata et al., contractures, these autonomous movements may be
2000; Leiguarda et al., 2003; Salter et al., 2004). limited. Most patients exhibit intermanual conflict,
Ideational apraxia is a dysfunction of the praxis mirror movements and difficulties with bimanual
conceptual system. It is an inability to sequence cor- coordination with compulsive synkinesias in the
rectly different movements needed to perform a speci- opposite limb. Posturing and levitation are asso-
fic task (Poeck, 1983). The performance in ideational ciated with alien-limb phenomenon in CBD more
apraxia is abnormal in the content and tool selection commonly than in other etiologies.
(including perseverations and pantomime-related Olfaction is not affected in CBD (Wenning et al.,
errors) and it may be observed in later stages of the 1995; Muller et al., 2002a; Hawkes, 2003).
disease or in patients with dementia and language dys- Eventually, many patients exhibit signs of corti-
function at presentation (De Renzi and Lucchelli, cospinal dysfunction, with extensor plantar responses
1988). In limb kinetic apraxia the manipulative beha- and hyperreflexia. The differentiation of spasticity
vior is affected by a decrease in dexterity and fine from rigidity in these patients is clinically very diffi-
movements in the affected limb (Peigneux et al., cult, but the loss of voluntary movement is almost cer-
2001). More advanced cases of CBD make it more tainly related in part to degeneration of primary and
difficult to appreciate apraxia due to the severe brady- secondary cortical motor regions that contribute to
kinesia, rigidity and dystonia in the same limb (Caselli the corticospinal tracts. In fairly advanced stages of
et al., 1999). CBD, frontal-release signs (grasp, glabellar and exag-
Speech apraxia was described in 2 patients (1 with gerated facial and palmomental reflexes) may become
pathological confirmation of CBD) as a presenting prominent.
sign in the evolution of the classical clinical motor Urinary symptoms, mainly consisting of decreased
syndrome (Rosenfield et al., 1991). Orofacial apraxia bladder capacity and detrusor overactivity, are fre-
with dysarthria has been reported in CBD patients quently seen in patients with CBD, especially in more
(Ozsancak et al., 2000). Apraxia has been described advanced stages (Sakakibara et al., 2004).
in PSP cases as well (Pharr et al., 2001; Denes, 2002).
Language disorders are frequently seen during the 48.4.3. Cognitive and neuropsychiatric symptoms
course of CBD (Frattali et al., 2000; Graham et al.,
2003b) and most of the cases initially diagnosed with Although many of the early CBD case reports described
PPA were shown to have pathological changes con- little cognitive dysfunction, it is now accepted that this
sistent with FTDP-17 and CBD/PSP in the pathologic may be the presenting and predominant feature in many
evaluation (Black, 2000). patients. Patients with CBD frequently show abnormal
Involuntary synkinesias of the less affected limb are cognitive and neuropsychological profiles in asso-
seen in CBD patients when they attempt complex ciation with motor and praxis disorders. A spectrum of
movements with the more affected, contralateral limb. different degrees of intellectual, memory and language
Abnormalities of two-point discrimination and impairment may develop (Cummings et al., 1994).
somatosensory extinction to double simultaneous sti- Storage and recall information and temporospatial
mulation may be present several years before apraxia orientation are usually preserved in early stages of
and other cortical symptoms become evident. In this CBD where motor symptoms predominate (Pillon
CORTICOBASAL DEGENERATION 357
et al., 1995; Levy et al., 1996). In one study with 11 cases Table 48.1
of neuropathologically confirmed CBD, all patients
Clinical manifestations of corticobasal degeneration
eventually developed cognitive deficits; the onset, nature
and severity of the impairment, however, varied widely. Motor signs and symptoms
Cognitive disturbances preceded or accompanied the Rigidity
onset of the movement disorder in 4 patients; an addi- Bradykinesia/akinesia
tional 3 patients developed memory loss, progressing to Dystonia (focal limb and blepharospasm)
more global dementia within 23 years prior to the onset Tremor: postural and action tremor
of motor symptoms. Another patient displayed mild Gait disorders/postural instability
Dysarthria
early memory impairment and, in 3 individuals, demen-
Dysphagia
tia was a late feature (Litvan et al., 1998).
Myoclonus
The memory disorders in CBD patients without Choreoathetosis
significant pathology in the temporal lobes and hippo- Orofacial dyskinesias
campus may be related to disruption of subcorticofrontal Hyperreflexia
circuits. This may explain the dysexecutive syndrome Impaired ocular movements
with abnormalities of dynamic motor control and execu- Impaired eyelid motion
tion similar to those described in other neurodegenera- Involuntary synkinesias
tive diseases related to CBD, most notably PSP (Bak Cortical signs and symptoms
et al., 2005). Those patients also have difficulties with Apraxia
motor programming, manifested by deficits in temporal Cortical sensory loss: abnormalities of two-point
discrimination, somatosensory extinction to double
organization, bimanual coordination and inhibition of
simultaneous stimulation, astereognosis, agraphesthesia
interfering motor activities, word fluency and switching
Alien-limb phenomenon
between different task sets and activities (Cummings Dysphasia/aphasia
and Litvan, 2000; Green, 2000; Pillon and Dubois, Frontal-release signs: grasp, glabellar, palmomental reflexes
2000). Those changes contrast with the findings in Alz- Cognitive and neuropsychiatric signs and symptoms
heimers disease, characterized by a true amnesic syn- Dementia
drome and linguistic disorders. CBD patients perform Apathy
worse on unimanual and bimanual dexterity compared Depression
with PSP patients. Verbal fluency and evocation of ver- Irritability/agitation
bal series are also impaired in CBD patients and there Disinhibition
is a tendency to lower scores in patients with initial Illusions
Delusions
right-sided symptoms (Monza et al., 2003).
Obsessive-compulsive behavior
The neuropsychological features most frequently
Sleep disorders
described in patients with CBD include depression, Somatic pain
apathy, irritability, anxiety, disinhibition, agitation,
disorders of sleep and high incidence of somatic
pain. It has been demonstrated that CBD patients with
left-sided symptoms (as a result of right hemisphere rates of depression to PD but lower rates of anxiety
involvement) manifested greater disinhibition, apathy, (Hargrave and Rafal, 1998). LBD has a high rate of
irritability and lower depression scores than patients visual hallucinations, a phenomenon not described in
with predominantly right-sided symptoms (Moretti pathologically confirmed CBD cases.
et al., 2005). Manifestations of obsessive-compulsive Rapid-eye movement (REM) sleep behavior disor-
disorder, including recurrent thoughts, repetitive acts, der has been associated with the synucleinopathies
indecisiveness, checking behaviors and preoccupation (Kimura et al., 1997; Boeve et al., 2001), but it has
with perfectionism, are also included in the neuropsy- also been reported in patients with CBD (Wetter
chological profile and they are described in both motor et al., 2002). Unilateral periodic limb movements of
and cognitive presentations of CBD. sleep have been seen in CBD as well (Iriarte et al.,
Contrasting the neuropsychiatric findings in CBD 2001) (Table 48.1).
with those found in other neurodegenerative disorders
is helpful in establishing a differential diagnosis. Com- 48.5. Pathology
pared with Alzheimers disease patients, CBD patients
have less apathy, agitation, anxiety and delusions. CBD as a distinctive pathological condition may have
Patients with FTDP-17 usually manifest more disinhi- several clinical presentations: on the other hand, a vari-
bition and euphoria. CBD patients manifest similar ety of pathologies can cause the classical syndrome that
358 N. P. STOVER ET AL.
was initially thought to be specific to the pathology of inferior olivary nuclei and cerebellar dentate nucleus
CBD. This may be better understood if the different suggests an alternative diagnosis such as MSA.
pathological findings are considered as part of a broader
syndrome. Bearing this in mind, we can describe the 48.5.2. Microscopic and biochemical findings
typical pathological hallmarks of the CBD syndrome.
In addition to the morphologic examination, immuno- The microscopic examination of cortical sections
histochemical stains and biochemical and genetic ana- stained with hematoxylin and eosin shows neuronal
lyses are directed to identify tau pathology and to rule loss and gliosis associated with vacuolar or spongiform
out other neurodegenerative processes (Feany et al., change of the upper cortical layers. The characteristic
1996; Litvan, 1997; Mirra and Hyman, 2002). cortical finding in CBD is the ballooned neuron (BN),
achromatic cells with swollen perikarya and eccentri-
48.5.1. Macroscopic findings cally displaced nuclei without Nissl substance. These
neurons have a pale and ballooned appearance (Mack-
Asymmetric atrophy of the frontoparietal cortex is enzie and Hudson, 1995) and they can be seen in other
the finding most consistently seen in typical cases tauopathies (Mori et al., 1996), Alzheimers disease
of CBD. Atrophy is usually more evident in the (Fujino et al., 2004), CJD (Nakazato et al., 1990) and
superior peri-Rolandic frontal gyrus, the pre- and amyotrophic lateral sclerosis. The presence of BN in
postcentral parasagittal gyri and the superior part of the peri-Rolandic region, however, is characteristic of
the parietal lobe. The asymmetry of the atrophy CBD. Immunocytochemical staining of the BN is posi-
usually correlates with the laterality of the clinical tive for phosphorylated neurofilament proteins, a--
manifestations. However, cases have been described crystallin and sometimes ubiquitin (Castellani et al.,
with asymmetric symptoms and the pathological 1995; Halliday et al., 1995). Notably, tau and a-synu-
exam showed no significant asymmetry with bilateral clein immunoreactivity are negative in BN (Fig. 48.1).
atrophy of the precentral gyrus and other cortical
regions variably affected. This lack of asymmetry is
usually seen late in patients with severe symptoms.
Cortical atrophy may be more generalized and include
the inferior frontal and temporal lobes in cases that
mainly present with cognitive problems and/or PPA.
The occipital lobe, hippocampus and parahippocampal
gyrus are usually spared. Of note, asymmetric parietal
lobe atrophy has been described in cases with pathol-
ogy consistent with Alzheimers disease (Kaida et al.,
1998).
As a consequence of the cortical atrophy, the ven-
tricles and cerebral aqueduct may be enlarged. If the
atrophy is significant, changes in the white matter
may be seen as well. The corpus callosum may be
atrophic (Yamauchi et al., 1998) and the anterior limb
of the internal capsule, corticospinal fascicles within
the base of the pons and the medullary pyramids may
show attenuation, in part related to retrograde cortical
atrophy. Other white-matter pathways, such as the
optic tract, anterior commissure and fornix, are usually
unaffected in the gross evaluation of CBD patients.
Involvement of subcortical gray matter varies
widely from case to case, but it is usually less severe
and variable than the cortical atrophy. The head of
the caudate nucleus may have a flattened appearance
and the thalamus in CBD cases tends to be smaller Fig. 48.1. A hematoxylin and eosin-stained section of frontal
than normal. Transverse sections of the brainstem cortex from the brain of a patient with corticobasal degeneration
show severe loss of neuromelanin pigment in the SN; shows a large ballooned neuron (large arrow) and another neu-
however, neuromelanin within the locus ceruleus ron with a cytoplasmic inclusion body (small arrow). Courtesy of
is usually preserved. Marked atrophy of the pons, Dr. K.A. Roth, UAB Division of Neuropathology.
CORTICOBASAL DEGENERATION 359
Astrocytic plaques carry significant diagnostic value erative disorders, including synucleinopathies and
and are considered the most specific histopathologic tauopathies; however, Alzheimers disease-type patho-
features in CBD by some authors (Ishizawa and Dick- logic features are more likely to coexist with synuclei-
son, 2001). They are mainly located in the neocortex, nopathies (Ingelson et al., 2001; Josephs et al., 2004b).
appearing as an annular cluster of short processes that Cholinergic neuronal loss has been described in
resemble the neuritic plaque of Alzheimers disease; patients with CBD and PSP pathological findings
however, these astrocytic plaques do not contain amy- (Kasashima and Oda, 2003). Spinal cord involvement
loid. Tau positivity in these lesions is derived from with the presence of neuropil threads and neuronal
astrocytes as demonstrated by double immunostaining inclusions has been described in CBD (Iwasaki et al.,
for tau and the astrocytic marker glial fibrillary acidic 2005) and PSP patients as well (Kikuchi et al., 1999).
protein. Other less characteristic tau-immunoareactive
astrocytic lesions are described in CBD, including 48.5.3. Tau pathology
thorn-shaped astrocytes, which are found in all tauopa-
thies (Armstrong et al., 2001; Dickson, 2003). Tau is a structural microtubule-associated phosphopro-
Glial threads, which indicate dystrophic axonal and tein that stabilizes microtubules and promotes tubulin
glial processes in the gray and white matter, are polymerization, playing an important role in maintain-
described in CBD (Komori et al., 1997; Richter-Lands- ing neuronal integrity and axoplasmic transport (Mirra
berg and Bauer, 2004). Neuritic threads are less com- et al., 1999; Forman et al., 2000). Tau undergoes
mon than in Alzheimers disease. A key feature that selective phosphorylation that determines its func-
distinguishes CBD from Alzheimers disease is the tional state (Hanger et al., 2002). Normal tau is soluble
lack of both neuritic plaques and diffuse plaques in sil- and heat-stable, but under pathological conditions it
ver-stained material. Conversely, Alzheimers disease becomes insoluble and forms aggregates as the result
does not exhibit the tau-positive glial inclusions of abnormal phosphorylation or abnormal ratios of
observed in CBD (Ikeda et al. 1994; Horoupian and different isoforms (Buee and Delacourte, 1999).
Wasserstein, 1999). The most important diagnostic finding in CBD and
Silver stain preparations typically show the pre- tauopathies in general is the presence of abnormal tau-
sence of NFT in affected areas. The tangles vary in positive deposits (Lee et al., 2001). The tau pathology
configuration from delicate thread-like structures to is extensive in CBD and is present within neurons and
compact globoid morphologies. The typical flame- glial cells of the cortex, subcortical nuclei and brain-
shaped appearance of NFT seen in Alzheimers stem (Cordato et al., 2001). In most affected neurons,
disease is less frequent in CBD (Ikeda et al., 1995; abnormal tau immunoreactivity is present as diffuse
Dickson et al., 1996; Dickson and Litvan, 2003). The or granular cytoplasmic deposits, called pretangles
ultrastructure of tangles in CBD consists of paired (Komori, 1999).
helical filaments or twisted filaments with a wider A characteristic feature of CBD and other tauopa-
diameter and longer periodicity than the filaments thies that distinguishes them from Alzheimers disease
present in Alzheimers disease. The tangles in PSP is the presence of tau-immunoreactive glial inclusions
are more uniform, compact and globoid in shape and in cell bodies and processes. The so-called astrocytic
they consist of straight filaments at the ultrastructural plaques are the most characteristic astrocytic lesion
level (Collins et al., 1995; Dickson, 1999a). in CBD and there are mainly located in the neocortex.
Cell loss in the SN is usually severe with extraneur- The astrocytic plaques resemble the neuritic plaques of
onal neuromelanin in phagocytes (melanophagia) and Alzheimers disease; however, they do not contain
residual neurons typically contain NFT. Lewy bodies amyloid or dystrophic neurites. Another lesion, the
are not seen in CBD. The locus ceruleus, raphe nuclei, tufted astrocyte, consists of fibrillary accumulation
tegmental gray matter, the subthalamic nuclei and the of tau within cell bodies and processes; it is believed
ventrolateral nucleus of the thalamus may also contain to be more characteristic of PSP than of CBD (Komori
NFT. The cerebellar dentate nucleus, inferior olivary et al., 1998).
nuclei, red nucleus, oculomotor complex and colliculi The tau-positive inclusions in oligodendroglia,
are relatively spared but may show a variable degree known as coiled bodies, are argyrophilic structures
of neuronal loss and gliosis. The cerebellar cortex that are frequently present in the tauopathies. These
may also show focal Purkinje cell axonal torpedoes inclusions are distinct from the glial cytoplasmic
and Bergmann gliosis (Su et al., 2000; Piao et al., inclusions, considered the hallmark of MSA, which
2002). are immunoreactive for ubiquitin and a-synuclein,
Apolipoprotein E epsilon 4 (APOE4) is a risk factor but negative for tau (Feany and Dickson, 1996;
for Alzheimers disease pathology in all neurodegen- Rademakers et al., 2004).
360 N. P. STOVER ET AL.
The caudate, putamen and globus pallidus may 48.5.4. Tau mutations and ultrastructure
contain tau-immunoreactive lesions, most frequently
pretangles, variable nerve cell depletion, gliosis and Tau is the major component of the intracellular fila-
NFT. The ventrolateral nucleus of the thalamus, mentous deposits in the tauopathies. Tau pathology
subthalamic nucleus, red nucleus, raphe nuclei and in Alzheimers disease is thought to arise secondary
tegmental gray matter may have similar lesions but to b-amyloid pathology (Bancher et al., 1989; Goedert
are usually less severely involved. NFT are common and Jakes, 2005).
in the locus ceruleus and SN. The latter structure Some of the tau mutations have their primary effect
and the basal nucleus of Meynert may contain tau- at the protein level, reducing the ability of the tau pro-
immunoreactive pre-tangles (Sergeant et al., 1999). tein to interact with microtubules (Stanford et al.,
White-matter areas not contiguous with cortical 2004). Other mutations have their effect at the RNA
atrophy may also contain tau-immunoreactive thread- level, modifying the normal ratio of tau isoforms.
like lesions and coiled bodies. The internal capsule The splicing of mRNA transcripts from a single gene
and thalamic fasciculus often have many thread-like on chromosome 17 generates a total of six isoforms
processes (Forman et al., 2002). in adult human brain, with either three- or four-repeat
The minimal criteria for the pathologic diagnosis of (3R or 4R) domains, depending upon whether exon 10
CBD are cortical and striatal tau-positive neuronal and is expressed or not. Three of these isoforms contain
glial lesions, especially astrocytic plaques and thread- three microtubule-binding domains (3R) and three
like lesions in white and gray matter and neuronal contain four microtubule-binding domains (4R) (Dela-
loss in cortical regions and in the SN (Lantos, 2000; courte, 1999). Abnormal protein aggregates in the
Dickson et al., 2002) (Fig. 48.2). brain can be isolated as insoluble tau fractions and
analyzed for isoform composition and phosphorylation
state. Insoluble tau from CBD, PSP and AGD show
elevated 4R/3R ratios relative to Alzheimers disease
and controls (Togo et al., 2002; Tolnay et al., 2002;
de Silva et al., 2003; Katsuse et al., 2003; Miserez
et al., 2003). The tau protein may be hyperphosphory-
lated or abnormally phosphorylated (Arai et al., 2001a).
The mechanisms that lead to these tau abnormalities in
Alzheimers disease are still not well understood
(Ishizawa et al., 2002). In contrast, tau aggregates in
Picks disease consist of elevated levels of 3R tau.
Therefore, in these disorders there may be a fundamen-
tally different abnormality in tau processing, leading to
abnormal isoform composition, hyperphosphorylation
and protein aggregation.
In FTDP-17 two major types of mutation may
occur: missense mutations of exon 10 may lead to
mutant proteins that exhibit diminished affinity
for microtubules; alternatively, intronic mutations
(and some exon 10 mutations) stabilize the splicing
site so that higher levels of 4R tau are generated.
This latter situation is similar to what occurs in
PSP, CBD and AGD. Recently, a selective loss or
reduction in the levels of all six brain tau isoforms has
been described in a subset of sporadic cases with fronto-
temporal dementia and those cases are classified as
tau-deficiency tauopathies (Kawasaki et al., 1996;
Fig. 48.2. Immunohistochemical staining for phosphory-
lated tau shows a rim of cytoplasmic reactivity in a
Ksiezak-Reding et al., 1998; Godbolt et al., 2005).
ballooned neuron (large arrow) and more diffuse staining Other genes and/or factors may also be essential for
of another neuron (small arrow). Numerous thread-like the neurodegenerative process to occur, as shown by
tau-immunoreactive processes are seen in the surrounding FTDP-17 cases without tau mutation or deposition
neuropil. Courtesy of Dr. K.A. Roth, UAB Division of and with ubiquitin-positive inclusions (van Swieten
Neuropathology. et al., 2004).
CORTICOBASAL DEGENERATION 361
Genetic analysis of tau has revealed several poly- Table 48.2
morphisms in the coding and non-coding regions that
represent two ancestral haplotypes, referred to as H1 Pathologic findings of corticobasal degeneration
and H2. In PSP and CBD, H1 homozygosity (H1/H1) Macroscopic
is overexpressed and found in close to 90% of patients Asymmetric atrophy: frontoparietal and parasagittal
(Houlden et al., 2001), as opposed to only 60% of nor- Enlarged ventricles
mal controls. These findings suggest that genetic poly- Corpus callosum atrophy
morphisms of the tau gene somehow influence the risk Variable atrophy of subcortical structures (basal ganglia,
for developing the sporadic 4R diseases, CBD and PSP thalamus)
(Oliveira et al., 2004; Pittman et al., 2005; Tuite et al., Pallor in substantia nigra
2005). H1/H1 genotype has also been associated with Atrophy of white matter
PPA (Sobrido et al., 2003). Microscopic
Neuronal loss and gliosis
Tau epitopes are similar in CBD and PSP but differ-
Ballooned, achromatic neurons
ent from those in Alzheimers disease (Berry et al.,
Astrocytic plaques, mainly in the cortex
2004). Molecular heterogeneity and posttranslational Glial threads in the cerebral cortex, white matter, striatum,
modifications may influence the morphology and thalamus, cerebral peduncles, cerebellar dentate nucleus
stability of the abnormal filaments. Electron micro- and pons
scopy of the paired helical filaments show that they Neurofibrillary tangles
are wider in CBD cases and have a longer periodicity Substantia nigra cell loss
of the helical twist, compared with filaments found Immunocytochemical Tau pathology
in Alzheimers disease. CBD filaments are primarily Neurons: granular cytoplasmic deposits (pretangles) or
composed of single- or double-stranded, highly phos- diffuse aggregates.
phorylated polypeptides (15 or 29 nm wide and 62 or Tau pathology in basal ganglia
Glia
133 kDa mass per unit length). Additionally, CBD fila-
Astrocytic plaques
ments have a less stable ultrastructure because they are
Tufted astrocytes
composed of two distinct protofilaments. In contrast, Coiled bodies in oligodendroglia
Alzheimers disease has three highly phosphorylated Tau pathology in white matter
polypeptides of tau (Ksiezak-Reding et al., 1996)
(Table 48.2).

48.6. Imaging studies MRI in FTDP-17 and Picks disease patients typi-
cally demonstrates marked brain atrophy in the frontal
An important characteristic to be considered when and temporal regions, usually in a symmetrical fash-
evaluating the brain imaging of patients with suspected ion. Widening of the interpeduncular fossa secondary
CBD is asymmetric cortical atrophy, a feature that to degeneration of frontopontine fibers is another ima-
is quite distinctive from other neurodegenerative ging finding frequently seen in the different neurode-
syndromes in the differential diagnosis of CBD generative diseases that have prominent cerebral
(Gimenez-Roldan et al., 1994; Savoiardo, 2003). cortical atrophy (Hosaka et al., 2002). The MRI of
Serial evaluation of brain computed tomography patients with PSP shows atrophy in the midbrain and
(CT) or magnetic resonance imaging (MRI) scans over brainstem without asymmetrical cortical atrophy.
612-month intervals is generally more useful than an Patients with Alzheimers disease have diffuse tem-
isolated study. The abnormalities are best detected poral and hippocampal atrophy and in MSA of the oli-
with MRI, particularly in the fluid attenuated inversion vopontocerebellar atrophy type the atrophy is more
recovery (FLAIR) sequences. evident in the pons and cerebellum (Savoiardo et al.,
Volumetric studies of the cortex and hemispheric 2000).
surface show that the more atrophic areas in CBD Asymmetric atrophy of the corpus callosum in the
are the parietal lobe and the anterior, middle and pos- most affected side with correspondent ventricular dila-
teroinferior frontal lobe. Asymmetric cortical atrophy tation is frequently seen in confirmed cases of CBD.
is frequently seen, contralateral to the most affected Significant loss of callosal volume may appear in
side of the body in asymmetric presentations (Blin Alzheimers disease, PSP and FTD compared to
et al., 1992; Hu et al., 2005). The subcortical white controls. This morphologic change has a strong corre-
matter underlying the atrophic cortex also exhibits lation with the degree of cognitive impairment in
decreased volume (Doi et al., 1999; Groschel et al., patients with Alzheimers disease and CBD, but not
2004). See Fig. 48.3. in PD and PD with dementia cases. Neither the cortical
362 N. P. STOVER ET AL.
or corpus callosum atrophy or subcortical and periven- ganglia, that may represent overlap with other disorders
tricular white-matter signal changes on MRI are speci- (Gerhard et al., 2004; Henkel et al., 2004).
fic to CBD (Laureys et al., 1999; Josephs et al., 2004a). Brain parenchyma sonography is a relatively new
Functional imaging with positron emission tomo- technique using ultrasound to display tissue echogeni-
graphy (PET), single-photon emission computed city of the brain through the skull. The results of these
tomography (SPECT) and proton magnetic resonance studies need to be interpreted with caution, since the
spectroscopy (PMRS) in suspected CBD cases can cases do not have neuropathological confirmation.
provide a sensitive way to study changes in regional Sonography has proved to be reliable and sensitive to
cerebral blood flow (CBF), cerebral metabolism determine basal ganglia abnormalities in several
(oxygen or glucose) and dopamine receptor binding. pathologies, including the SN in PD, the caudate
SPECT study findings in CBD are variable and include nucleus in Huntingtons disease and the lenticular
asymmetric frontotemporal perfusion and a wide range nuclei in dystonia. A study attempted to discriminate
of prefrontal abnormalities. Other cases show bifrontal between CBD and PSP with ultrasound, but the cases
hypoperfusion with varying degrees of temporal and/or did not have pathologic confirmation and the results
parietal involvement (Eidelberg et al., 1991; Brooks, were inconclusive. CBD exhibits marked bilateral SN
2000). The presence of isolated, asymmetrical hypo- hyperechogenicity; this is reported as the characteristic
perfusion contralateral to the most affected side of finding in PD. According to some authors, cases of
the body is highly suggestive of CBD. PSP or MSA have normal echogenicity in the SN
PET studies demonstrate asymmetrical cortical and (Walter et al., 2004).
basal ganglia metabolism in patients clinically Studies using proton magnetic resonance neuro-
diagnosed with CBD. Cases of MSA show decreased spectroscopy correlated with EEG cartography con-
metabolism in the basal ganglia and cerebellum and firmed the asymmetrical features of CBD cases with
in PSP there is decreased medial frontal metabolism only clinical diagnosis (Vion-Dury et al., 2004).
and increased metabolism of the lentiform nucleus in
relation to that of patients with PD. This study created 48.7. Electrophysiological studies
a template to serve as a statistical aid in diagnosing
different neurodegenerative disorders based upon Electrophysiological studies may support the diagnosis
imaging findings. Studies of CBD show a relative of CBD and give us information about asymmetrical
reduction in both metabolic rate and glucose radiotra- dysfunction. The EEG is usually normal during the
cer uptake in several cortical areas, as well as the basal initial stages of the disease and may reveal non-speci-
ganglia and the insula, contralateral to the most fic asymmetric slowing in the more affected side in
affected side (Eckert et al., 2005). later stages. The non-specific diffuse slowing can be
18
Fluorodopa (18F-dopa) PET and SPECT labeling bilateral in advanced cases or in cases with predomi-
of the dopamine transporter by 2-carboxymethoxy nant cognitive dysfunction at presentation. Electrophy-
3(4-odophenyl)-tropane [123I] -CIT have demon- siological studies of the tremor with accelerometry and
strated homogeneous reduction in caudate and puta- electromyography (EMG) have distinguished tremor
men signal to as low as 25% of normal values in of CBD from the parkinsonian tremor: it is faster,
clinically diagnosed CBD, PSP and MSA, compared more irregular, more variable in amplitude, more evi-
with the asymmetric reduction in PD where binding dent during action and has a myoclonic component.
is selectively reduced in the putamen. The characteris- The myoclonus seen in CBD is a reflex myoclonus
tic pattern of asymmetrically reduced frontoparietal but the cortical spike is not always recognized.
cerebral cortical metabolism and/or CBF coupled with Results from studies of somatosensory evoked
equal reduction of 18F-dopa uptake in the caudate and potentials have not been conclusive in CBD. The
putamen provides strong supportive evidence in a N30 frontal component has been found to be absent
patient with a clinical diagnosis of CBD (Sawle unilaterally, bilaterally and with increased latency in
et al., 1989; Kreisler et al., 2005; Plotkin et al., 2005). patients with clinically probable CBD. Motor evoked
A PET study using a marker for peripheral benzo- potentials are reported as being prolonged in patients
diazepine-binding sites which are expressed by acti- with CBD (Okuda et al., 1998).
vated microglia addressed the role of microglial The P100 wave of the visual evoked potentials has
inflammatory responses in neurodegenerative disorders. been reported to not differ significantly between differ-
This study located the changes in the caudate nucleus, ent types of dementia and age-matched control subjects.
the putamen, the SN, the pons, the pre- and postcentral Brainstem auditory evoked potentials are reported to be
gyri and the frontal lobe. There was a broad heterogene- abnormal with prolonged P300 and N200 latencies in
ity within individual patients, especially in the basal CBD (Homma et al., 1996; Lu et al., 1998). Routine
CORTICOBASAL DEGENERATION 363
EMG is normal in early stages or may show changes CBD has been described as well (Benito-Leon et al.,
consistent with dystonia in the affected limbs. Nerve 2004). If a patient presents with a rapidly progressive
conduction studies may show focal or generalized CBD-like picture, prion-related disease should be con-
neuropathy that is usually subclinical. sidered (Cannard et al., 1998; Kovacs et al., 2002;
Josephs et al., 2004c; Kleiner-Fisman et al., 2004; Mor-
48.8. Laboratory studies eaud et al., 2005).
PSP is the disorder most likely to be confused with
Serum copper and ceruloplasmin levels and heavy- the motor presentation of CBD. PSP classically pre-
metal screens in blood and urine are normal in CBD sents with prominent axial rigidity, postural instability
patients. Cerebrospinal fluid (CSF) analysis in patients with early falls and abnormal vertical eye movements.
with CBD, PSP and Alzheimers disease show reduced Atypical features such as asymmetric onset, oculomo-
levels of b-amyloid peptide compared with control tor impairment that is only mild, focal dystonia and
subjects. Total tau protein levels are increased in involuntary limb levitation resembling the alien-limb
Alzheimers disease and the rest of the tauopathies phenomenon are the most frequent features that may
and may help to differentiate them from normal aging, cause confusion with CBD (Table 48.3).
depression, synucleinopathies and vascular dementia.
However, the tau level in the CSF is inadequate to dif- 48.10. Therapy
ferentiate among the different tauopathies (Arai et al.,
1997; Mitani et al., 1998; Paraskevas et al., 2005). Pharmacotherapy for CBD is currently of limited bene-
Levels of somatostatin were reported to be decreased fit. This may be explained by the widespread pathologi-
in some patients with autopsy confirmation of CBD. cal involvement of cortical and subcortical systems and
the lack of biochemical and molecular explanations for
48.9. Differential diagnosis and heterogeneity the pathophysiology of the disease. New therapeutic
approaches will be directed towards the development
The typical motor presentation of CBD is usually of compounds that modify, correct or block the underly-
sufficiently distinctive to allow a confident clinical diag- ing pathological mechanisms of CBD.
nosis. PD is the most common misdiagnosis early in the Some cases may show initial benefit with levodopa
course of the disease (Boeve et al., 2002; Doran et al., or with dopamine agonists, but this benefit is usually
2003; Kertesz et al., 2005). The best early clues that temporary. The primary treatment is symptomatic,
can help to distinguish CBD from idiopathic PD are the focused on alleviating the rigidity, dystonia, tremor,
lack of sustained beneficial response to dopaminergic myoclonus, neuropsychologic manifestations and other
medication and signs of cortical dysfunction, most nota- symptoms (Lang, 2005).
bly apraxia and/or cortical sensory impairment. Based on
pathological findings of cases diagnosed as CBD, the 48.10.1. Treatment of motor symptoms
differential diagnosis needs to include Picks disease
(Lang et al., 1994b), PSP (Wakabayashi and Takahashi, Improvement was reported in only 24% of clinically
2004), dementia lacking distinctive histology or tau- diagnosed CBD patients with carbidopa/levodopa
negative inclusions of the motor neuron disease type, treatment. The rest of the dopaminergic agents (bro-
FTDP-17 (Reed et al., 2001) and PPA (Ferrer et al., mocriptine, pergolide, pramipexole, ropinirole) may
2003). Primary dementing disorders should also be produce more side-effects with less clinical benefit in
included in the differential diagnosis of CBD. Other this group of patients. Clonazepam and other benzo-
diagnoses to consider are MSA, including olivoponto- diazepines have been the most beneficial agents for
cerebellar atrophy and striatonigral degeneration action tremor and myoclonus. Baclofen and tizanidine
(Adams et al., 1964), diffuse Lewy body disease, Alzhei- may improve rigidity. Anticholinergics have not been
mers disease with extrapyramidal features, AGD beneficial and they have been poorly tolerated due to
(Jellinger, 1998), parkinsonismdementiaamyotrophic cognitive side-effects and amantadine is of little or
lateral sclerosis complex, widespread cerebrovascular no benefit (Kompoliti et al., 1998). Botulinum toxin
disease, variants of Binswangers disease, progressive injections may be useful in the treatment of painful
multifocal leukodystrophies (Bhatia et al., 1996; Van focal limb dystonias, as well as in blepharospasm
Zanducke and Dehaene, 2000), variants of Azorean dis- (Cordivari et al., 2001; Muller et al., 2002b). Stereo-
ease (Nakano et al., 1972; Sachdev et al., 1982) and tactic surgery for relief of severe painful limb dystonia
hemiparkinsonismhemiatrophy (Giladi and Fahn, and parkinsonism has little or no benefit and is not
1992). CBD has presented as a Balints syndrome indicated in the treatment of atypical parkinsonian
(Mendez, 2000). A case of neurosyphilis resembling syndromes (Fazzini et al., 1995).
364 N. P. STOVER ET AL.
Table 48.3
Differential diagnosis of patients with neurodegenerative diseases with motor and cognitive dysfunction

Diagnosis Clinical features Features which suggest an alternative diagnosis

CBD Asymmetric bradykinetic/akinetic parkinsonism Prominent ocular involvement, axial dystonia,


with lack of response to dopaminergic aphasia, autonomic dysfunction, rest tremor
treatment, dystonia, cortical sensory loss,
apraxia, asymmetric dystonia or rigidity,
alien-limb phenomenon, myoclonus,
cognitive dysfunction
FTD/PPA/PiD Behavioral disturbances and problems with Gait disorder, early ocular involvement,
interpersonal interactions, disinhibition, hallucinations, prominent extrapyramidal
hyperorality, personality changes, dietary symptoms, levodopa responsiveness,
changes, speech and language symptoms, progressive apraxia without dementia
word-finding difficulties, memory loss
PSP Ophthalmoplegia (particularly vertical gaze Lack of ocular involvement, apraxia, cortical
paresis), early gait impairment, frequent sensory loss, prominent autonomic
falls, axial rigidity, dysarthria, dysphagia, dysfunction
suboptimal response to levodopa therapy
MSA Symmetric rigidity, absence of rest tremor, Early ocular involvement, apraxia, cortical
cerebellar and autonomic dysfunction, rapid signs, dementia
course, suboptimal response to levodopa
therapy, gait disorder
PD with dementia Asymmetric rest tremor, bradykinesia and Symmetric rigidity, early ocular involvement,
rigidity with dementia, memory loss, suboptimal response to levodopa therapy,
responsive to levodopa therapy apraxia, cortical sensory loss, prominent
autonomic dysfunction
AD with parkinsonism Early cognitive dysfunction (especially Parkinsonism present prior to onset of
memory loss), cortical dysfunction dementia, aphasia or apraxia without
(visuospatial deficits, aphasia, apraxia), significant memory loss, early gait
mild bradykinesia and rigidity impairment, axial rigidity, ophthalmoplegia,
alien-limb phenomenon
DLB Fluctuating symptoms, cognitive Axial rigidity or dystonia, early ocular
dysfunction, psychosis induced or involvement
worsened by dopaminergic therapy,
parkinsonism, gait disorder
CJD Rapid course of dementia, personality Protracted course, isolated parkinsonism or
changes, myoclonus, upper motor neuron cortical dysfunction, levodopa
signs, cerebellar or extrapyramidal signs responsiveness

CBD, corticobasal degeneration; FTD, frontotemporal dementia; PPA, primary progressive aphasia; PiD, Picks disease; PSP, progressive
supranuclear palsy; MSA, multiple system atrophy; PD, Parkinsons disease; AD, Alzheimers disease; DLB, dementia with Lewy bodies;
CJD, CreutzfeldtJakob disease.

48.10.2. Treatment of cognitive and including tricyclic compounds, may exacerbate confu-
neuropsychiatric symptoms sion. Small doses of atypical neuroleptic medications
such as quetiapine, olanzapine or clozapine may be help-
There is no clear evidence that treatment of cognitive ful for paranoid delusions, psychotic behavior, agitation,
dysfunction with medications that enhance cholinergic irritability and sleep problems. The use of typical antipsy-
neurotransmission has much benefit; however, they may chotic medications such as haloperidol may worsen motor
be considered in early stages of the disease. Depression symptoms of CBD and are not recommended.
and obsessive-compulsive symptomatology may be trea- Small doses of sedative hypnotic agents may be help-
ted effectively with selective serotonin reuptake inhibitors ful for insomnia, but they may also exacerbate confusion
(SSRIs), but cautious titration and low doses are recom- and agitation. Clonazepam is usually helpful and agents
mended because these medications can exacerbate agita- such as diphenhydramine, chloral hydrate and zolpidem
tion. Antidepressants with anticholinergic side-effects, may be useful as well, but they should be used cautiously.
CORTICOBASAL DEGENERATION 365
48.10.3. Treatment of other symptoms The urinary symptoms of CBD include urgency and
frequency and they may improve with the use of hyos-
Gastrointestinal symptoms in CBD patients include cyamine, tolterodine or oxybutynin. Close observation
hypersalivation, dysphagia, nausea and constipation. for central and peripheral side-effects is warranted
Excessive salivation may respond to small doses of when using these or related medications.
anticholinergic therapy, but side-effects are a limiting Symptomatic orthostatic hypotension, although
factor in most cases. more frequently seen in MSA, may be treated with
The goal of treatment of the dysphagia is to main- fludrocortisone or midodrine.
tain safe and efficient nutrition and hydration. Evalua- Other aspects of patient care, not involving pharma-
tion with a barium swallow study may be necessary. cotherapy, can be of special importance for these patients
Treatment includes dietary modifications, positional and their families (Litvan, 2001). Physiotherapy is very
changes, swallowing maneuvers and exercises and sur- helpful for maintenance of mobility and prevention of
gical interventions. Selection of foods with a consis- contractures. Pain related to dystonic posturing can be
tency that facilitates swallowing is critical. If patients lessened by maintenance of good range of motion and
are not able to ingest enough food to meet nutritional occasionally splinting can be helpful. Occupational ther-
requirements, percutaneous feeding gastrostomy tube apy can help patients maintain some degree of functional
placement may be necessary. The decision to place a independence by providing specially made devices such
gastrostomy in a patient with a chronically progressive as eating utensils with large handles. Speech therapy
neurodegenerative disease must be handled on an indi- may offer practical suggestions and exercises to optimize
vidual basis, keeping in mind the wishes of the patient speech function and guard against aspiration secondary
or the patients surrogate. to swallowing difficulty. Good home care assistance
Constipation in parkinsonism is due to colonic can help prolong the time a patient remains at home
hypomotility, colonic outlet dysfunction or both. Con- before requiring nursing-home placement.
stipation has also been associated with pelvic floor Despite our best therapeutic efforts, regardless of
dystonia. The use of stool softeners, increased fluid whether it presents with motor or cognitive symptoms,
intake, foods rich in fiber and laxatives may be benefi- CBD generally progresses to a state of bilateral rigid
cial. Polyethylene glycol is another alternative for the immobility due to increased tone or apraxia and patients
treatment of severe constipation. usually die from aspiration pneumonia or urosepsis.

Fig. 48.3. (A) and (B) T1 and T2-weighted brain images of a patient with corticobasal degeneration showing the asymmetric
cortical atrophy, contralateral to the most affected side
366 N. P. STOVER ET AL.
References Boeve BF, Maraganore DM, Parisi JE et al. (2002). Cortico-
basal degeneration and frontotemporal dementia presenta-
Adams RD, Van Bogaert L, Vander Eecken H (1964). Strao- tions in a kindred with nonspecific histopathology.
nigral degeneration. J Neuropathol Exp Neurol 23: Dement Geriatr Cogn Disord 13 (2): 8090.
584608. Brooks DJ (2000). Corticobasal degeneration. Functional
Arai H, Morikawa Y, Higuchi M et al. (1997). Cerebrospinal imaging studies in corticobasal degeneration. In: I Litvan,
fluid tau levels in neurodegenerative diseases with distinct CG Goetz, AE Lang (Eds.), Corticobasal Degeneration
tau-related pathology. Biochem Biophys Res Commun Advances in Neurology, Vol. 82. Williams & Wilkins,
236 (2): 262264. Philadelphia, pp. 209215.
Arai T, Ikeda K, Akiyama H et al. (2001a). Distinct isoforms Brown J, Lantos PL, Rossor MN (1998). Familial dementia
of tau aggregated in neurons and glial cells in brains of lacking specific pathological features presenting with
patients with Picks disease, corticobasal degeneration clinical features of corticobasal degeneration. J Neurol
and progressive supranuclear palsy. Acta Neuropathol Neurosurg Psychiatry 65 (4): 600603.
(Berl) 101 (2): 167173. Brunt ER, van Weerden TW, Pruim J et al. (1995). Unique
Arai Y, Yamazaki M, Mori O et al. (2001b). Alpha-synuclein- myoclonic pattern in corticobasal degeneration. Mov
positive structures in cases with sporadic Alzheimers Disord 10 (2): 132142.
disease: morphology and its relationship to tau aggregation. Buee L, Delacourte A (1999). Comparative biochemistry of
Brain Res 888 (2): 287296. tau in progressive supranuclear palsy, corticobasal degen-
Armstrong RA, Cairns NJ, Lantos PL (2001). What does the eration, FTDP-17 and Picks disease. Brain Pathol 9 (4):
study of the spatial patterns of pathological lesions tell us 681693.
about the pathogenesis of neurodegenerative disorders? Cairns NJ, Lee VM, Trojanowski JQ (2004). The cytoskeleton
Neuropathology 21 (1): 112. in neurodegenerative diseases. J Pathol 204 (4): 438449.
Bak TH, Rogers TT, Crawford LM et al. (2005). Cognitive Cannard KR, Galvez-Jimenez N, Watts RL (1998). Creutz-
bedside assessment in atypical parkinsonian syndromes. feldt-Jakob disease presenting and evolving as rapidly pro-
J Neurol Neurosurg Psychiatry 76 (3): 420422. gressive corticobasal degeneration. Neurology 50 (4):
Ball JA, Lantos PL, Jackson M et al. (1993). Alien hand sign A95.
in association with Alzheimers histopathology. J Neurol Carella F, Scaioli V, Franceschetti S et al. (1991). Focal reflex
Neurosurg Psychiatry 56 (9): 10201023. myoclonus in corticobasal degeneration. Funct Neurol
Ballan G, Tison F (1997). A historical case of probable cor- 6 (2): 165170.
ticobasal degeneration? Mov Disord 12 (6): 10731074. Carella F, Ciano C, Panzica F et al. (1997). Myoclonus in
Bancher C, Brunner C, Lassmann H et al. (1989). Accumula- corticobasal degeneration. Mov Disord 12 (4): 598603.
tion of abnormally phosphorylated tau precedes the forma- Caselli RJ, Stelmach GE, Caviness JN et al. (1999). A kine-
tion of neurofibrillary tangles in Alzheimers disease. matic study of progressive apraxia with and without
Brain Res 477 (12): 9099. dementia. Mov Disord 14 (2): 276287.
Banks G, Short P, Martinez J et al. (1989). The alien hand Castellani R, Smith MA, Richey PL et al. (1995). Evidence
syndrome. Clinical and postmortem findings. Arch Neurol for oxidative stress in Pick disease and corticobasal degen-
46 (4): 456459. eration. Brain Res 696 (12): 268271.
Benito-Leon J, Alvarez-Linera J, Louis ED (2004). Neurosy- Caviness JN (2003). Myoclonus and neurodegenerative
philis masquerading as corticobasal degeneration. Mov diseasewhats in a name? Parkinsonism Relat Disord
Disord 19 (11): 13671370. 9 (4): 185192.
Bergeron C, Davis A, Lang AE (1998). Corticobasal gang- Charcot JM (18871888). Lecons du mardi: Policlinique de
lionic degeneration and progressive supranuclear palsy la Salpetriere Bureaux du Progres Medical, Paris.
presenting with cognitive decline. Brain Pathol 8 (2): Charcot JM, Vulpian A (18611862). De la paralysie agi-
355365. tante. Gazette Hebdomadaire de Medecine et de Chirurgie
Berry RW, Sweet AP, Clark FA et al. (2004). Tau epitope 8: 765767, 8: 816820, 9: 5459.
display in progressive supranuclear palsy and corticobasal Chen R, Ashby P, Lang AE (1992). Stimulus-sensitive myo-
degeneration. J Neurocytol 33 (3): 287295. clonus in akinetic-rigid syndromes. Brain 115: 18751888.
Bhatia KP, Morris JH, Frackowiak RS (1996). Primary Collins SJ, Ahlskog JE, Parisi JE et al. (1995). Progressive
progressive multifocal leukoencephalopathy presenting as supranuclear palsy: neuropathologically based diagnostic
an extrapyramidal syndrome. J Neurol 243 (1): 9195. clinical criteria. J Neurol Neurosurg Psychiatry 58 (2):
Black SE (2000). Aphasia in corticobasal degeneration. Adv 167173.
Neurol 82: 123133. Cordato NJ, Halliday GM, McCann H et al. (2001). Cortico-
Blin J, Vidailhet MJ, Pillon B et al. (1992). Corticobasal degen- basal syndrome with tau pathology. Mov Disord 16 (4):
eration: decreased and asymmetrical glucose consumption as 656667.
studied with PET. Mov Disord 7 (4): 348354. Cordivari C, Misra VP, Catania S et al. (2001). Treatment of
Boeve BF, Silber MH, Ferman TJ et al. (2001). Association dystonic clenched fist with botulinum toxin. Mov Disord
of REM sleep behavior disorder and neurodegenerative 16 (5): 907913.
disease may reflect an underlying synucleinopathy. Mov Cummings JL, Litvan I (2000). Corticobasal degeneration.
Disord 16 (4): 622630. Neuropsychiatric aspects of corticobasal degeneration. In: I
CORTICOBASAL DEGENERATION 367

Litvan, CG Goetz, AE Lang (Eds.), Corticobasal Degenera- Eckert T, Barnes A, Dhawan V et al. (2005). FDG PET in
tion. Advances in Neurology, Vol. 82. Williams & Wilkins, the differential diagnosis of parkinsonian disorders. Neu-
Philadelphia, pp. 147152. roimage 26 (3): 912921.
Cummings JL, Mega M, Gray K et al. (1994). The neuropsy- Eidelberg D, Dhawan V, Moeller JR et al. (1991). The meta-
chiatric inventory: comprehensive assessment of psycho- bolic landscape of cortico-basal ganglionic degeneration:
pathology in dementia. Neurology 44 (12): 23082314. regional asymmetries studied with positron emission
Delacourte A (1999). Biochemical and molecular characteri- tomography. J Neurol Neurosurg Psychiatry 54: 856862.
zation of neurofibrillary degeneration in frontotemporal Fazzini E, Dogali M, Beric A et al. (1995). The effects of
dementias. Dement Geriatr Cogn Disord 10 (Suppl 1): unilateral ventral posterior medial pallidotomy in patients
7579. with Parkinsons disease and Parkinsons plus syndromes.
Denes G (2002). Comparison of apraxia in corticobasal degen- In: WC Koller, G Paulson (Eds.), Therapy of Parkinsons
eration and progressive supranuclear palsy. Neurology Disease. Marcel Dekker, New York, pp. 353379.
58 (8): 1317. Feany MB, Dickson DW (1996). Neurodegenerative disor-
DePold Hohler A, Ransom BR, Chun MR et al. (2003). The ders with extensive tau pathology: a comparative study
youngest reported case of corticobasal degeneration. and review. Ann Neurol 40 (2): 139148.
Parkinsonism Relat Disord 10 (1): 4750. Feany MB, Mattiace LA, Dickson DW (1996). Neuropatholo-
De Renzi E, Lucchelli F (1988). Ideational apraxia. Brain gic overlap of progressive supranuclear palsy, Picks dis-
111 (Pt 5): 11731185. ease and corticobasal degeneration. J Neuropathol Exp
de Silva R, Lashley T, Gibb G et al. (2003). Pathological Neurol 55 (1): 5367.
inclusion bodies in tauopathies contain distinct comple- Ferrer I, Hernandez I, Boada M et al. (2003). Primary pro-
ments of tau with three or four microtubule-binding repeat gressive aphasia as the initial manifestation of corticobasal
domains as demonstrated by new specific monoclonal degeneration and unusual tauopathies. Acta Neuropathol
antibodies. Neuropathol Appl Neurobiol 29 (3): 288302. (Berl) 106 (5): 419435.
Dickson DW (1999a). Neuropathologic differentiation of Fisher CM (2000). Alien hand phenomena: a review with the
progressive supranuclear palsy and corticobasal degenera- addition of six personal cases. Can J Neurol Sci 27 (3):
tion. J Neurol 246 (Suppl 2): II6II15. 192203.
Dickson DW (1999b). Tau and synuclein and their role in Forman MS, Lee VM, Trojanowski JQ (2000). New insights
neuropathology. Brain Pathol 9 (4): 657661. into genetic and molecular mechanisms of brain degen-
Dickson DW (2003). Neuropathology of parkinsonism. In: eration in tauopathies. J Chem Neuroanat 20 (34): 225244.
R Pahwa, KE Lyons, WC Koller (Eds.), Handbook Forman MS, Zhukareva V, Bergeron C et al. (2002). Signature
of Parkinsons Disease. Marcel Dekker, Inc, New York, tau neuropathology in gray and white matter of corticobasal
pp. 203220. degeneration. Am J Pathol 160 (6): 20452053.
Dickson DW, Litvan I (2003). Corticobasal degeneration. Frattali CM, Sonies BC (2000). Speech and swallowing
In: DW Dickson, (Eds.), Neurodegeneration: The Molecu- disturbances in corticobasal degeneration. Adv Neurol
lar Pathology of Dementia and Movement Disorder. Neu- 82: 153160.
ropath Press, Basel, Switzerland, pp. 115123. Frattali CM, Grafman J, Patronas N et al. (2000). Language
Dickson DW, Feany MB, Yen SH et al. (1996). Cytoskeletal disturbances in corticobasal degeneration. Neurology
pathology in non-Alzheimer degenerative dementia: new 54 (4): 990992.
lesions in diffuse Lewy body disease, Picks disease, and Fujino Y, Delucia MW, Davies P et al. (2004). Ballooned
corticobasal degeneration. J Neural Transm Suppl 47: neurones in the limbic lobe are associated with Alzheimer
3146. type pathology and lack diagnostic specificity. Neuro-
Dickson DW, Bergeron C, Chin SS et al. (2002). Office pathol Appl Neurobiol 30 (6): 676682.
of Rare Diseases of the National Institutes of Health. Gerhard A, Watts J, Trender-Gerhard I et al. (2004). In vivo
Office of Rare Diseases neuropathologic criteria for corti- imaging of microglial activation with [11C] (R)-PK11195
cobasal degeneration. J Neuropathol Exp Neurol 61 (11): PET in corticobasal degeneration. Mov Disord 19 (10):
935946. 12211226.
Di Maria E, Tabaton M, Vigo T et al. (2000). Corticobasal Giannakopoulos P, Hof PR, Bouras C (1995). Dementia lack-
degeneration shares a common genetic background ing distinctive histopathology: clinicopathological evalua-
with progressive supranuclear palsy. Ann Neurol 47 (3): tion of 32 cases. Act Neuropathol (Berl) 89 (4): 346355.
374377. Gibb WR, Luthert PJ, Marsden CD (1989). Corticobasal
Doi T, Iwasa K, Makifuchi T et al. (1999). White matter degeneration. Brain 112: 11711192.
hyperintensities on MRI in a patient with corticobasal Giladi N, Fahn S (1992). Hemiparkinsonism-hemiatrophy
degeneration. Acta Neurol Scand 99 (3): 199201. syndrome may mimic early-stage cortical-basal ganglionic
Doody RS, Jankovic J (1992). The alien hand and related degeneration. Mov Disord 7 (4): 384385.
signs. J Neurol Neurosurg Psychiatry 55 (9): 806810. Gimenez-Roldan S, Mateo D, Benito C et al. (1994). Progressive
Doran M, du Plessis DG, Enevoldson TP et al. (2003). Patho- supranuclear palsy and corticobasal ganglionic degeneration:
logical heterogeneity of clinically diagnosed corticobasal differentiation by clinical features and neuroimaging techni-
degeneration. J Neurol Sci 216 (1): 127134. ques. J Neural Transm Suppl 42: 7990.
368 N. P. STOVER ET AL.

Godbolt AK, Josephs KA, Revesz T et al. (2005). Sporadic Hosaka K, Ishii K, Sakamoto S et al. (2002). Voxel-based
and familial dementia with ubiquitin-positive tau-negative comparison of regional cerebral glucose metabolism
inclusions: clinical features of one histopathological between PSP and corticobasal degeneration. J Neurol Sci
abnormality underlying frontotemporal lobar degenera- 199 (12): 6771.
tion. Arch Neurol 62 (7): 10971101. Houlden H, Baker M, Morris HR et al. (2001). Corticobasal
Goedert M, Jakes R (2005). Mutations causing neurodegenera- degeneration and progressive supranuclear palsy share a
tive tauopathies. Biochim Biophys Acta 1739 (23): 240250. common tau haplotype. Neurology 56 (12): 17021706.
Goedert M, Ghetti B, Spillantini MG (2000). Tau gene muta- Hu WT, Josephs KA, Ahlskog JE et al. (2005). MRI correlates
tions in frontotemporal dementia and parkinsonism linked of alien leg-like phenomenon in corticobasal degeneration.
to chromosome 17 (FTDP-17). Their relevance for under- Mov Disord 20 (7): 870873.
standing the neurodegenerative process. Ann NY Acad Sci Ikeda K, Akiyama H, Haga C et al. (1994). Argyrophilic
920: 7483. thread-like structure in corticobasal degeneration and
Goldberg G, Bloom KK (1990). The alien hand sign. Locali- supranuclear palsy. Neurosci Lett 174 (2): 157159.
zation, lateralization and recovery. Am J Phys Med Reha- Ikeda K, Akiyama H, Kondo H et al. (1995). A study
bil 69 (5): 228238. of dementia with argyrophilic grains. Possible cytoske-
Graham NL, Bak TH, Hodges JR (2003a). Corticobasal degen- letal abnormality in dendrospinal portion of neurons
eration as a cognitive disorder. Mov Disord 18 (11): and oligodendroglia. Acta Neuropathol (Berl) 89 (5):
12241232. 409414.
Graham NL, Bak T, Patterson K et al. (2003b). Language Ingelson M, Fabre SF, Lilius L et al. (2001). Increased risk
function and dysfunction in corticobasal degeneration. for frontotemporal dementia through interaction between
Neurology 61 (4): 493499. tau polymorphisms and apolipoprotein E epsilon4.
Green J (2000). Neuropsychological profiles in corticobasal Neuroreport 12 (5): 905909.
degeneration. In: J Green (Ed.), Neuropsychological Evalua- Iriarte J, Alegre M, Arbizu J et al. (2001). Unilateral periodic
tion of the Older Adult. Academic Press, San Diego, CA, limb movements during sleep in corticobasal degenera-
pp. 142143. tion. Mov Disord 16 (6): 11801183.
Grimes DA, Lang AE, Bergeron CB (1999). Dementia as the Ishiwata T, Suzuki A, Mochizuki H et al. (2000). [Progres-
most common presentation of cortical-basal ganglionic sive limb-kinetic apraxia with myoclonus focal atrophy
degeneration. Neurology 53: 19691974. in the postcentral gyrus and the supplementary motor
Groschel K, Hauser TK, Luft A et al. (2004). Magnetic reso- area.] No To Shinkei 52 (10): 925928.
nance imaging-based volumetry differentiates progressive Ishizawa K, Dickson DW (2001). Microglial activation par-
supranuclear palsy from corticobasal degeneration. Neuro- allels system degeneration in progressive supranuclear
image 21 (2): 714724. palsy and corticobasal degeneration. J Neuropathol Exp
Hachinski V (1997). Frontotemporal degeneration, Pick dis- Neurol 60 (6): 647657.
ease, and corticobasal degeneration. Three entities or 1? Ishizawa T, Ko LW, Cookson N et al. (2002). Selective neu-
Arch Neurol 54 (11): 1429. rofibrillary degeneration of the hippocampal CA2 sector is
Halliday GM, Davies L, McRitchie DA et al. (1995). Ubiqui- associated with four-repeat tauopathies. J Neuropathol
tin-positive achromatic neurons in corticobasal degenera- Exp Neurol 61 (12): 10401047.
tion. Acta Neuropathol (Berl) 90 (1): 6875. Iwasaki Y, Yoshida M, Hattori M et al. (2005). Widespread
Hamilton SR (2000). Neuro-ophthalmology of movement spinal cord involvement in corticobasal degeneration. Acta
disorders. Curr Opin Ophthalmol 11 (6): 403407. Neuropathol (Berl) 109 (6): 632638.
Hanger DP, Gibb GM, de Silva R et al. (2002). The complex Jacobs DH, Adair JC, Heilman KM (1994). Visual grasp in
relationship between soluble and insoluble tau in tauopa- corticobasal degeneration. Ann Neurol 36 (4): 679680.
thies revealed by efficient dephosphorylation and specific Jacobs DH, Adair JC, Macauley B et al. (1999). Apraxia in
antibodies. FEBS Lett 531 (3): 538542. corticobasal degeneration. Brain Cogn 40 (2): 336354.
Hargrave R, Rafal R (1998). Depression in corticobasal Jellinger KA (1998). Dementia with grains (argyrophilic
degeneration. Psychosomatics 39 (5): 481482. grain disease). Brain Pathol 8 (2): 377386.
Hawkes C (2003). Olfaction in neurodegenerative disorder. Josephs KA, Tang-Wai DF, Edland SD et al. (2004a). Cor-
Mov Disord 18 (4): 364372. relation between antemortem magnetic resonance imaging
Henkel K, Karitzky J, Schmid M et al. (2004). Imaging of findings and pathologically confirmed corticobasal degen-
activated microglia with PET and [11C]PK 11195 in cor- eration. Arch Neurol 61 (12): 18811884.
ticobasal degeneration. Mov Disord 19 (7): 817821. Josephs KA, Tsuboi Y, Cookson N et al. (2004b). Apolipo-
Homma A, Harayama H, Kondo H et al. (1996). P300 find- protein E epsilon 4 is a determinant for Alzheimer-type
ings in patients with corticobasal degeneration. Brain and pathologic features in tauopathies, synucleinopathies, and
Nerve 48 (10): 925929. frontotemporal degeneration. Arch Neurol 61 (10):
Horoupian DS, Wasserstein PH (1999). Alzheimers disease 15791584.
pathology in motor cortex in dementia with Lewy bodies Josephs KA, Tsuboi Y, Dickson DW (2004c). Creutzfeldt-
clinically mimicking corticobasal degeneration. Acta Jakob disease presenting as progressive supranuclear
Neuropathol (Berl) 98 (3): 317322. palsy. Eur J Neurol 11 (5): 343346.
CORTICOBASAL DEGENERATION 369

Kaida K, Takeda K, Nagata N et al. (1998). Alzheimers dis- Ksiezak-Reding H, Tracz E, Yang LS et al. (1996). Ultrastruc-
ease with asymmetric parietal lobe atrophy: a case report. tural instability of paired helical filaments from corticobasal
J Neurol Sci 160 (1): 9699. degeneration as examined by scanning transmission electron
Kasashima S, Oda Y (2003). Cholinergic neuronal loss in the microscopy. Am J Pathol 149 (2): 639651.
basal forebrain and mesopontine tegmentum of progres- Ksiezak-Reding H, Yang G, Simon M et al. (1998). Assembled
sive supranuclear palsy and corticobasal degeneration. tau filaments differ from native paired helical filaments as
Acta Neuropathol (Berl) 105 (2): 117124. determined by scanning transmission electron microscopy
Katsuse O, Iseki E, Arai T et al. (2003). 4-Repeat tauopathy (STEM). Brain Res 814 (12): 8698.
sharing pathological and biochemical features of cortico- Kumar R, Bergeron C, Pollanen MS et al. (1998). Corticalba-
basal degeneration and progressive supranuclear palsy. sal ganglionic degeneration. In: J Jankovic, E Tolosa
Acta Neuropathol (Berl) 106 (3): 251260. (Eds.), Parkinsons Disease and Movement Disorders,
Kawasaki K, Iwanaga K, Wakabayashi K et al. (1996). Corti- 3rd edn. Williams & Wilkins, Baltimore, pp. 297316.
cobasal degeneration with neither argyrophilic inclusions Kurz AF (2005). Uncommon neurodegenerative causes of
nor tau abnormalities: a new subgroup? Acta Neuropathol dementia. Int Psychogeriatr 17 (Suppl 1): S35S49.
91 (2): 140144. Lalive PH, Personeni O, Slosman DO et al. (2000). Lower
Kertesz A, Munoz D (2004). Relationship between fronto- limb corticobasal degeneration. Eur Neurol 44 (4):
temporal dementia and corticobasal degeneration/progres- 248249.
sive supranuclear palsy. Dement Geriatr Cogn Disord Lang AE (1992). Cortico basal ganglionic degeneration pre-
17 (4): 282286. senting with progressive loss of speech output and orofa-
Kertesz A, Martinez-Lage P, Davidson W et al. (2000). The cial dyspraxia. J Neurol Neurosurg Psychiatry 55: 1101.
corticobasal degeneration syndrome overlaps progressive Lang AE (2000). Parkinsonism in corticobasal degeneration.
aphasia and frontotemporal dementia. Neurology 55 (9): In: I Litvan, CG Goetz, AE Lang (Eds.), Corticobasal
13681375. Degeneration. Advances in Neurology, Vol. 82. Williams
Kertesz A, McMonagle P, Blair M et al. (2005). The evolu- & Wilkins, Philadelphia, pp. 8389.
tion and pathology of frontotemporal dementia. Brain Lang AE (2005). Treatment of progressive supranuclear
128 (Pt 9): 19962005. palsy and corticobasal degeneration. Mov Disord 20 (Suppl
Kikuchi H, Doh-ura K, Kira J et al. (1999). Preferential neu- 12): S83S91.
rodegeneration in the cervical spinal cord of progressive Lang AE, Riley DE, Bergeron C (1994a). Cortical-basal
supranuclear palsy. Acta Neuropathol (Berl) 97 (6): ganglionic degeneration. In: DB Calne, (Eds.), Neurode-
577584. generative Diseases. WB Saunders, Philadelphia, pp.
Kimura K, Tachibana N, Aso T et al. (1997). Subclinical 877894.
REM sleep behavior disorder in a patient with corticobasal Lang AE, Bergeron C, Pollanen MS et al. (1994b). Parietal
degeneration. Sleep 20 (10): 891894. Picks disease mimicking cortical-basal ganglionic degen-
Kleiner-Fisman G, Bergeron C, Lang AE (2004). Presenta- eration. Neurology 44: 14361440.
tion of Creutzfeldt-Jakob disease as acute corticobasal Lantos PL (2000). Diagnostic criteria for corticobasal degen-
degeneration syndrome. Mov Disord 19 (8): 948949. eration. J Neurol Neurosurg Psychiatry 69 (5): 705706.
Komori T (1999). Tau-positive glial inclusions in progres- Laureys S, Salmon E, Garraux G et al. (1999). Fluorodopa
sive supranuclear palsy, corticobasal degeneration and uptake and glucose metabolism in early stages of cortico-
Picks disease. Brain Pathol 9 (4): 663679. basal degeneration. J Neurol 246 (12): 11511158.
Komori T, Arai N, Oda M et al. (1997). Morphologic dif- Lee VM, Goedert M, Trojanowski JQ (2001). Neurodegen-
ference of neuropil threads in Alzheimers disease, erative tauopathies. Annu Rev Neurosci 24: 11211159.
corticobasal degeneration and progressive supranuclear Leiguarda R, Merello M, Balej J (2000). Apraxia in cortico-
palsy: a morphometric study. Neurosci Lett 233 (23): basal degeneration. Adv Neurol 82: 103121.
8992. Leiguarda RC, Merello M, Nouzeilles MI et al. (2003).
Komori T, Arai N, Oda M et al. (1998). Astrocytic plaques Limb-kinetic apraxia in corticobasal degeneration: clinical
and tufts of abnormal fibers do not coexist in corticobasal and kinematic features. Mov Disord 18 (1): 4959.
degeneration and progressive supranuclear palsy. Acta Lerner A, Friedland R, Riley D et al. (1992). Dementia with
Neuropathol (Berl) 96 (4): 401408. pathological findings of corticobasal ganglionic degenera-
Kompoliti K, Goetz CG, Boeve BF et al. (1998). Clinical tion. Ann Neurol 32: 271.
presentation and pharmacological therapy in corticobasal Levy ML, Miller BL, Cummings JL et al. (1996). Alzheimer
degeneration. Arch Neurol 55 (7): 957961. disease and frontotemporal dementias. Behavioral distinc-
Kovacs GG, Trabattoni G, Hainfellner JA et al. (2002). Muta- tions. Arch Neurol 53 (7): 687690.
tions of the prion protein gene phenotypic spectrum. Litvan I (1997). Progressive supranuclear palsy and cortico-
J Neurol 249 (11): 15671582. basal degeneration. Baillieres Clin Neurol 6 (1): 167185.
Kreisler A, Defebvre L, Lecouffe P et al. (2005). Corticobasal Litvan I (1999). Recent advances in atypical parkinsonian
degeneration and Parkinsons disease assessed by HmPaO disorders. Curr Opin Neurol 12 (4): 441446.
SPECT: the utility of factorial discriminant analysis. Mov Litvan I (2001). Therapy and management of frontal lobe
Disord 20 (11): 14311438. dementia patients. Neurology 56 (11 Suppl 4): S41S45.
370 N. P. STOVER ET AL.

Litvan I, Agid Y, Jankovic J et al. (1996). Accuracy of clin- eration clinical and MRI data of a case. J Neurol
ical criteria for the diagnosis of progressive supranuclear 252 (10): 12831284.
palsy (Steele-Richardson-Olszewski syndrome). Neurol- Moretti R, Torre P, Antonello RM et al. (2005). Cognitive
ogy 46 (4): 922930. impairment in the lateralized phenotype of corticobasal
Litvan I, Agid Y, Goetz C et al. (1997). Accuracy of the clin- degeneration. Dement Geriatr Cogn Disord 20 (23):
ical diagnosis of corticobasal degeneration: a clinicopatho- 158162.
logic study. Neurology 48 (1): 119125. Mori H, Oda M, Mizuno Y (1996). Cortical ballooned neu-
Litvan I, Cummings JL, Mega M (1998). Neuropsychiatric rons in progressive supranuclear palsy. Neurosci Lett
features of corticobasal degeneration. J Neurol Neurosurg 209 (2): 109112.
Psychiatry 65 (5): 717721. Muller A, Mungersdorf M, Reichmann H et al. (2002a).
Litvan I, Grimes DA, Lang AE et al. (1999). Clinical fea- Olfactory function in Parkinsonian syndromes. J Clin Neu-
tures differentiating patients with postmortem confirmed rosci 9 (5): 521524.
progressive supranuclear palsy and corticobasal degenera- Muller J, Wenning GK, Verny M et al. (2001). Progression
tion. J Neurol 246 (Suppl 2): II1II5. of dysarthria and dysphagia in postmortem-confirmed par-
Litvan I, Bhatia KP, Burn DJ et al. (2003). Movement kinsonian disorders. Arch Neurol 58 (2): 259264.
Disorders Society Scientific Issues Committee report: Muller J, Wenning GK, Wissel J et al. (2002b). Botulinum
SIC Task Force appraisal of clinical diagnostic criteria toxin treatment in atypical parkinsonian disorders asso-
for Parkinsonian disorders. Mov Disord 18 (5): 467486. ciated with disabling focal dystonia. J Neurol 249 (3):
Lleo A, Rey MJ, Castellvi M et al. (2002). Asymmetric myo- 300304.
clonic parietal syndrome in a patient with Alzheimers Nakano KK, Dawson DM, Spence A (1972). Machado dis-
disease mimicking corticobasal degeneration. Neurologia ease. A hereditary ataxia in Portuguese emigrants to Mas-
17 (4): 223226. sachusetts. Neurology 22: 4955.
Lu CS, Ikeda A, Terada K et al. (1998). Electrophysiological Nakazato Y, Hirato J, Ishida Y et al. (1990). Swollen cortical
studies of early stage corticobasal degeneration. Mov neurons in Creutzfeldt-Jakob disease contain a phosphory-
Disord 13 (1): 140146. lated neurofilament epitope. J Neuropathol Exp Neurol 49:
Mackenzie IR, Hudson LP (1995). Achromatic neurons in 197205.
the cortex of progressive supranuclear palsy. Acta Neuro- Obeso JA, Rothwell JC, Lang AE et al. (1983). Myoclonic
pathol (Berl) 90 (6): 615619. dystonia. Neurology 33: 825830.
Maraganore DM, Ahlskog JE, Petersen RC (1992). Progres- Obeso JA, Rothwell JC, Marsden CD (1985). The spectrum
sive asymmetric rigidity with apraxia: a distinctive clinical of cortical myoclonus. From focal reflex jerks to sponta-
entity. Mov Disord 7 (1): 80. neous motor epilepsy. Brain 108: 193124.
Mendez MF (2000). Corticobasal ganglionic degeneration Oide T, Ohara S, Yazawa M et al. (2002). Progressive supra-
with Balints syndrome. J Neuropsychiatry Clin Neurosci nuclear palsy with asymmetric tau pathology presenting
12 (2): 273275. with unilateral limb dystonia. Acta Neuropathol (Berl)
Merians AS, Clark M, Poizner H et al. (1999). Apraxia dif- 104 (2): 209214.
fers in corticobasal degeneration and left-parietal stroke: Okuda B, Tachibana H (1994). The nature of apraxia in cor-
a case study. Brain Cogn 40 (2): 314335. ticobasal degeneration. J Neurol Neurosurg Psychiatry
Mima T, Nagamine T, Ikeda A et al. (1998). Pathogenesis of 57 (12): 15481549.
cortical myoclonus studied by magnetoencephalography. Okuda B, Tachibana H, Takeda M et al. (1998). Asymmetric
Ann Neurol 43 (5): 598607. changes in somatosensory evoked potentials correlate with
Mirra SS, Hyman B (2002). Aging and dementia. In: D Gra- limb apraxia in corticobasal degeneration. Acta Neurol
ham, P Lantos (Eds.), Greenfields Neuropathology. Gray Scand 97 (6): 409412.
Publishing, New York, pp. 195271. Okuma Y, Urabe T, Mochizuki H et al. (2000). Asymmetric
Mirra SS, Murrell JR, Gearing M et al. (1999). Tau pathol- cortico-cortical inhibition in patients with progressive
ogy in a family with dementia and a P301L mutation in limb-kinetic apraxia. Acta Neurol Scand 102 (4): 244248.
tau. J Neuropathol Exp Neurol 58 (4): 335345. Oliveira SA, Scott WK, Zhang F et al. (2004). Linkage
Miserez AR, Clavaguera F, Monsch AU et al. (2003). Argyr- disequilibrium and haplotype tagging polymorphisms in
ophilic grain disease: molecular genetic difference to other the Tau H1 haplotype. Neurogenetics 5 (3): 147155.
four-repeat tauopathies. Acta Neuropathol (Berl) 106 (4): Otsuki M, Soma Y, Yoshimura N et al. (1997). Slowly progres-
363366. sive limb-kinetic apraxia. Eur Neurol 37 (2): 100103.
Mitani K, Furiya Y, Uchihara T et al. (1998). Increased CSF tau Ozsancak C, Auzou P, Hannequin D (2000). Dysarthria and
protein in corticobasal degeneration. J Neurol 245 (1): 4446. orofacial apraxia in corticobasal degeneration. Mov
Monza D, Ciano C, Scaioli V et al. (2003). Neurophysiologi- Disord 15 (5): 905910.
cal features in relation to clinical signs in clinically diag- Paraskevas GP, Kapaki E, Liappas I et al. (2005). The
nosed corticobasal degeneration. Neurol Sci 24 (1): 1623. diagnostic value of cerebrospinal fluid tau protein in
Moreaud O, Monavon A, Brutti-Mairesse MP et al. (2005). dementing and nondementing neuropsychiatric disorders.
Creutzfeldt-Jakob disease mimicking corticobasal degen- J Geriatr Psychiatry Neurol 18 (3): 163173.
CORTICOBASAL DEGENERATION 371

Peigneux P, Salmon E, Garraux G et al. (2001). Neural and Rivaud-Pechoux S, Vidailhet M, Gallouedec G et al. (2000).
cognitive bases of upper limb apraxia in corticobasal Longitudinal ocular motor study in corticobasal degenera-
degeneration. Neurology 57 (7): 12591268. tion and progressive supranuclear palsy. Neurology 54 (5):
Pharr V, Uttl B, Stark M et al. (2001). Comparison of 10291032.
apraxia in corticobasal degeneration and progressive Rosenfield DB, Bogatka CJ, Viswanath NS et al. (1991).
supranuclear palsy. Neurology 56 (7): 957963. Speech apraxia in corticobasal ganglionic degeneration.
Piao YS, Hayashi S, Wakabayashi K et al. (2002). Cerebellar Ann Neurol 30: 296297.
cortical tau pathology in progressive supranuclear palsy Rottach KG, Riley DE, DiScenna AO et al. (1996). Dynamic
and corticobasal degeneration. Acta Neuropathol (Berl) properties of horizontal and vertical eye movements in
103 (5): 469474. parkinsonian syndromes. Ann Neurol 39 (3): 368377.
Pierrot-Deseilligny C (1994). Saccade and smooth-pursuit Sachdev HS, Forno LS, Kane CA (1982). Joseph disease: a
impairment after cerebral hemispheric lesions. Eur Neurol multisystem degenerative disorder of the nervous system.
34 (3): 121134. Neurology 32: 192195.
Pierrot-Deseilligny C, Rivaud S, Pillon B et al. (1989). Sakakibara R, Uchiyama T, Yamanishi T et al. (2004). Urin-
Lateral visually-guided saccades in progressive supranuc- ary function in patients with corticobasal degeneration;
lear palsy. Brain 112 (Pt 2): 471487. comparison with normal subjects. Neurourol Urodyn
Pillon B, Dubois B (2000). Memory and executive processes 23 (2): 154158.
in corticobasal degeneration. Adv Neurol 82: 91101. Salter JE, Roy EA, Black SE et al. (2004). Gestural imitation
Pillon B, Blin J, Vidailhet M et al. (1995). The neuropsycho- and limb apraxia in corticobasal degeneration. Brain Cogn
logical pattern of corticobasal degeneration: comparison 55 (2): 400402.
with progressive supranuclear palsy and Alzheimers dis- Savoiardo M (2003). Differential diagnosis of Parkinsons
ease. Neurology 45 (8): 14771483. disease and atypical parkinsonian disorders by magnetic
Pittman AM, Myers AJ, Abou-Sleiman P et al. (2005). Link- resonance imaging. Neurol Sci 24 (Suppl 1): S35S37.
age disequilibrium fine-mapping and haplotype associa- Savoiardo M, Grisoli M, Girotti F (2000). Corticobasal
tion analysis of the tau gene in progressive supranuclear degeneration. Magnetic resonance imaging in CBD,
palsy and corticobasal degeneration. J Med Genet related atypical parkinsonian disorders, and dementias.
42 (11): 837846. In: I Litvan, CG Goetz, AE Lang (Eds.), Corticobasal
Plotkin M, Amthauer H, Klaffke S et al. (2005). Combined Degeneration. Advances in Neurology, Vol. 82. Williams
123I-FP-CIT and 123I-IBZM SPECT for the diagnosis of & Wilkins, Philadelphia, pp. 197208.
parkinsonian syndromes: study on 72 patients. J Neural Sawle GV, Brooks DJ, Thompson PD et al. (1989). PET
Transm 112 (5): 677692. studies on the dopaminergic system and regional cortical
Poeck K (1983). Ideational apraxia. J Neurol 230 (1): 15. metabolism in corticobasal degeneration. Neurology
Rademakers R, Cruts M, van Broeckhoven C (2004). The 39 (1): 163.
role of tau (MAPT) in frontotemporal dementia and Schneider JA, Watts RL, Gearing M et al. (1997). Corticoba-
related tauopathies. Hum Mutat 24 (4): 277295. sal degeneration: neuropathologic and clinical heterogene-
Rebeiz JJ, Kolodny EH, Richardson EP Jr (1967). Cortico- ity. Neurology 48 (4): 959969.
dentatonigral degeneration with neuronal achromasia: a Sergeant N, Wattez A, Delacourte A (1999). Neurofibrillary
progressive disorder of late adult life. Trans Am Neurol degeneration in progressive supranuclear palsy and corti-
Assoc 92: 2326. cobasal degeneration: tau pathologies with exclusively
Rebeiz JJ, Kolodny EH, Richardson EP Jr (1968). Cortico- exon 10 isoforms. J Neurochem 72 (3): 12431249.
dentatonigral degeneration with neuronal achromasia. Shibasaki H (1995). Myoclonus. Curr Opin Neurol 8 (4):
Arch Neurol 18: 2033. 331334.
Reed LA, Wszolek ZK, Hutton M (2001). Phenotypic corre- Sobrido MJ, Abu-Khalil A, Weintraub S et al. (2003). Possi-
lations in FTDP-17. Neurobiol Aging 22 (1): 89107. ble association of the tau H1/H1 genotype with primary
Richter-Landsberg C, Bauer NG (2004). Tau-inclusion body progressive aphasia. Neurology 60 (5): 862864.
formation in oligodendroglia: the role of stress proteins Soliveri P, Piacentini S, Paridi D et al. (2003). Distal-proxi-
and proteasome inhibition. Int J Dev Neurosci 22 (7): mal differences in limb apraxia in corticobasal degenera-
443451. tion but not progressive supranuclear palsy. Neurol Sci
Riley DE, Lang AE (1993). Cortical-basal ganglionic degen- 24 (3): 213214.
eration. In: MB Stern, WC Koller (Eds.), Parkinsonian Soliveri P, Piacentini S, Girotti F (2005). Limb apraxia in
Syndromes. Dekker, New York, pp. 379392. corticobasal degeneration and progressive supranuclear
Riley DE, Lang AE (2000). Corticobasal degeneration. Clin- palsy. Neurology 64 (3): 448453.
ical diagnostic criteria. In: I Litvan, CG Goetz, AE Lang Stanford PM, Brooks WS, Teber ET et al. (2004). Frequency
(Eds.), Corticobasal Degeneration. Advances in Neurology, of tau mutations in familial and sporadic frontotemporal
Vol. 82. Williams & Wilkins, Philadelphia, pp. 2934. dementia and other tauopathies. J Neurol 251 (9):
Rinne JO, Lee MS, Thompson PD et al. (1994). Corticobasal 10981104.
degeneration. A clinical study of 36 cases. Brain 117 (Pt 5): Steele JC, Richardson JC, Olszewski J (1964). Progressive
11831196. supranuclear palsy. Arch Neurol 10: 333359.
372 N. P. STOVER ET AL.

Stover NP, Watts RL (2001). Corticobasal degeneration. Van Zanducke M, Dehaene I (2000). A cortico-basal degen-
Semin Neurol 21: 4958. eration-like syndrome as first sign of progressive multifocal
Su M, Yoshida Y, Hirata Y et al. (2000). Degeneration of the leukoencephalopathy. Acta Neurol Belg 100 (4): 242245.
cerebellar dentate nucleus in corticobasal degeneration: Verin M, Rancurel G, De Marco O et al. (1997). First familial
neuropathological and morphometric investigations. Acta cases of corticobasal degeneration. Mov Disord 12 (1): 55.
Neuropathol (Berl) 99 (4): 365370. Vidailhet M, Rivaud-Pechoux S (2000). Corticobasal degen-
Tang-Wai DF, Josephs KA, Boeve BF et al. (2003). Pathologi- eration. Eye movement disorders in corticobasal deg-
cally confirmed corticobasal degeneration presenting with eneration. In: I Litvan, CG Goetz, AE Lang (Eds.),
visuospatial dysfunction. Neurology 61 (8): 11341135. Corticobasal Degeneration. Advances in Neurology, Vol.
Tanner CM (1996). Epidemiologic approaches to cortical- 82. Williams & Wilkins, Philadelphia, pp. 161167.
basal ganglionic degeneration. Mov Disord 11: 346357. Vidailhet M, Rivaud S, Gouider-Khouja N et al. (1994).
Thompson PD (1995). Myoclonus in corticobasal degenera- Eye movements in parkinsonian syndromes. Ann Neurol
tion. Clin Neurosci 3 (4): 203208. 35 (4): 420426.
Thompson PD, Shibasaki H (2000). Myoclonus in corticobasal Vion-Dury J, Rochefort N, Michotey P et al. (2004). Proton
degeneration and other neurodegenerations. In: I Litvan, magnetic resonance neurospectroscopy and EEG cartogra-
CG Goetz, AE Lang (Eds.), Corticobasal Degeneration. phy in corticobasal degeneration: correlations with neu-
Advances in Neurology, Vol. 82. Williams & Wilkins, ropsychological signs. J Neurol Neurosurg Psychiatry
Philadelphia, pp. 6981. 75 (9): 13521355.
Thompson PD, Day BL, Rothwell JC et al. (1994). The myo- Wakabayashi K, Takahashi H (2004). Pathological heteroge-
clonus in corticobasal degeneration. Evidence for two forms neity in progressive supranuclear palsy and corticobasal
of cortical reflex myoclonus. Brain 117 (Pt 5): 11971207. degeneration. Neuropathology 24 (1): 7986.
Togo T, Sahara N, Yen SH et al. (2002). Argyrophilic grain Walter U, Dressler D, Wolters A et al. (2004). Sonographic
disease is a sporadic 4-repeat tauopathy. J Neuropathol discrimination of corticobasal degeneration vs progressive
Exp Neurol 61 (6): 547556. supranuclear palsy. Neurology 63 (3): 504509.
Tolnay M, Clavaguera F (2004). Argyrophilic grain disease: Watts RL, William RS, Growdon JH et al. (1985). Corticoba-
a late-onset dementia with distinctive features among sal ganglionic degeneration. Neurology 35 (1): 178.
tauopathies. Neuropathology 24 (4): 269283. Watts RL, Mirra SS, Richardson EP (1994). Corticobasal gang-
Tolnay M, Probst A (2003). The neuropathological spectrum lionic degeneration. In: CD Marsden, S Fahn (Eds.), Move-
of neurodegenerative tauopathies. IUBMB Life 55 (6): ment Disorders 3. Butterworths, London, pp. 282299.
299305. Wenning GK, Ben Shlomo Y, Magalhaes M et al. (1994).
Tolnay M, Sergeant N, Ghestem A et al. (2002). Argyrophilic Clinical features and natural history of multiple system
grain disease and Alzheimers disease are distinguished by atrophy. An analysis of 100 cases. Brain 117 (Pt 4):
their different distribution of tau protein isoforms. Acta 835845.
Neuropathol (Berl) 104 (4): 425434. Wenning GK, Shephard B, Hawkes C et al. (1995). Olfactory
Tuite PJ, Clark HB, Bergeron C et al. (2005). Clinical and function in atypical parkinsonian syndromes. Acta Neurol
pathologic evidence of corticobasal degeneration and pro- Scand 91 (4): 247250.
gressive supranuclear palsy in familial tauopathy. Arch Wenning GK, Litvan I, Jankovic J et al. (1998). Natural his-
Neurol 62 (9): 14531457. tory and survival of 14 patients with corticobasal degen-
Vanek ZF, Jankovic J (2000). Corticobasal degeneration. eration confirmed at postmortem examination. J Neurol
Dystonia in corticobasal degeneration. In: I Litvan, CG Neurosurg Psychiatry 64 (2): 184189.
Goetz, AE Lang (Eds.), Corticobasal Degeneration. Wetter TC, Brunner H, Collado-Seidel V et al. (2002). Sleep
Advances in Neurology, Vol. 82. Williams & Wilkins, and periodic limb movements in corticobasal degenera-
Philadelphia, pp. 6167. tion. Sleep Med 3 (1): 3336.
Vanek Z, Jankovic J (2001). Dystonia in corticobasal degen- Yamauchi H, Fukuyama H, Nagahama Y et al. (1998).
eration. Mov Disord 16 (2): 252257. Atrophy of the corpus callosum, cortical hypometabolism,
van Swieten JC, Rosso SM, van Herpen E et al. (2004). and cognitive impairment in corticobasal degeneration.
Phenotypic variation in Frontotemporal dementia and Arch Neurol 55 (5): 609614.
parkinsonism linked to chromosome 17. Dement Geriatr Zadikoff C, Lang AE (2005). Apraxia in movement disor-
Cogn Disord 17: 261264. ders. Brain 128 (Pt 7): 14801497.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 49

Infectious basis to the pathogenesis of Parkinsons disease

V. DHAWAN1,2 AND K. RAY CHAUDHURI13*

1
Regional Movement Disorders Unit, Kings College Hospital, London, UK
2
University Hospital Lewisham, London, UK
3
Guys, Kings and St. Thomas School of Biomedical Medicine and Kings College, London, UK

49.1. Introduction 49.2. A Possible immunological basis and


a link with infection
The etiology of Parkinsons disease (PD) is unknown
but a considerable body of research suggests that a Several studies have displayed evidence of immune
variety of occupational, environmental and genetic abnormalities in PD. These include the occurrence of
factors may play an important role (Ben-Shlomo, antineuronal antibodies, increases in human leukocyte
1997). Infections have also been suggested as a cause antigen-DR (HLA)-DR expression on cerebrospinal
of PD both directly and indirectly (through occu- fluid (CSF) monocytes and increases in HLA-DR
pational exposure), stemming from the observation activated microglia in substantia nigra (SN). In one
that development of parkinsonism often followed the study examining immune mediators in peripheral
influenza pandemic of the 1910s and encephalitis lymphocytes of patients with PD, patients with PD
lethargica (von Economos disease) often preceded displayed a significantly greater population of circulat-
it (Ben-Shlomo, 1997; Lai et al., 2002). Since then, ing CD3 CD4 bright CD8 dull lymphocytes than
several other possibilities of an infective basis to PD age-matched control subjects (Hisanaga et al., 2001).
and parkinsonism have been put forward and this These cells are known to increase after some specific
review examines the basis behind these observations. viral infections, hence raising the possibility of an
PD occurs throughout the world across all races and association between postinfectious immune abnormal-
increases exponentially with age. The late onset ities and pathogenesis of PD. CD8dull and
and slow-progressing nature of the disease have CD4bright T cells have been associated with
prompted the consideration of environmental expo- EpsteinBarr virus (EBV), cytomegalovirus, human
sure to agrochemicals, including pesticides, as a risk herpes 6, human T-cell leukemia virus type-1 and
factor. Moreover, increasing evidence suggests that human immunodeficiency virus (HIV). However,
early-life occurrence of inflammation in the brain, as these have also been demonstrated in patients with
a consequence of either brain injury or exposure to neoplasms as well as in small populations of healthy
infectious agents, may play a role in the pathogenesis adults and in patients with multiple sclerosis, myasthe-
of PD. Most importantly, there may be a self- nia gravis and during rejection of renal transplants.
propelling cycle of inflammatory process involving The precise reasons behind why CD4 and CD8
brain immune cells (microglia and astrocytes) that T cells increase in PD and whether these circulating
drives the slow yet progressive neurodegenerative lymphocytes contribute to the pathogenesis of neuro-
process. Differences in prevalence between identical nal death in PD remain inconclusive and require
ethnic groups in different countries support the role further investigation.
of an environmental factor, including an infectious Salman and colleagues (1999) reported decreased
basis (Richards and Chaudhuri, 1996; Stoessl, 1999; phagocytic function exhibited by peripheral blood
Chaudhuri et al., 2000). polymorphonuclears of PD patients, as shown by their

*Correspondence to: Dr. K. Ray Chaudhuri, 9th floor, Ruskin Wing, Kings College Hospital, Denmark Hill, London SE5 9RS,
UK. E-mail: Ray.chaudhuri@uhl.nhs.uk, Tel: 44-(0)208-333-3030, Ext. 6184, Fax: 44-(0)208-333-3093.
374 V. DHAWAN AND K. R. CHAUDHURI
impaired ability to engulf latex particles. Function of forms of this protein in PD and related neurodegenerative
phagocytes is significant in trying to combat bacterial synucleinopathies.
invasion or destruction of any foreign material, It is possible that similar cross-reacting immune
thereby increasing the susceptibility of PD patients to responses are generated as part of the immune response
infection, but could be secondary to the disease rather to natural infection with a virus. The cross-reactivity
than playing a direct causative role in its pathogenesis. between a virus and protein may bear important implica-
Other immune abnormalities described in PD tions in elucidating infectious mechanisms in the patho-
include the occurrence of autoantibodies against neu- genesis of PD. More specifically, studies have focused
ronal structures and an elevated number of microglia on the possible role of the following organisms in the
cells expressing the histocompatibility glycoprotein pathogenesis of PD and related syndromes (Fig. 49.1):
HLA-DR in the SN. An increased gamma delta ()
 HIV
T-cell population and immunoglobulin G immunity
 Prion protein
in CSF to heat shock proteins has been found in PD
 Nocardia asteroides
(Fiszer, 2001). Impaired cytokine production by the
 Japanese encephalitis virus
peripheral immune system in PD patients with a sig-
 Influenza A virus
nificant decrease in the production of tumor necrosis
 Helicobacter pylori
factor-a (TNF-a), interleukin (IL)-1a, IL-1b and IL-6
 Toxoplasma gondii
by peripheral blood mononuclear cells and TNF-a by
peripheral blood macrophages was demonstrated
49.3. Review of clinical case reports
by Hasegawa et al. (2000).
Rats with nigral injection of immunoglobulin 49.3.1. HIV spontaneous-onset extrapyramidal
G (IgG) purified from the serum of 5 patients with PD symptoms in patients with HIV/AIDS
and 10 disease control patients suggested that PD IgG
can initiate a relatively specific inflammatory destruc- Movement disorders are recognized as a complication
tion of SN pars compacta neurons in vivo (Chen et al., of HIV/acquired immune deficiency syndrome (AIDS)
1998). and may be both hypo- and hyperkinetic in nature.
A cytokine/CD23-dependent activation pathway of Opportunistic infections or, occasionally, drug thera-
inducible nitric oxide synthase, toxic oxidative species pies may be responsible, although in some cases there
and of proinflammatory mediators in glial cells and may be no identifiable precipitant. Estimates of preva-
their involvement in the pathophysiology of PD has lence of movement disorders in HIV-seropositive
been suggested in a study by Hunot et al. (1999). Also patients range between 0.006 and 14%, depending on
in a study in 1999, Linda et al. demonstrated that motor concurrent pharmacological therapy and the method of
neurons and nigral dopaminergic neurons (i.e. those analysis employed (Nath et al., 1987; Mirsattari et al.,
neurons that are susceptible to neurodegeneration in 1998; De Mattos et al., 2002; Tse et al., 2004). Cardoso
diseases such as PD and amyotrophic lateral sclerosis), (2002) proposed that clinically relevant movement disor-
in the brainstem of the adult rat, displayed high levels ders are identified in 3% of patients with HIV infection
of both major histocompatibility complex (MHC) seen at tertiary referral centers and prospective follow-
class I heavy-chain and b2-microglobulin mRNAs. up shows that 50% of patients with AIDS develop
A deficiency in expression of MHC class I is thought tremor, parkinsonism or other extrapyramidal features.
to impede CD8 T-cell recognition and target cell kill- Parkinsonism has been described in several cases with
ing. These mechanisms may protect neurons and other HIV infection and AIDS, which result from lesions
cells with low regenerative capacity from destruction caused by opportunistic infections such as toxoplasmo-
by T cells. sis, which damage the basal ganglia connections
Antibodies raised against viral determinants some- (Table 49.1). Parkinsonism, tremor and dystonia can also
times cross-react with host proteins in immunohisto- result from dopaminergic dysfunction caused by cellular
chemical studies and such cross-reacting antibodies action of dopamine-blocking agents (Table 49.2).
may thereby play a role in disease pathogenesis. Extrapyramidal symptoms associated with HIV/
a-Synuclein, a neuronal protein, is reported to be a major AIDS were described by Berger et al. (1984), who
component of Lewy bodies (LBs) in PD, dementia with noted the presence of tremor in 3 out of 165 patients
LBs and common variants of Alzheimers disease. Gias- (0.02%) recently diagnosed with AIDS in Florida,
son et al. (2000) developed a panel of antisynuclein USA. Three years later Nath et al. (1987) published a
antibodies and performed immunohistochemical studies case report of 2 patients with AIDS and rest tremor.
showing that these antibodies label numerous LBs in The patients were both male, aged 57 and 37 years,
the SN of PD, thereby suggesting a role of pathological and had been diagnosed with AIDS 17 and 22 months
INFECTIOUS BASIS TO THE PATHOGENESIS OF PARKINSONS DISEASE 375

A B

C D
Fig. 49.1. Possible infectious organisms implicated in cause of Parkinsons disease and parkinsonism. (A) Human immunode-
ficiency virus; (B) influenza; (C) Helicobacter pylori; (D) CJD (prion particles); (E) Toxoplasma; (F) Nocardia asteroides.

previously. Serological investigations and neuroima- In 1989 Carrazana et al. published a case report
ging failed to identify obvious causes of parkinsonism of acute-onset parkinsonism associated with AIDS in
in these patients and a direct effect of HIV-1 on the a 66-year-old female. The patient presented with a
basal ganglia was hypothesized. Brain biopsy later 2-week history of lethargy, tremor and slow, shuffling
revealed cerebral Whipples disease in 1 of the patients gait and a 2-day history of fever. Her family had noted
and it was suggested this may have contributed to the her facial expression was reduced. Examination
clinical presentation. revealed bilateral cogwheel rigidity, infrequent
376 V. DHAWAN AND K. R. CHAUDHURI

E F
Fig. 49.1. (Continued)

blinking, drooling, right pronator drift, right-sided Parkinsonian symptoms of tremor, hypomimia and
hyperreflexia and an extensor plantar response on the gait disturbance caused by mesencephalic cryptococ-
right. Serum and CSF toxoplasma titers were elevated cal abscesses in a 63-year-old man with AIDS
and computed tomographic (CT) neuroimaging have also been reported (Bouffard et al., 2003). The
revealed bilateral ring-enhancing lesions in the ante- patients symptoms were initially attributed to early
rior limb of the internal capsule, suggestive of cerebral PD; diagnoses of AIDS and cryptococcal infection
toxoplasmosis. HIV-1 in serum was positive. There were made following his death due to cardiac arrest
was a poor clinical response to levodopa and pyri- 8 months after the onset of his neurological symptoms.
methamine and sulfadiazine therapy were discontinued Autopsy revealed bilateral cryptococcal abscesses in
following the development of pancytopenia. The the SN without any characteristic changes of cerebral
patient died secondary to septicemia within 5 months HIV infection or other obvious underlying pathology
of the onset of parkinsonian symptoms. Tolge and Fac- (Bouffard et al., 2003). Bradykinesia and rigidity were
tor (1991) reported a case of isolated focal dystonia in also reported in association with central nervous
the left hand and arm of a 29-year-old male, 5 months system (CNS) tuberculosis in a 49-year-old male,
after having received the diagnosis of HIV-1 infection. as the presenting manifestation of HIV infection (de La
Serum toxoplasma titer was elevated and a CT scan Fuente-Aguado et al., 1996). The diagnosis of CNS
revealed ring-enhancing lesions, typical of Toxo- tuberculosis was made on the basis of neuroimaging
plasma abscesses, in the right lenticular nucleus and appearances (Fig. 49.3) and the growth of acid-fast
right thalamus (Fig. 49.2). bacilli on a CSF smear. CD4 count at diagnosis was
Maggi et al. (2000) reported an AIDS patient who 133 cells/mm3. The case is interesting as parkinsonian
developed cerebral opportunistic granulomatous features resolved fully following 12 days of antitubercu-
lesions and, subsequently, a parkinsonian akinetic- losis therapy plus intravenous dexamethasone.
rigid syndrome. The parkinsonian syndrome only
developed when the lesions involved basal ganglia 49.3.2. Postencephalitic parkinsonism (PEP)
bilaterally.
Werring and Chaudhuri (1996) also described a 49.3.2.1. Epidemiological aspects
rapidly developing akinetic-rigid syndrome progres- A pandemic of encephalitis lethargica occurred
sing to akinetic mutism and death in a patient with between 1919 and 1926, resulting in the emergence
recently acquired HIV. of the syndrome of PEP. A younger age of onset of
Table 49.1

INFECTIOUS BASIS TO THE PATHOGENESIS OF PARKINSONS DISEASE


Summary of case reports of spontaneous-onset parkinsonism in patients with human immunodeficiency virus (HIV)/acquired immune deficiency syndrome (AIDS)

Werring and De la
Berger et al. Nath et al. Carrazana Chaudhuri Fuente-Aguado Hersh et al. De Mattos Bouffard
Case reports of parkinsonism (1984) (1987) et al. (1989) (1996) et al. (1996 ) (2001) et al. (2002) et al. (2003) Total

Male 3 2 1 1 1 10 1 19
Female 1 3 4
Mean age (years) 47 66 38 49 37 37 63 44.1
Latency between Dx HIV/AIDS
and onset of EPS (months) 17 to22 0.5 1 0 4 5 0
Baseline CD4 count (cells/mm3) 1/1 110 438 133 227
EPS attributed to toxoplasmosis 1/14 2/23
EPS attributed to cryptococcosis 1/1 1/23
EPS attributed to CNS
tuberculosis 1/1 1/23
EPS attributed to PML 1/1 1/1
No cause found to explain EPS 2/2 1/1 12/13 15/23
Improvement with
antitoxoplasmosis Tx No No 0/2
Improvement with anti-TB Tx 1/1 1/1
Improvement with levodopa No No Yes Yes5/9 6/12
Improvement with HAART Yes 1/1
Deaths 1 1 8 1 11
Latency between onset of EPS
and death (months) 5 5 8 6

Dx, diagnosis; EPS, extrapyramidal symptoms; CNS, central nervous system; PML, progressive multifocal leukoencephalopathy; Tx, treatment; TB, tuberculosis; HAART, highly active antiretroviral
treatment; implies EPS began after diagnosis of HIV/AIDS; implies EPS began before HIV diagnosis.

377
378 V. DHAWAN AND K. R. CHAUDHURI
Table 49.2
Summary of case reports of dystonia in patients with human immunodeficiency virus (HIV)/acquired immune deficiency
syndrome (AIDS)

Nath et al. Tolge and De Mattos


Case reports of dystonia (1987) Factor (1991) et al. (2002) Total

Right-sided Male 1 1
Right-sided Female
Left-sided Male 1 1
Left-sided Female
Bilateral Male 1 1
Bilateral Female
Mean age (years) 35 29 28 30.7
Mean time between diagnosis of HIV/AIDS and onset of EPS 4 months 5 months
EPS attributed to basal ganglia toxoplasmosis 1/1 1/1 2/3
No cause found to explain EPS 1/1 1/3
Clinical improvement with antitoxoplasmosis treatment No No 0/2
Deaths 0

EPS, extrapyramidal symptoms.


implies EPS began after diagnosis of HIV/AIDS; implies EPS began before HIV diagnosis.

Fig. 49.2. Contrasted computed tomographic image in a patient presenting with focal dystonia. Cerebral toxoplasmosis
abscess in right lenticular nucleus and left temporal cortex (A) and right thalamus (B). Reproduced from Tolge and
Factor (1991), with permission.

disease (36.8 years) was reported from a group of that the pandemic of 19191926 may have influenced
London hospitals compared to an age of onset of the earlier onset of disease in this group of patients.
54.7 years between 1900 and 1920 (Duvoisin et al., Mortality data from England and Wales related to PD
1963) At this time the age of onset of PEP (between showed that, although there is a fivefold increase in
1921 and 1942) was 27.4 years. Studies from the USA death rates among people over 80 years of age, younger
(Massachusetts General Hospital) also suggest that age people with PD had a lower mortality rate (Martyn,
of diagnosis of parkinsonism between 1920 and 1924 1997). Although the possibility of a birth cohort rate
was 27 years less than those diagnosed between 1919 cannot be excluded, these data provide indirect
and 1926 (Poskanzer and Schwab, 1963), suggesting evidence that people born at the beginning of the
INFECTIOUS BASIS TO THE PATHOGENESIS OF PARKINSONS DISEASE 379
these patients improved and were able to mobilize
and walk. Reversible mutism was observed in 3
patients during the acute phase. Response to levodopa,
amantadine and trihexiphenedyl was partial. All 5
patients were followed for more than 1 year, during
which time the parkinsonian features recovered
substantially.
Peatfield (1987) described 2 patients with a chronic
non-progressive illness beginning with excessive slee-
piness and personality change. Both had an atypical
movement disorder, clearly distinct from PD, with
impairment of memory and learning and relative pre-
servation of arithmetical, language and visuospatial
tasks, suggesting parkinsonism with a subcortical
dementia. CT scans of the brain showed atrophy of
deep structures and elevated Coxsackievirus antibody
titers. The author speculated that insidious viral ence-
phalitis (perhaps these cases would previously have
been described as encephalitis lethargica) could lead
to subcortical dementia.
Calne and Lees (1988) performed a detailed retro-
Fig. 49.3. Computed tomography image in a patient present- spective analysis of 11 long-standing (greater than 40
ing with parkinsonism. Hypodense cerebral lesion affecting years) inmates in a hospital with neurological deficits
left external capsule, posterior limb of internal capsule and due to encephalitis lethargica. Retrospective informa-
lower region of left lenticular nuclei. The lesion did not show tion was gathered from physicians and nurses
enhancement to the intravenous contrast and was diagnosed records, photographs, published narrative and hospital
as central nervous system tuberculosis on the basis of cere- charts dating back as far as 1931. They concluded that
brospinal fluid analysis. Reproduced from de La Fuente- neurological disabilities attributable to basal ganglia
Aguado et al. (1996), with permission. damage frequently increase in late life, with deteriora-
tion being most marked in motor function and largely
20th century suffered from an unusually high rate sparing the intellect, special senses and somatosensory
of parkinsonism, thus supporting the notion that system.
encephalitis lethargica could have been implicated. Picard et al. (1996) described a patient who went on
Reports from a recent study on 22 patients with an ence- to develop a severe parkinsonian syndrome a few years
phalitis lethargica-like phenotype advocate that it is still after being diagnosed with an encephalitic syndrome
prevalent and is a postinfectious autoimmune CNS dis- associated with a prolonged lethargic state in his
order with autoantibody targeting of basal ganglia and childhood. He presented with axial dystonia and
SN neurons (Dale et al., 2002). stereotyped abnormal movements of the upper limbs,
with normal brain imaging. Fluorodopa positron emis-
49.3.2.2. Case report-related observations sion tomography (PET) showed a significant bilateral
The Japanese B encephalitis virus is the first demon- reduction of tracer accumulation in both putamens,
strated virus capable of producing nigral lesions and similar to that observed in patients with idiopathic
causing parkinsonism. Pradhan et al. (1999) selected PD, and treatment with levodopa suppressed akinetic,
5 patients from 52 patients with Japanese B encephali- dystonic and dyskinetic symptoms in this patient.
tis seen in an endemic zone, on the basis of isolated These symptoms appeared to be related to a limited
lesions in the SN (revealed on imaging with magnetic lesion of the dopaminergic neurons of the zona
resonance imaging (MRI)) and performed detailed compacta of the SN.
clinical and laboratory evaluations. Examination dur- Ghaemi et al. (2000) reported another case of a
ing acute illness revealed restricted eye movements, 74-year-old woman who developed an akinetic-rigid
opsoclonus, upbeating nystagmus and cogwheel Parkinson syndrome with tremor, hypokinesia, hypo-
rigidity with reversal of consciousness and eye signs mimia, rigidity and cogwheel phenomenon in all four
after the acute illness. Parkinsonian features, such as extremities following viral encephalitis. The diagnosis
positive glabellar tap, facial impassivity, bradykinesia, was supported by a reactive CSF assay and a positive
tremors and postural instability, became apparent as influenza A IgA antibody titer (1:>160). PET studies
380 V. DHAWAN AND K. R. CHAUDHURI
showed an altered pattern of glucose and dopa metabo- ganisms and thus disputed the link of parkinsonism
lism, clearly distinguishing it from idiopathic Parkin- with Nocardia.
son syndrome. Subsequently, Hubble et al. (1995) used a serodiag-
nostic panel to determine antibodies specific for
49.3.2.3. Clinical features Nocardia in PD patients by comparing sera from
Litvan and colleagues (1998) reported results of a healthy volunteers and from patients with culture-
retrospective clinicopathologic study to determine the proven nocardiosis. No difference in seropositivity
validity of the clinical diagnosis of PEP by presenting was found when PD patients were compared with their
105 records with neuropathologic diagnoses of PEP age- and gender-matched controls (n 140). The
(n 7), progressive supranuclear palsy (n 24), PD results reveal a high exposure rate of humans to
(n 15), dementia with LBs (n 14), multiple system nocardial antigens, especially among men and older
atrophy (n 16), corticobasal degeneration (n 10), individuals, and do not confirm the hypothesis that
CreutzfeldtJakob disease (CJD: n 4) and other nocardiosis may cause PD or parkinsonism, although
dementia disorders (n 15) as clinical vignettes to the authors acknowledge that serological testing may
six neurologists who were unaware of the autopsy not be optimal for the detection of Nocardia-related
findings. The neurologists own clinical diagnoses, CNS infection (Hubble et al., 1995).
when compared with neuropathologic diagnoses, N. asteroides are composed of both filamentous and
displayed high reliability, sensitivity and positive filterable forms. In a further study, Kohbata et al.
predictive values for the clinical diagnosis of PEP. (1998) investigated the presence of acid-fast spherical
According to their data set, the best predictors for the structures similar to filterable Nocardia at the
diagnosis of PEP included: midbrain nigral lesions of 3 patients with PD. Many
clusters of acid-fast lipochrome bodies were present
1. Onset below middle age
in the vicinity of melanin-pigmented neurons in the
2. Symptom duration lasting more than 10 years
3 PD patients studied and none were observed in 3
3. Presence of oculogyric crisis
control patients. Examination of adjacent hematoxylin
A relevant history of encephalitis lethargica, pre- and eosin-stained sections indicated that they consisted
sent in most PEP cases, was an important individual of yellow-green granules, bodies and aggregates in
diagnostic predictor. Other reported features include: ballooned glial cells. However, the results do not
confirm or refute the proposed link between Nocardia
1. Sleep disturbances
and Parkinsons disease, and they suggest that the
2. Excessive daytime sleepiness
immunological and genetic relationship between the
However, as the reported case histories suggest, acid-fast lipochrome bodies and filterable Nocardia
there is a wide phenotypic variation in cases labelled should be investigated. In another experiment with
as PEP, ranging from a levodopa-responsive syndrome the midbrain sections of PD patients and controls,
with PET appearances of idiopathic PD to a syndrome acid-fast stain and antifilterable Nocardia antiserum
of parkinsonism with early dementia. were used and confirmed the presence of filterable
forms of N. asteroides in the midbrain nigral lesions
49.3.3. Nocardia asteroides that occur with PD (Kohbata et al., 2002).
Recently LeWitt and colleagues (2004) reliably
Nocardia is a genus of aerobic Gram-positive bacteria verified that, in mica models, prior infection with
which forms filamentous cells that fragment into rod- certain strains of N. asteroides or, possibly, other
shaped or coccoid elements or L-forms (see Fig. 49.1). neurotoxic microorganisms in the environment caused
It is found in soil, stagnant water and farming areas. neuronal degeneration and various levodopa-
The link between Nocardia infection and parkinson- responsive and persisting motor disorders resembling
ism was suggested by Beaman, a leading worker in parkinsonism. They postulated that this might be an
this field based on work in animal models (see section etiological basis for this or for other neurodegenerative
49.4, below). Kohbata and Shimokawa (1993) detected disorders.
antibodies to coccoid and rod-shaped cells of Nocardia
in the serum of patients with PD. The antibodies 49.3.4. Helicobactor pylori
were demonstrated in all the 20/20 patients with PD
at a titer greater than 1:10 and 10 out of 14 controls. Altschuler (1996) suggested that gastric Helicobacter
The results suggested that PD patients amid age- pylori infection might be a cause for both idiopathic
matched healthy volunteers are routinely exposed to PD and non-arteritic anterior optic ischemic neuropa-
and naturally infected with Nocardia-related microor- thy, given the higher prevalence of gastrointestinal
INFECTIOUS BASIS TO THE PATHOGENESIS OF PARKINSONS DISEASE 381
ulcers in both these diseases than reported in age- and a patient with histopathologically verified sporadic CJD
sex-matched controls or in the general population. presented initially with diplopia, sleep disturbances
Charlett et al. (1999) investigated the role of H. pylori and levodopa-responsive parkinsonism. Unusually, he
of PD and the issue of cross-infection in familial did not dement and initially failed to fulfill the clinical
aggregation by checking H. pylori seropositivity in criteria for possible CJD after more than a year of
33 elderly subjects with idiopathic parkinsonism and slow progression. It was proposed that CJD should be
39 siblings and controls). They found that both parkin- included in the differential diagnosis of parkinson
sonians and siblings differed from controls in the odds plus syndromes until a different etiology has been
of being H. pylori-seropositive (odds ratios 3.04 (95% found or a histopathologic examination performed.
confidence interval: 1.22, 7.63) and 2.94 (1.26, 6.86) Nitrini et al. (2001) presented the clinical features
respectively, P < 0.02); seropositivity was present in of 12 patients from a family with CJD associated with
0.70 of sufferers. Familial transmission of chronic a point mutation at codon 183 of the prion protein
infection was linked with causality. gene. The duration of the symptoms until death ranged
H. pylori is one of the commonest of known from 2 to 9 years. Nine patients were first seen by
bacterial infections linked with peptic ulcer/non-ulcer psychiatrists for behavioral disturbances. Eight
dyspepsia, immunosuppression and autoimmunity. patients manifested parkinsonian signs. The authors
Dobbs et al. (2000) challenged the conventional con- concluded that these clinical features bear a consider-
cept of a role of a neurotoxin as an environmental able resemblance to those described in frontotemporal
cause of idiopathic parkinsonism by suggesting that dementia and parkinsonism linked to chromosome 17
H. pylori-related acquired immunosuppression, predis- (FTDP-17), not usually considered in the differential
posing to autoimmunity, could serve as a model for diagnosis of prion diseases.
the pathogenesis of parkinsonism and eradication of A Japanese case of sporadic CJD presenting as
H. pylori has the potential to change the approach to progressive supranuclear palsy with slowly progressing
parkinsonism. neurological deficits for 2 years after onset was reported
The theory remains controversial, and a study in 2003 (Shimamura et al., 2003). Needle biopsy
conducted by Wlodarek et al. (2003) found no relation revealed deposition of prion protein of a patchy/
between PD and H. pylori infection. Another issue is perivacuolar type with spongiform degeneration.
that H. pylori infection positivity affects levodopa Of late, 2 further cases of CJD presenting with a
pharmacokinetics in PD, through direct degradation progressive supranuclear palsy-like syndrome and
of the drug or changes of gastroduodenal environment autopsy confirmation have been reported from the
(Brusa et al., 2004). USA (Josephs et al., 2004).

49.3.5. EpsteinBarr virus 49.3.7. Leprosy

Woulfe et al. (2000) suggested cross-reactivity In early 2003, an international team of scientists con-
between monoclonal antibodies against EBV and a- ducted a genome scan in Vietnamese multiplex leprosy
synuclein in the human brain. a-Synuclein has been families and found that susceptibility to leprosy was
reported to be a major component of LBs in PD, demen- significantly linked to region q25 on the long arm of
tia with LBs and common variants of Alzheimers chromosome 6 (Buschman and Skamene, 2004). In a
disease. However, in continuation of the study, similar continuation of these findings, the team has pinpointed
experiments in 2002 failed to demonstrate elevated the chromosome 6 susceptibility locus to the 5 regula-
anti-a-synuclein and anti-EBV latent membrane protein tory promoter region shared by both the PD gene
antibodies in PD, thus refuting the association between PARK2 and its co-regulated gene PACRG. The surpris-
the virus and PD (Woulfe et al., 2002). ing discovery has important implications for the under-
standing of leprosy pathogenesis and for the strategy of
49.3.6. Transmissible spongiform encephalopathy genetic analysis of infectious diseases.
(prion protein-related disorders)
49.4. Animal studies
There are reports of parkinsonism developing in the
context of CJD. A case of coexistent clinical findings Potential associations between PD or parkinsonism-
of idiopathic PD and CJD was described in 1985 and like disease and exposure to various infectious agents
confirmed at autopsy (Ezrin-Waters et al., 1985). have been demonstrated in some animal studies.
An unusual case of CJD was identified in 1998, in The development of a rhythmic, uncontrolled
Geneva, Switzerland (Vingerhoets et al., 1998), where vertical shake of the head (45/second), tremulous
382 V. DHAWAN AND K. R. CHAUDHURI
movement, stooped posture and restlessness in female rhesus monkeys during the asymptomatic stage of the
BALB/c mice (head shakes were temporarily stopped infection (Jenuwein et al., 2004). Changes in cyclic
by treatment with levodopa) following intravenous adenosine monophosphate (cAMP) and cAMP
injection of various doses of log-phase Nocardia aster- response element-binding protein (CREB), two factors
oides GUH-2 has been reported (Kohbata and Beaman, involved in the signaling pathway of dopamine, were
1991). Studies show localization and growth of nocar- also investigated. The brain regions examined were
dial cells within the brains and loss of Nissl substance the nucleus accumbens and the corpus amygdaloi-
and tyrosine hydroxylase immunoreactivity in the deum, which are limbic structures of the basal ganglia
neurons of SN. Therefore, the authors speculate that that are involved in the pathophysiology of psychiatric
mice infected with N. asteroides may serve as a model disorders and substance abuse. Dopamine content was
for studying parkinsonian signs and other degenerative reduced in both regions, nucleus accumbens and the
diseases involving extrapyramidal and pyramidal corpus amygdaloideum (limbic structures of the basal
systems. ganglia that are involved in the pathophysiology of
In another study, in vivo and in vitro models were psychiatric disorders) of SIV-infected monkeys
utilized to measure the ability of N. asteroides GUH-2, compared to uninfected animals. Moreover, dopamine
a neuroinvasive strain, to induce the apoptotic death of deficits were associated with a decrease in the expres-
dopaminergic cells (Tam et al., 2002). Apoptosis has sion of total CREB. Changes in dopamine signaling
been implicated in dopaminergic neuronal dropout in were not related to pathology or viral load of the
Parkinsons patients, causing a levodopa-responsive investigated animals.
movement disorder resembling parkinsonism.
Following infection with GUH-2, dopaminergic apop- 49.5. Conclusion
totic cells were identified in the SN of animals by in situ
end-labeling, which detects DNA fragmentation, com- The precise cause of PD remains unknown.
bined with fluorescent immunolabeling of tyrosine Environmental factors, with their possible effects on
hydroxylase-positive cells. genetic susceptibility, are possible causal mechanisms.
Nocardia otitidiscaviarum (GAM-5), isolated from Infections may form part of the environmental insult
a patient with an actinomycetoma, produced signs which may predispose to PD or parkinsonism.
similar to PD following a non-lethal dose of intrave- However, the evidence is weak and largely based on
nous injection into Naval Medical Research Institute anecdotal reports. The most robust causative link of
(NMRI) mice (Diaz-Corrales et al., 2004). Fourteen PD/parkinsonism and infection is the emergence of
days after bacterial infection, most of the 60 mice PEP, although the current rates of prevalence and
injected exhibited parkinsonian features characterized incidence of PD do not suggest a stray/infective
by vertical head tremor, akinesia/bradykinesia, flexed encephalopathic basis.
posture and postural instability. Dopamine levels were
reduced from 110  32.5 to 58  16.5 ng/mg protein Acknowledgments
(47.2% reduction) in brain from infected mice exhibit-
ing impaired movements, whereas serotonin levels We acknowledge the contribution of the following
were unchanged (191  44 ng/mg protein in control fourth-year medical students, Gillian Lapthorn and
and 175  39 ng/mg protein in injected mice). Yolanda Sydney Augustin, who performed their study
Chapman et al. (2003) developed an in situ hybridi- modules based on the theme of this article at Guys,
zation technique to detect nocardial 16S ribosomal Kings and St. Thomass School of Medicine, London,
RNA, using a Nocardia-specific probe (B77) in the UK.
cerebral cortical specimens from cynomolgus monkeys
at 48 hours, 3.5 months and 1 year after experimental
infection with N. asteroides. This in situ hybridization References
procedure, applied in a blinded fashion to human SN
specimens with LBs, detected similar reactivity in Altschuler E (1996). Gastric Helicobacter pylori infection as
a cause of idiopathic Parkinson disease and non-arteric
inclusions with the appearance of LBs, suggesting a
anterior optic ischemic neuropathy. Med Hypotheses 47:
possible association between Nocardia and neurode-
413414.
generative disorders in which LBs are present. In Ben-Shlomo Y (1997). The epidemiology of Parkinsons
2004 a German study measured levels of dopamine disease. Baillieres Clin Neurol 6: 5568.
and its metabolites, homovanillic acid and 3,4- Berger JR, Moskowitz L, Fischl M et al. (1984). The neurologic
dihydroxyphenylacetic acid, in brains of uninfected complications of AIDS: Frequently the initial manifestation.
and simian immunodeficiency virus (SIV)-infected Neurology 34: 134135.
INFECTIOUS BASIS TO THE PATHOGENESIS OF PARKINSONS DISEASE 383

Bouffard JP, Mena H, Ripple M et al. (2003). Mesencepha- Ghaemi M, Rudolf J, Schmulling S et al. (2000). FDG- and
lic cryptococcal abscesses presenting with parkinsonism Dopa-PET in postencephalitic parkinsonism. J Neural
as an initial manifestation of AIDS. Mov Disord 18: Transm 107: 12891295.
13541357. Giasson BI, Jakes R, Goedert M et al. (2000). A panel of epi-
Brusa L, Pietroiusti A, Pierantozzi M et al. (2004). Long tope-specific antibodies detects protein domains distribu-
term effects of Helicobacter pylori eradication on L- ted throughout human alpha-synuclein in Lewy bodies of
DOPA absorption in Parkinsons disease patients. Mov Parkinsons disease. J Neurosci Res 59: 528533.
Disord 19: S387S399. Hasegawa Y, Inagaki T, Sawada M et al. (2000). Impaired
Buschman E, Skamene E (2004). Linkage of leprosy suscept- cytokine production by peripheral blood mononuclear
ibility to Parkinsons disease genes. Int J Lepr Other cells and monocytes/macrophages in Parkinsons disease.
Mycobact Dis 72: 169170. Acta Neurol Scand 101: 159164.
Calne DB, Lees AJ (1988). Late progression of post-encephalitic Hersh BP, Rajendran PR, Battinelli D (2001). Parkinsonism
Parkinsons syndrome. Can J Neurol Sci 15: 135138. as the presenting manifestation of HIV infection: Improve-
Cardoso F (2002). HIV-related movement disorders: Epide- ment on HAART. Neurology 56: 278279.
miology, pathogenesis and management. CNS Drugs 16: Hisanaga K, Asagi M, Itoyama Y et al. (2001). Increase in
663668. peripheral CD4 bright CD8 dull T cells in Parkinson
Carrazana E, Rossitch E Jr, Samuels MA (1989). Parkinso- disease. Arch Neurol 58: 15801583.
nian symptoms in a patient with AIDS and cerebral toxo- Hubble JP, Cao T, Kjelstrom JA et al. (1995). Nocardia
plasmosis. J Neurol Neurosurg Psychiatry 52: 14451447. species as an etiologic agent in Parkinsons disease:
Chapman G, Beaman BL, Loeffler DA et al. (2003). In situ Serological testing in a case-control study. J Clin Microbiol
hybridization for detection of nocardial 16S rRNA: Reactiv- 33: 27682769.
ity within intracellular inclusions in experimentally infected Hunot S, Dugas N, Faucheux B et al. (1999). FcepsilonRII/
cynomolgus monkeysand in Lewy body-containing CD23 is expressed in Parkinsons disease and induces,
human brain specimens. Exp Neurol 184: 715725. in vitro, production of nitric oxide and tumor necrosis
Charlett A, Dobbs RJ, Dobbs SM et al. (1999). Parkinsonism: factor-alpha in glial cells. J Neurosci 19: 34403447.
Siblings share Helicobacter pylori seropositivity and Jenuwein M, Scheller C, Neuen-Jacob E et al. (2004).
facets of syndrome. Acta Neurol Scand 99: 2635. Dopamine deficits and regulation of the cAMP second
Chaudhuri KR, Hu MT, Brooks DJ (2000). Atypical parkin- messenger system in brains of simian immunodeficiency
sonism in Afro-Caribbean and Indian immigrants to UK. virus-infected rhesus monkeys. J Neurovirol 10: 163170.
Mov Disord 15: 1823. Josephs KA, Tsuboi Y, Dickson DW (2004). Creutzfeldt-
Chen S, Le WD, Xie WJ et al. (1998). Experimental destruc- Jakob disease presenting as progressive supranuclear
tion of substantia nigra initiated by Parkinson disease palsy. Eur J Neurol 11: 343346.
immunoglobulins. Arch Neurol 55: 10751080. Kohbata S, Beaman BL (1991). L-dopa-responsive move-
Dale RC, Church AJ, Surtees RA et al. (2002). Re-emergence ment disorder caused by Nocardia asteroides localized in
of encephalitis lethargica-like syndrome: Evidence of the brains of mice. Infect Immun 59: 181191.
CNS autoimmunity. Mov Disord 17: S260. Kohbata S, Shimokawa K (1993). Circulating antibody to
De La Fuente-Aguado J, Bordon J, Moreno J et al. (1996). Nocardia in the serum of patients with Parkinsons
Parkinsonism in an HIV-infected patient with hypodense disease. Adv Neurol 60: 355357.
cerebral lesion. Tubercle Lung Dis 77: 191192. Kohbata S, Tamura T, Hayashi R (1998). Accumulation of
De Mattos JP, De Rosso AL, Correa RB et al. (2002). Move- acid-fast lipochrome bodies in glial cells of the midbrain
ment disorders in 28 HIV-infected patients. Arq Neuropsi- nigral lesion in Parkinsons disease. Clin Diagn Lab
quiatr 60: 525530. Immunol 5: 888893.
Diaz-Corrales FJ, Colasante C, Contreras Q et al. (2004). Kohbata S, Hayashi R, Tamura T (2002). An infection with
Nocardia otitidiscaviarum (GAM-5) induces parkinso- filterable forms of Nocardia asteroides in the midbrain
nian-like alterations in mouse. Braz J Med Biol Res 37: nigral lesion in Parkinsons disease. Mov Disord 17: S62.
539548. Lai BC, Marion SA, Teschke K et al. (2002). Occupational
Dobbs SM, Dobbs RJ, Weller C et al. (2000). Link between and environmental risk factors for Parkinsons disease.
Helicobacter pylori infection and idiopathic parkinsonism. Parkinsonism Relat Disord 8: 297309.
Med Hypotheses 55: 9398. LeWitt PA, Beaman BL, Camp DM et al. (2004). Nocardia
Duvoisin RC, Yahr MD, Schweitzer MD et al. (1963). Par- asteroides: A possible environmental cause of Parkinsons
kinsonism before and since the epidemic of encephalitis disease? Mov Disord 19: S222.
lethargica. Arch Neurol 9: 232236. Linda H, Hammarberg H, Piehl F et al. (1999). Expression of
Ezrin-Waters C, Resch L, Lang AE (1985). Coexistence of MHC class I heavy chain and beta2-microglobulin in rat
idiopathic Parkinsons disease and Creutzfeldt-Jakob brainstem motoneurons and nigral dopaminergic neurons.
disease. Can J Neurol Sci 12: 272273. J Neuroimmunol 101: 7686.
Fiszer U (2001). Does Parkinsons disease have an immuno- Litvan II, Jankovic J, Goetz CG et al. (1998). Accuracy of
logical basis? The evidence and its therapeutic implica- the clinical diagnosis of postencephalitic parkinsonism:
tions. BioDrugs 15: 351355. A clinicopathologic study. Eur J Neurol 5: 451457.
384 V. DHAWAN AND K. R. CHAUDHURI

Maggi P, De Mari M, Moramarco A et al. (2000). Parkinson- Shimamura M, Uyama E, Hirano T et al. (2003). A unique
ism in a patient with AIDS and cerebral opportunistic case of sporadic Creutzfeldt-Jacob disease presenting as
granulomatous lesions. Neurol Sci 21: 173176. progressive supranuclear palsy. Intern Med 42: 195198.
Martyn CN (1997). Infection in childhood and neurological Stoessl AJ (1999). Etiology of Parkinsons disease. Can J
diseases in adult life. Br Med Bull 53: 2439. Neurol Sci 26 (Suppl 2), S5S12.
Mirsattari SM, Power C, Nath A (1998). Parkinsonism with Tam S, Barry DP, Beaman L et al. (2002). Neuroinvasive
HIV infection. Mov Disord 13: 684689. Nocardia asteroides GUH-2 induces apoptosis in the sub-
Nath A, Jankovic J, Pettigrew LC (1987). Movement stantia nigra in vivo and dopaminergic cells in vitro. Exp
disorders and AIDS. Neurology 37: 3741. Neurol 177: 453460.
Nitrini R, Teixeira da Silva LS, Rosemberg S et al. (2001). Tolge CF, Factor SA (1991). Focal dystonia secondary to
Prion disease resembling frontotemporal dementia and cerebral toxoplasmosis in a patient with acquired immue
parkinsonism linked to chromosome 17. Arq Neuropsi- deficiency syndrome. Mov disord 6: 6972.
quiatr 59: 161164. Tse W, Cersosimo M, Gracies J et al. (2004). Movement dis-
Peatfield RC (1987). Basal ganglia damage and subcortical orders and AIDS: A review. Parkinsonism Relat Disord
dementia after possible insidious Coxsackie virus ence- 10: 323334.
phalitis. Acta Neurol Scand 76: 340345. Vingerhoets FJ, Hegyi I, Aguzzi A et al. (1998). An unusual
Picard F, de Saint-Martin A, Salmon E et al. (1996). Post- case of Creutzfeldt-Jakob disease. Neurology 51: 617619.
encephalitic stereotyped involuntary movements respon- Werring DJ, Chaudhuri KR (1996). Human Immunodefi-
sive to L-Dopa. Mov Disord 11: 567570. ciency virus-related progressive multifocal leukoencepha-
Poskanzer DC, Schwab RS (1963). Cohort analysis of lopathy presenting with an akinetic rigid syndrome. Mov
Parkinsons disease: Evidence for a single etiology related Disord 11: 758761.
to subclinical infection about 1920. J Chronic Dis 16: Wlodarek D, Pakszys W, Bujko J (2003). Helicobacter
961973. pylori infection and its influence on Parkinsons disease.
Pradhan S, Pandey N, Shashank S et al. (1999). Parkinsonism Pol Merkuriusz Lek 15: 428431.
due to predominant involvement of substantia nigra in Woulfe J, Hoogendoorn H, Tarnopolsky M et al. (2000).
Japanese encephalitis. Neurology 53: 17811786. Monoclonal antibodies against Epstein-Barr virus cross-
Richards M, Chaudhuri KR (1996). Parkinsons disease in react with alpha-synuclein in human brain. Neurology
populations of African origin. Neuroepidemiology 15: 55: 13981401.
214221. Woulfe JM, Duke R, Middeldorp JM et al. (2002). Absence
Salman H, Bergman M, Djaldetti R et al. (1999). Decreased of elevated anti-alpha-synuclein and anti-EBV latent
phagocytic function in patients with Parkinsons disease. membrane protein antibodies in PD. Neurology 58:
Biomed Pharmacother 53: 146148. 14351436.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 50

Toxic causes of parkinsonism

NIRIT LEV*, ELDAD MELAMED AND DANIEL OFFEN

Laboratory of Neuroscience and Department of Neurology, Rabin Medical Center,


Petah-Tikva, Tel Aviv University, Tel Aviv, Israel

50.1. Introduction unwanted proteins leading to the accumulation and


aggregation of cytotoxic proteins, has recently been
Parkinsons disease (PD) is the most common neurode- shown to play a major role in the pathogenesis of both
generative movement disorder, affecting 1% of the familial and sporadic forms of PD (McNaught et al.,
population over 65 years of age. Clinically, most 2002; Elkon et al., 2004).
patients present with a motor disorder and suffer from PD is a multifactorial disease caused by both genetic
bradykinesia, resting tremor, rigidity and postural and environmental factors. However, most PD patients
instability. Other manifestations include behavioral, suffer from a sporadic disease. The etiology of sporadic
cognitive and autonomic disturbances. Pathologically, PD is largely unknown and epidemiological studies
PD is characterized by a progressive loss of dopaminer- are conducted in order to define factors increasing the
gic neurons in the substantia nigra pars compacta. How- risk for developing PD. Factors that may predispose
ever, PD is a widespread neurodegenerative disease, to dopaminergic cell loss, including mitochondrial
which affects multiple areas in the central as well as dysfunction and oxidative damage, could be common
the peripheral nervous systems. The pathologic hall- to separate genetic and environmental etiologies.
mark of PD is the appearance of cytoplasmic inclusions, Several studies were conducted in order to assess
named Lewy bodies. Lewy bodies contain a heteroge- the influence of hereditary factors of PD by studying
neous mixture of insoluble, filamentous proteins and monozygotic (MZ) and dizygotic (DZ) twin pairs.
lipids, including a-synuclein, ubiquitin, neurofilaments These studies, relying on [18F]-dopa positron emission
and oxidized/nitrated proteins (Betarbet et al., 2002). tomography (PET) findings, did find higher concor-
Nigral pathology in parkinsonian patients has been dance for subclinical striatal dopaminergic dysfunction
shown to be associated with oxidative stress, mito- in twins (Burn et al., 1992; Holthoff et al., 1994;
chondrial dysfunction, excitotoxicity and inflamma- Piccini et al., 1999; Laihinen et al., 2000). However,
tion. Oxidative stress-related changes, including several large studies based on clinical evaluation of
oxidative damage to proteins, lipids and DNA, as well PD found low concordance rates for twins, emphasizing
as decreased levels of reduced glutathione, have been environmental factors as the major risk factors for PD
detected in PD brains (Jenner, 1998; Sherer et al., (Duvoisin et al., 1981; Ward et al., 1983; Marttila
2003). Reactive oxygen species (ROS) are generated et al., 1988; Vieregge et al., 1992, 1999; Tanner et al.,
during dopamine metabolism and by mitochondrial 1999; Wirdefeldt et al., 2004, 2005). These findings
respiration (Bahat-Stroomza et al., 2005). Within com- indicate that genetic factors do not play a major role
plex I there is a site of electron leakage that produces in causing typical PD (when the disease begins after
ROS (Kushnareva et al., 2002) and impaired complex the age of 50 years).
I activity enhances ROS formation (Betarbet et al., The search for environmental risk factors for PD
2002; Kushnareva et al., 2002). Failure of another sys- revealed several risk factors as well as protective fac-
tem, the ubiquitin-proteasome system (UPS), to clear tors associated with PD. An inverse association

*Correspondence to: Dr. Nirit Lev, Laboratory of Neuroscience and Department of Neurology, Rabin Medical Center,
Petah-Tikva 49100, Israel. E-mail: lev.nirit@gmail.com, Tel: 972-3-937-6276, Fax: 972-3-937-6277.
386 N. LEV ET AL.
between smoking and PD was reported in many helped develop animal models for this disease. The
studies (Grandinetti et al., 1994; Hernan et al., 2001; two most common, classic animal models of PD rely
Wirdefeldt et al., 2005). Nevertheless, the presence on toxic-induced parkinsonism induced by 1-methyl-
of a confounder has not been ruled out. A recent study 4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and
found an increased risk for PD with several occupa- 6-hydroxydopamine (6-OHDA) (Schober, 2004).
tions: farming, health care and teaching (Goldman Other animal models were generated following the dis-
et al., 2005). There is a growing body of evidence covery that agricultural chemicals, such as rotenone,
implicating rural living, consumption of well water, maneb and paraquat, when administered systemically,
farming and pesticide exposure as environmental risk can also induce specific features of PD (Betarbet
factors for PD (Liou et al., 1997; Priyadarshi et al., et al., 2002). In this chapter, we describe the current
2001). A meta-analysis of 22 epidemiologic studies knowledge of PD induced by neurotoxic compounds.
done by Priyadarshi et al. (2001) found that the odds
ratio (OR) for rural living is 1.56 (95% confidence
50.2. Agricultural chemicals
interval (CI) 1.172.07) and that the highest risk was
4.9 (95% CI 1.418.2) for living in a rural area for
Several clinical and epidemiologic studies have
more than 40 years. Exposure to well water and the risk
demonstrated that exposures to certain chemicals are
of contracting PD yielded a combined OR of 1.26 (95%
associated with increased incidence of PD (Landrigan
CI 0.961.64), increasing to 3.28 (95% CI 0.9311.51)
et al., 2005). Exposure to pesticides yielded an
for exposure to well water for at least 1 year (Priyadarshi
increased risk for getting PD with combined OR of
et al., 2001). Farming was also associated with an
1.85 (95% CI 1.312.60), with exposure for over 10
increased risk of getting PD, with combined OR of
years increasing the risk to 5.81 (95% CI 1.9916.97)
1.42 (95% CI 1.051.91), (OR 5.2, 95% CI 1.617.7,
(Priyadarshi et al., 2001). Baldi et al. (2003) found
for farming over 20 years) (Priyadarshi et al., 2001).
an association between past occupational exposure to
The association of rural living, farming and well-water
pesticides, low cognitive performance and an
drinking with PD may be related to exposure to potential
increased risk of developing PD. Levels of organo-
neurotoxins present in these areas, such as pesticides.
chlorines have been found to be elevated in the brains
In contrast, Tanner et al. (1989) found that in China, liv-
of persons with PD (Fleming et al., 1994; Corrigan
ing in a village was associated with a decreased risk for
et al., 2000). Epidemiologic data suggest a positive
PD, whereas occupational or residential exposure to
doseresponse relationship between lifetime cumulative
industrial chemicals, printing plants or quarries was
exposure to paraquat and risk of PD (Liou et al., 1997).
associated with an increased risk of developing PD.
These findings are consistent with the hypothesis that
environmental exposure to certain agricultural or indus- 50.2.1. Rotenone
trial chemicals may be related to the development of PD.
The concept of silent neurotoxicity suggests that a Rotenone (Fig. 50.1A) is a natural compound, used as
developmental exposure to a toxin may significantly an insecticide and a herbicide. Rotenone is a naturally
increase the vulnerability of the dopaminergic system, occurring complex ketone, derived from the roots of
which is unmasked by later challenges to the dopamine Lonchocarpus species (Uversky, 2004). It is a com-
system (Thiruchelvam et al., 2002; Uversky, 2004; monly used pesticide and is also used in lakes and reser-
Cory-Slechta et al., 2005). Exposure to pesticides dur- voirs to kill fish that are perceived as pests. Gardeners
ing the postnatal period can produce permanent and commonly use it as the active ingredient of derris dust
progressive lesions of the nigrostriatal dopamine sys- or liquid preparations of derris, described as natural
tem and enhanced adult susceptibility to these pesti- herbicide, which are commonly found on the shelves
cides. Inflammation of the brain in early life caused by of garden centers (Jenner, 2001). Rotenone is a highly
exposure to infectious agents, toxicants or environmen- lipophilic compound and readily crosses the blood
tal factors has been suggested as a possible cause or con- brain barrier (Betarbet et al., 2002; Uversky, 2004).
tributor to the later development of PD (Liu et al., Rotenone is a highly selective inhibitor of complex
2003). The inflammatory process in such cases may I of the mitochondrial respiratory chain (Sherer et al.,
involve activation of brain immune cells (microglia 2003). Sherer et al. (2003) demonstrated that rotenone
and astrocytes), which release inflammatory and neuro- toxicity involved an oxidative mechanism. In neuro-
toxic factors that in turn produce neurodegeneration blastoma cells and a chronic midbrain slice model,
(Liu and Hong, 2003). rotenone toxicity depended on an interaction with
The finding of toxic causes of PD had a tremendous complex I and was attenuated by antioxidants, sug-
importance on the scientific study of PD since it gesting a causal role for oxidative damage. Rotenone
TOXIC CAUSES OF PARKINSONISM 387
H2C
CH3

O O
O H3C+N N+CH3
Cr Cr

O O

CH3 O
CH3
A B

NHC2H5
N H2 N
H C
HS C N CS2 Cl N
H2 H N CH3
HS Mn S
H H2 NHCH
N C SH
S2C C N CH3
H2 H
C D
Fig. 50.1. Pesticides and herbicides: chemical structure of (A) rotenone, (B) paraquat, (C) maneb and (D) atrazine.

toxicity depended on a direct interaction with complex I, bodies similar to Lewy bodies (Betarbet et al., 2002).
because cells expressing the rotenone-insensitive, sin- It also induces oxidative stress and mitochondrial
gle-subunit NADH dehydrogenase of yeast, NDI1, complex I inhibition, found in human patients (Betarbet
were resistant to rotenone toxicity. In this system, et al., 2000, 2002; Liu et al., 2003; Sherer et al., 2003).
electrons from complex I substrates are shunted Rotenone infusion by osmotic pumps can induce a
through NDI1 into downstream portions of the elec- chronically progressive degeneration of dopaminergic
tron transport chain, thereby allowing mitochondrial and of some non-dopaminergic neurons in both the
respiration. Rotenone toxicity does not result solely basal ganglia and the brainstem (Hirsch et al., 2003).
from a bioenergetic defect since brain levels of rote- Behaviorally, rotenone induces hypokinesia, flexed
none after chronic exposure are not sufficient to alter posture and, in some rats, rigidity and paw shaking,
mitochondrial respiration in isolated brain mitochon- clinical findings that are similar to PD (Betarbet et al.,
dria (Betarbet et al., 2000). In addition, Sherer et al. 2002).
(2003) found that similar adenosine triphosphate The rotenone-induced degeneration of dopaminergic
depletion induced by 2-deoxyglucose was not toxic as neurons may not be solely attributable to an impairment
rotenone exposure. Panov et al. (2005) recently demon- of neuronal mitochondrial complex I activity but may
strated that it is not the primary inhibitory effect of rote- also involve the activation of microglia (Gao et al.,
none on the electron transport activity of complex I, 2002, 2003b). In vitro, rotenone-induced microglial
but the resulting secondary mechanism, increased activation occurs before apparent neurodegeneration
superoxide radical production, that is responsible (Gao et al., 2002). Activated microglia upregulate
for the development of further structural and func- cell surface markers such as the macrophage antigen
tional damages to the mitochondria. They also found complex I and produce a variety of proinflammatory
that in vivo rotenone toxicity results in a substantial cytokines, leading to ROS production. Synergistic
loss of activity not only of complex I, but also of dopaminergic neurotoxicity was found with combined
complex II, probably mediated by ROS (Panov exposure to rotenone and the inflammogen lipopolysac-
et al., 2005). charide (Gao et al., 2003a).
The animal model induced by chronic systemic Therefore, the rotenone-induced animal model
rotenone administration includes clinical and patholo- for PD recapitulates most of the pathological and clin-
gic features similar to those of human PD, including ical features of PD, as well as central pathophysio-
selective chronic and progressive degeneration of the logical mechanisms of the disease, strengthening the
nigrostriatal system and the formation of inclusion environmental hypothesis of PD.
388 N. LEV ET AL.
50.2.2. Paraquat 50.2.4. Atrazine

An etiologic link has been suggested between PD Atrazine (2-chloro-4-ethylamino-6-isopropylamino-S-


and the herbicide paraquat (1,10 -dimethyl-4,40 -bipyri- triazine) (Fig. 50.1D), a chlorinated member of the
dinium) (Fig. 50.1B) (Brooks, 1999; McCormack family of S-substituted triazines, is one of the most
et al., 2002). Paraquat is structurally similar to 1- widely employed herbicides in the world. It acts to
methyl-4-phenylpyridinium ion (MPP), the active suppress photosynthesis by inhibiting electron trans-
metabolite of MPTP (Uversky, 2004). The major neu- fer at the reducing site of chloroplast complex II
rotoxic effect of paraquat is through the production of (Rodriguez et al., 2005). Although it has limited solu-
ROS (Uversky, 2004). bility in water, atrazine is frequently detected in
Paraquat crosses the bloodbrain barrier slowly, ground and surface waters in agricultural regions
inefficiently and to a limited extent. However, detect- (Rodriguez et al., 2005).
able levels of the herbicide have been measured in the Rodriguez et al. (2005) demonstrated that sustained
central nervous system after its systemic injection into low-level atrazine exposure in the diet can adversely
rodents (Corasaniti et al., 1998). In experimental studies affect dopaminergic tracts of the central nervous sys-
in which paraquat has been administered to animals, tem, resulting in persistent increases in locomotor
researchers have observed loss of substantia nigra dopa- activity, alterations in responsivity to the indirect
minergic neurons, depletion of dopamine, reduced dopamine agonist amphetamine, changes in monoamine
ambulatory activity and apoptotic cell death (Liu and levels and loss of neurons in the midbrain. Chronic atra-
Hong, 2003; Liu et al., 2003). Subchronic exposure to zine exposure caused cell loss of both tyrosine hydr-
paraquat causes a dose-dependent decrease in substantia oxylase (TH)-immunoreactive cells and TH-negative
nigra dopaminergic neurons, some decrease in striatal cells in the ventral tegmental area and substantia nigra
dopamine nerve terminal density and also induces neu- (Rodriguez et al., 2005).
robehavioral syndrome characterized by reduced ambu-
latory activity (Brooks et al., 1999; Di Monte, 2001). 50.2.5. Organochlorine compounds
The exposure of mice to paraquat leads to a significant
increase in brain levels of a-synuclein and accumula- High levels of organochlorine compounds in the substan-
tion of a-synuclein-containing inclusions within neu- tia nigra of PD brains were found, as compared to the
rons of the substantia nigra pars compacta, similar to levels in cortical Lewy body dementia, Alzheimers
Lewy bodies (Manning-Bog et al., 2002). disease and non-demented non-parkinsonian controls
(Corrigan et al., 2000). The levels of the gamma iso-
50.2.3. Maneb mer of hexachlorocyclohexane (gamma-HCH, lindane;
Fig. 50.2A) were significantly higher in PD tissues
The organomanganese fungicide maneb (Fig. 50.1C), than in the other three groups (P < 0 .05). Dieldrin
which is largely used in agricultural regions for the (HEOD) was higher in PD brain than in Alzheimers dis-
control of field crop pathologies, has been implicated ease or control brain, whereas 1,10 -(2,2-dichloroethenyl
in PD. Maneb contains a major active fungicidal com- diene)-bis(4-chlorobenzene) ( p, p-DDE; Fig. 50.2B) and
ponent, manganese ethylene-bis-dithiocarbamate (Mn- total Aroclor-matched polychlorinated biphenyls were
EBDC) and belongs to the dithiocarbamate fungicide
family. Permanent parkinsonism has been reported in
a man with chronic exposure to maneb (Meco et al., Cl
1994). Administration of maneb to experimental ani-
mal models has a depressant-like effect on the central
nervous system, involving the dopaminergic systems Cl
(Uversky, 2004). In animal studies, early-life exposure
to a combination of paraquat and maneb produced Cl Cl
destructive effects on the nigrostriatal dopaminergic Cl
system and abnormalities in motor response that were
more severe than those produced by either agent alone Cl Cl
Cl
(Thiruchelvam et al., 2000). These effects were esca-
Cl Cl
lated by aging (McCormack et al., 2002; Thiruchelvam A B
et al., 2003). Exposure to maneb was also reported to Fig. 50.2. Organochlorine compounds: chemical structure of
enhance MPTP uptake and to amplify its neurotoxicity (A) gamma-hexachlorocyclohexane (lindane) and (B) 1,10 -
(Uversky, 2004). (2,2-dichloroethenyl diene)-bis(4-chlorobenzene ( p,p-DDE).
TOXIC CAUSES OF PARKINSONISM 389
higher in PD compared with cortical Lewy body 50.3.1. Iron
dementia. Bhatt et al. (1999) described 5 patients who
developed acute reversible parkinsonism following A central role of iron in the pathogenesis of PD, due to
organophosphate pesticide exposure. Three of these its increased levels in substantia nigra pars compacta
patients were family-related, therefore a genetic sus- dopaminergic neurons (Dexter et al., 1991, 1992; Double
ceptibility to organochlorine pesticide-induced parkin- et al., 2000) and reactive microglia and its capacity
sonism may account for susceptibility for developing to enhance production of ROS, has been discussed for
this syndrome. many years. Moreover, analysis of Lewy bodies in the
These findings support the hypothesis derived from parkinsonian substantia nigra revealed high levels of iron
epidemiological work and animal studies that organo- and the presence of aluminum (Hirsch et al., 1991).
chlorine insecticides produce a direct toxic action on Besides the accumulation of iron in the substantia
the dopaminergic tracts of the substantia nigra and nigra of PD brains, derangements in iron metabolism
may contribute to the development of PD. were identified. The substantia nigra of the PD brain
is characterized by a shift in the Fe2/Fe3 ratio
50.3. Metals in favor of Fe3 (Berg et al., 2001). Furthermore,
mutated iron metabolism genes have now been shown
The possible involvement of heavy metals in the etiol- to be involved in neurodegeneration (Felletschin et al.,
ogy of PD follows primarily from the results of epide- 2003; Grunblatt et al., 2004). The observations of the
miological studies. In particular, exposure to ability of iron to induce aggregation and toxicity of
manganese, copper, lead, iron, mercury, zinc and alu- a-synuclein have reinforced the critical role of iron
minum appears to be a risk factor for PD (Uversky in the pathogenesis of nigrostriatal injury (Berg et al.,
et al., 2001). Long-term occupational exposure to cop- 2001). Thus, iron redox state constitutes a pivotal fac-
per and manganese individually, as well as to dual tor contributing to the extent of protein misfolding and
combinations of lead, copper and iron, were found to aggregation in the substantia nigra of PD patients.
indicate a significant risk for PD (Gorell et al., 1997, Unilateral injection of FeCl3 into the substantia nigra
1999, 2004). Analysis of the PD mortality rates in of adult rats resulted in a substantial selective decrease
Michigan (19861988) with respect to potential of striatal dopamine (95%) (Ben-Shachar and Youdim,
heavy-metal exposure revealed that counties with an 1991). Dopamine-related behavioral responses, such as
industry in the paper, chemicals, iron or copper-related spontaneous movements in a novel space and rearing, were
categories had significantly higher PD death rates than significantly impaired, whereas amphetamine administra-
counties without these industries (Rybicki et al., tion induced ipsilateral rotation in the iron-treated rats
1993). Another epidemiological study established an (Ben-Shachar and Youdim, 1991). Wesemann et al.
increased risk for PD with occupational exposure to (1994) found that intranigral-injected iron progressively
manganese, iron and aluminum, especially when the reduces striatal dopamine metabolism.
duration of exposure is longer than 30 years (Zayed Neuromelanin has the ability to bind a variety of
et al., 1990). metals and up to 20% of the total iron contained in
Quantifications of aluminum, calcium, copper, iron, the substantia nigra from normal subjects is bound
magnesium, manganese, silicon and zinc were per- within neuromelanin. Increased tissue iron found
formed in urine, serum, blood and cerebrospinal fluid in the parkinsonian substantia nigra may saturate
(CSF) of 26 PD patients (Forte et al., 2004). The iron-chelating sites on neuromelanin (Gerlach et al.,
results indicate the involvement of iron and zinc 2003). It is hypothesized that this redox-active iron
(increased concentration in blood) as well as of copper could be released and involved in a Fenton-like reac-
(decreased serum level) in PD (Forte et al., 2004). tion, leading to an increased production of oxidative
Postmortem analysis of brain tissues from patients radicals. The resultant radical-mediated cytotoxicity
with PD revealed an increase in total iron and zinc may contribute to cellular damage observed in PD.
content of the parkinsonian substantia nigra, whereas These studies indicate that the nigrostriatal dopaminer-
copper levels were reduced and manganese levels were gic neurons are susceptible to the presence of ionic
unchanged (Dexter et al., 1991, 1992). Moreover, sev- iron and support the assumption that iron causes
eral di- and trivalent metal ions caused significant dopaminergic neurodegeneration in PD.
acceleration in the rate of a-synuclein fibril formation
(Uversky et al., 2001). Aluminum was the most effec- 50.3.2. Manganese
tive, along with copper, iron, cobalt and manganese.
The effectiveness correlated with increasing ion Manganese, an essential trace element, is one of the
charge density (Uversky et al., 2001). most used metals in the industry. It is employed in
390 N. LEV ET AL.
the manufacture of steel and batteries, in water purifi- subsequent activation of caspase-9 and caspase-3. Cas-
cation, in bactericidal and fungicide agents and as an pase-3-dependent proteolytic activation of protein
antiknock agent in gasoline. Recently, several new man- kinase C (PKC) delta was demonstrated to play a role
ganese compounds have been introduced as fungicide, as in oxidative stress-mediated apoptosis in dopaminergic
antiknock agent in petrol (Mn tricarbonyl, MMT) and as cells after exposure to manganese (Anantharam et al.,
contrasting agent in nuclear magnetic resonance tomo- 2002).
graphy (MnDPDP) (Gerber et al., 2002). Manganese is
relatively non-toxic to the adult organism, except to the 50.3.3. Copper
brain, where it causes Parkinson-like symptoms when it
is chronically inhaled, even at moderate amounts (Gerber Copper(II) was found to be the most effective metal
et al., 2002). Manganism was reported in manganese ion affecting a-synuclein to form oligomers (Paik
miners (Mergler and Baldwin, 1997). Hudnell (1999) et al., 1999). Copper was recently found to promote
found that the risk of a Parkinson-like syndrome diag- a-synuclein aggregation at physiologically relevant
nosis is increased with continued manganese exposure concentrations by binding to the N-terminus (for
and aging. Chronic exposure to high levels of this metal which copper has the highest affinity) (Rasia et al.,
causes accumulation in the basal ganglia, resulting 2005).
in manganism, characterized by tremors, rigidity and These findings support the notion of PD as a metal-
psychosis. associated neurodegenerative disorder.
A cohort of patients who worked in a manganese-
smelting plant in Taiwan developed parkinsonism that 50.4. MPTP
was almost certainly due to manganese intoxication. Six
of 13 individuals chronically exposed to a very high In 1982 severe Parkinson-like symptoms were described
ambient concentration of manganese developed basal among a group of drug users in northern California who
ganglia syndrome, characterized by gait dysfunction, had taken synthetic heroin contaminated with MPTP
with particular difficulty walking backwards, bradyki- (Fig. 50.3A) (Davis et al., 1979; Langston et al., 1983,
nesia, micrographia and hypophonia (Olanow, 2004). 1999; Ballard et al., 1985). Exposure to MPTP produced
Some of the patients also had dystonia and intermittent bradykinesia and rigidity almost identical to those of
tremor (Olanow, 2004). Patients with manganese- idiopathic PD and severe loss of dopaminergic neurons
induced parkinsonism were reported to have a normal in the substantia nigra (Davis et al., 1979; Langston
striatal fluorodopa uptake on PET (Olanow, 2004). et al., 1999). These patients responded to treatment with
However, a recent study reported decreased putamenal levodopa or bromocriptine, but rapidly developed treat-
fluorodopa uptake on PET (Racette et al., 2005). Dopa- ment-related complications, including fluctuations and
mine transporter (DAT) scan showed a slight decrease dyskinesias (Ballard et al., 1985; Langston et al.,
in the putamen of manganism patients as compared 1999). In some patients, MPTP-induced PD appeared
with that of the normal controls. It seems, then, that almost immediately after exposure, whereas in others,
the presynaptic dopaminergic terminals are not the main onset became evident only months or years later,
targets of chronic manganese intoxication (Huang et al., reflecting a progressive neurodegenerative process.
2003). MPTP was shown to act selectively, specifically in-
Exposure of dopaminergic cells to an organic form juring dopaminergic neurons in the nigrostriatal system
of manganese compound, methylcyclopentadienyl man- in humans as well as in experimental animals (Langston
ganese tricarbonyl (MMT), resulted in a rapid increase et al., 1999). Evidence was also found for ongoing
in the generation of ROS, followed by the release dopaminergic nerve cell loss without Lewy body forma-
of mitochondrial cytochrome c into the cytoplasm and tion in these patients (Langston et al., 1999). Many years

NH2

HO

N CH3
OH

A B OH
Fig. 50.3. Chemical structure of (A) 1-methyl-4-phenylpyridinium ion (MPP) and (B) 6-hydroxydopmaine.
TOXIC CAUSES OF PARKINSONISM 391
later, postmortem examination of brains from persons effects of MPTP on nigral dopaminergic cells were most
exposed to MPTP showed a marked microglial prolif- evident in primates, therefore MPTP-lesioned monkeys
eration in the substantia nigra pars compacta (Orr were developed and have now become the most relevant
et al., 2002). and extensively used animal model of PD (Burns et al.,
MPTP is highly lipophilic and after systemic 1983; Stern, 1990). However, the MPTP-treated primate
administration rapidly crosses the bloodbrain barrier. is a model of selective nigral destruction and does not
Subsequently, the protoxin MPTP is converted to reflect other pathological features of PD. MPTP is
1-methyl-4-phenyl-2,3-dihydropyridium (MPDP) exclu- mainly used in non-human primates and in mice but also
sively in non-dopaminergic cells (especially in astrocytes in several other species, such as dogs, cats, sheep, rats
and serotonergic neurons) by monoamine oxidase B and goldfish (Schober, 2004).
(MAO-B) and then spontaneously oxidizes to MPP Biochemical and histological investigations have
and is released into the extracellular space (Nicklas demonstrated that MPTP-induced parkinsonism reflects
et al., 1987; Przedborski et al., 2001; Przedborski and the human disease and has been important for under-
Vila, 2003). MPP enters the dopaminergic neurons standing the pathophysiology of PD as well as for the
through DAT, as well as via the norepinephrine and ser- evaluation of the effects of various drug therapies and
otonin transporters (Javitch et al. 1985; Mayer et al., efficacy of new surgical techniques such as fetal grafts,
1986). Inside dopaminergic neurons, MPP can bind pallidotomy and deep brain stimulation (Jenner, 2003).
to the vesicular monoamine transporter, which is asso-
ciated with an incorporation of MPP into synaptic vesi- 50.5. Isoquinoline derivatives
cles containing dopamine (Del Zompo et al., 1993).
In addition, MPP can accumulate within mitochondria, Various isoquinoline derivatives were found in the brain
where it inhibits complex I (Nicklas et al., 1987; Mizuno and are considered to be the endogenous neurotoxins
et al., 1987), or it can remain inside the cytoplasm and with neurochemical properties similar to those of MPTP
interact with cytosolic enzymes (Klaidman et al., (Nagatsu, 1997; Antkiewicz-Michaluk, 2002; Naoi et al.,
1993). Since MPP is selectively taken up into dopami- 2002; Storch et al., 2002). Among them, 1,2,3,4-tetrahy-
nergic neurons through the DAT and acts as a potent droisoquinoline (TIQ), 1-benzyl-TIQ and 1-methyl-5,
inhibitor of mitochondrial complex I, it selectively poi- 6-dihydroxy-TIQ (salsolinol) have the most potent neu-
sons the dopaminergic neurons. Surprisingly, Smeyne rotoxic action (Nagatsu, 1997; Antkiewicz-Michaluk,
et al. (2005) found that the susceptibility of substantia 2002).
nigra dopaminergic neurons to MPTP was inversely N-methyl-(R)-salsolinol, a dopamine-derived alka-
related to the number of astrocytes. They also found that loid, selectively occurs in human brains and accu-
Swiss Webster (MPTP-resistant) mice have a greater mulates in the nigrostriatal system (Naoi et al.,
number of astrocytes and a lower number of microglia 2002). N-methyl-(R)-salsolinol is synthesized in the
than C57Bl/6J (MPTP-sensitive) mice. Astrocytes human brain by two enzymes, an (R)-salsolinol
appear to provide protection to neurons against ROS- synthase and an N-methyltransferase, and accumu-
mediated toxicity by multidimensional mechanisms lates in the nigrostriatum in human brains (Maruyama
involving glutathione, phase II detoxifying enzymes et al., 2000). The activity of a neutral N-methyltrans-
and neuronal growth factors (Smeyne et al., 2005). ferase in the striatum was found to determine the
One of the major implications of the discovery of level of MPP-like 1,2-dimethyl-6,7-dihydroxyiso-
MPTP was the production of an effective experimental quinolinium ion, an oxidation product of N-methyl-
model of PD by systemic toxin administration (Jenner, (R)-salsolinol in the substantia nigra (Maruyama
2003). When injected into mice, MPTP causes a PD- et al., 2000). N-methyl-(R)-salsolinol is increased
like syndrome with massive loss of nigral dopaminer- in the CSF of PD patients (Maruyama et al., 1999)
gic neurons and striatal dopamine. MPTP injections and the activity of (R)-salsolinol N-methyltransferase
do not induce neuronal inclusions characteristic of is increased in lymphocytes from PD patients
PD even after repeated injections (Vila et al., 2000). (Maruyama et al., 2000; Naoi et al., 2002).
Fornai et al. (2005) have recently developed a mouse The studies of animal and cellular models of PD
PD model that is based on continuous MPTP administra- proved that salsolinol is selectively cytotoxic to dopa-
tion with an osmotic minipump that produced progres- mine neurons. Exposure of human dopaminergic
sive behavioral changes and triggered severe striatal neuroblastoma cells to salsolinol-induced apoptosis
dopamine depletion with the formation of nigral inclu- by the activation of the apoptotic cascade initiated in
sions immunoreactive for ubiquitin and a-synuclein. the mitochondria (Akao et al., 1999; Naoi et al.,
This continuous MPTP administration also led to the 2000). 2[N]-methylated isoquinoline derivatives struc-
inhibition of the UPS (Fornai et al., 2005). The toxic turally related to MPTP/MPP are selectively toxic to
392 N. LEV ET AL.
dopaminergic cells via uptake by the DAT and there- 6-OHDA models, also known as hemiparkinson
fore may play a role in the pathogenesis of PD (Storch model, the intact hemisphere serves as internal con-
et al., 2002). Cell death induced by salsolinol is due trol structure (Perese et al., 1989). Unilateral 6-OHDA
to impairment of cellular energy supply, caused in injection causes an asymmetric and quantifiable motor
particular by inhibition of mitochondrial complex II behavior (rotations) induced by systemic administra-
(succinate Q reductase) (Storch et al., 2000). On the tion of dopaminergic receptor agonists (e.g. apomor-
other hand, (R)-salsolinol proved to scavenge hydroxyl phine), levodopa or dopamine-releasing drugs (e.g.
radical produced by oxidation of dopamine (Naoi amphetamine).
et al., 1998). The neurotoxicity and neuroprotection
of catechol isoquinolines may be ascribed to their 50.7. Cycad (amyotrophic lateral sclerosis
oxidation and scavenging of radicals. Since PD is a parkinsonism dementia complex of Guam)
slowly progressing neurodegenerative disease, long-
term neurotoxic effects of isoquinolines may have a The Chamorro people of Guam have been afflicted
central role in its progression. with a complex of neurodegenerative diseases known
as amyotrophic lateral sclerosisparkinsonism demen-
50.6. 6-hydroxydopamine tia complex (ALS-PDC) with similarities to ALS,
Alzheimers disease and PD at a far higher rate than
6-OHDA (Fig. 50.3B)-induced nigrostriatal damage other populations throughout the world. Several other
is today one of the most common animal models used foci of endemic ALS-PDC were found in Asia and
in the research of PD. 6-OHDA is a hydroxylated ana- Oceania. Epidemiologic study has shown that prefer-
log of the natural neurotransmitter dopamine (Schober, ence for traditional Chamorro food is significantly asso-
2004). 6-OHDA-induced toxicity is relatively selective ciated with an increased risk for PD (Durlach et al.,
for catecholaminergic neurons, resulting from a prefer- 1997). The toxic cycad seed that was used in traditional
ential uptake of 6-OHDA by dopamine and noradre- food and medicine was found to contain various toxins
nergic transporters (Schober, 2004). The mechanisms and particularly have an excitotoxic amino acid beta-N-
involved in 6-OHDA toxicity include respiratory methylamino-L-alanine (L-BMAA) (Fig. 50.4) (Durlach
inhibition and oxidative stress, induced by free radical
formation. It is toxic to mitochondrial complex I
(Betarbet et al., 2002) and leads to the formation of
superoxide free radicals (Schober, 2004). Furthermore,
it has been shown that 6-OHDA treatment reduces
striatal glutathione and superoxide dismutase enzyme
activity (Schober, 2004).
Several studies have reported the presence of
6-OHDA in both rat and human brains as well as in
the urine of levodopa-treated PD patients (Curtius
et al., 1974; Andrew et al., 1993; Liao et al., 2003;
Maharaj et al., 2005). Dopamine chemically reacts with
iron and ascorbate, resulting in hydroxylated forms of
dopamine products, such as 6-OHDA (Maharaj et al.,
2005). 6-OHDA is easily oxidized and can also take
part in free radical-forming reactions, such as the meta-
bolic monoamine oxidation. Due to the high content of
dopamine, hydrogen peroxide and free iron in dopami-
nergic neurons, a non-enzymatic reaction between these
elements may possibly lead to the endogenous 6-OHDA
formation (Jellinger et al., 1995; Linert et al., 1996). A
Animal models based on 6-OHDA toxicity are
commonly used in the study of PD. Since systemically H2N CH2NHCH3
administered 6-OHDA fails to cross the bloodbrain
barrier, 6-OHDA has to be injected stereotactically COOH
into the brain. Preferred injection sites are the substan- B
tia nigra, medial forebrain bundle and striatum (Perese Fig. 50.4. Cycad toxin. (A) Cycad tree; (B) chemical struc-
et al., 1989; Przedborski et al., 1995). In the unilateral ture of beta-methylaminoalanine (BMAA).
TOXIC CAUSES OF PARKINSONISM 393
et al., 1997; Spencer et al., 2005). Traditional feasting as effective as rotenone but 50-fold more potent
on flying foxes may be related to the prevalence of neu- than the prototypical dopaminergic neurotoxin MPP
ropathologic disease in Guam, since consumption of a (Lannuzel et al., 2003).
single flying fox may have resulted in an equivalent
BMAA dose obtained from eating 1741014 kg of 50.9. Methanol
processed cycad flour, sufficiently high to result in
ALS-PDC neuropathologies (Cox and Sacks, 2002; Several case reports describe the development of
Banack and Cox, 2003). Therefore, cycad neurotoxins parkinsonism as an acute or delayed toxic effect of
are biomagnified within the Guam ecosystem. exposure to vapors of methanol (McLean et al.,
Apoptosis was identified in histological sections of 1980; Ley and Gali, 1983; Verslegers et al., 1988;
brain and gut tissue of adult mice fed with seed prepara- Indakoetxea et al., 1990; Davis and Adair, 1999;
tions of cycad (Gobe, 1994). Shaw and Wilson (2003) Hageman et al., 1999; Finkelstein and Vardi, 2002).
developed an animal model of ALS-PDC which mimics Methanol intoxication caused development of perma-
all the essential features of the disease by feeding the nent parkinsonism (McLean et al., 1980; Ley and
animals with washed cycad seeds. Gali, 1983; Verslegers et al., 1988; Indakoetxea et al.,
1990). Chronic exposure, even without episodes of
50.8. Annonacin acute intoxication, caused parkinsonism, pyramidal
signs, cognitive decline and unresponsiveness to levo-
An unusually high percentage of atypical forms of par- dopa (Hageman et al., 1999; Finkelstein and Vardi,
kinsonism were discovered in Guadeloupe (French 2002). The setting of prolonged exposure to methanol
West Indies). The patients are characterized by levo- vapors may exist in research laboratories and other
dopa-resistant symmetrical bradykinesia and rigidity, environments and pose the risk of similar delayed toxic
postural instability, dementia and, after 25 years, influences.
pseudobulbar palsy. The early occurrence of postural
instability followed by vertical supranuclear palsy sup- 50.10. Carbon monoxide poisoning
ported the diagnosis of progressive supranuclear palsy
(PSP) in one-third of patients (Caparros-Lefebvre Carbon monoxide (CO) exposure is a common cause
et al., 1999, 2002). of toxic brain damage and its effects range from tran-
The neurological syndrome was linked to regular sient neurological dysfunction to coma and death
consumption of tropical plants of the Annonaceae (Prockop, 2005). Parkinsonism is a common sequel
family, in particular Annona muricata (synonyms: cor- of CO poisoning (Lee and Marsden, 1994; Choi and
ossol, soursop, guanabana, graviola), used for alimen- Cheon, 1999; Prockop, 2005). Choi and Cheon
tary and medicinal purposes (Lannuzel et al., 2003). (1999) followed 242 patients with CO poisoning.
A similar clinical syndrome has also been reported in Delayed movement disorder affected 13.2% of these
Afro-Caribbean and Indian immigrants in England patients, of whom 71.9% suffered from parkinsonism
who regularly consumed imported annonacea products (9.5% of all patients with CO poisoning) (Choi and
(Chaudhuri et al., 2000). The validity of this associa- Cheon, 1999). Other movement disorders described
tion was strengthened by the partial regression of after CO poisoning include dystonia, chorea, athetosis,
symptoms in young patients who stopped consuming tremor and myoclonus (Choi and Cheon, 1999).
these plants (Caparros-Lefebvre et al., 1999). Movement disorders can develop during or immedi-
Annonacin (Fig. 50.5) is highly toxic to dopaminer- ately after acute CO poisoning, but are usually delayed
gic and other mesencephalic neurons via a mechanism for weeks after acute anoxia. The median latency
that is not excitotoxic and occurs independently of between CO poisoning and the onset of parkinsonism
ROS (Lannuzel et al., 2003). Some of the compounds was 4 weeks (Choi and Cheon, 1999). Most patients
found in A. muricata are potent complex I inhibitors suffering from parkinsonism recovered gradually
(Lannuzel et al., 2003). Annonacin was approximately within 6 months (Choi and Cheon, 1999).

O O CH3
OH OH OH OH
O
H3C (H2C)10 (H2C)4 (H2C)5

Fig. 50.5. Chemical structure of annonacin.


394 N. LEV ET AL.
Brain computed tomography findings of patients exposure to toxins may also induce an increased vulner-
with parkinsonism after CO poisoning shows low- ability and enhanced adult susceptibility to toxins.
density lesions bilaterally in the white matter of the cere- These models of synergistic toxicity, as well as chemi-
bral cortex and in the globus pallidus, which can also be cal-induced inflammatory response, may underlie the
seen in non-parkinsonian patients with CO poisoning effects of environmental agents on the pathogenesis of
(Lee and Marsden, 1994; Choi and Cheon, 1999). A spec- PD. Another important factor is the individual sus-
trum of severity of magnetic resonance imaging (MRI) ceptibility to a certain toxin defined by susceptibility
findings after CO poisoning was described, including glo- genes in PD, therefore a combination of genetic and
bus pallidus and white-matter lesions (Sohn et al., 2000; environmental factors is needed to produce the disease.
Parkinson et al., 2002; Prockop, 2005). MR spectroscopy Revelation of the environmental factors that lead to
(MRS) shows decreased n-acetyl aspartase in the white PD enabled the development of toxin-induced animal
matter and basal ganglia in some of the patients (Sohn models for PD. These models have a critical role in
et al., 2000; Prockop, 2005). the research of PD and enable the development and
Hara et al. (2002) studied the effects of CO expo- assessment of new therapeutic strategies.
sure on the dopaminergic system in rats by in vivo
brain microdialysis. Exposure of rats to CO (up to References
0.3%) caused a marked increase in extracellular dopa-
mine in the striatum, probably through stimulation of Akao Y, Nakagawa Y, Maruyama W et al. (1999). Apoptosis
sodium-dependent dopamine release in addition to induced by an endogenous neurotoxin, N-methyl(R)salso-
suppressing dopamine metabolism. CO withdrawal linol, is mediated by activation of caspase 3. Neurosci Lett
and subsequent reoxygenation enhanced the oxidative 267: 153156.
metabolism of the increased extracellular dopamine, Anantharam V, Kitazawa M, Wagner J et al. (2002). Cas-
preferentially mediated by MAO-A (Hara et al., pase-3-dependent proteolytic cleavage of protein kinase
2002). These findings might imply that CO toxicity Cdelta is essential for oxidative stress-mediated dopami-
to the dopaminergic system is mediated, at least par- nergic cell death after exposure to methylcyclopentadienyl
manganese tricarbonyl. J Neurosci 22: 17381751.
tially, through the toxicity of dopamine and its reactive
Andrew R, Watson DG, Best SA et al. (1993). The determi-
substances, such as quinones and activated oxygen
nation of hydroxydopamines and other trace amines in the
species, generated via dopamine oxidation. urine of parkinsonian patients and normal controls. Neuro-
chem Res 18: 11751177.
50.11. Conclusions Antkiewicz-Michaluk L (2002). Endogenous risk factors in
Parkinsons disease: dopamine and tetrahydroisoquino-
PD is a common and mostly sporadic disease. The lines. Pol J Pharmacol 54: 567572.
pathogenesis of PD is likely multifactorial and involves Bahat-Stroomza M, Gilgun-Sherki Y, Offen D et al. (2005).
genetic and environmental interactions. The discovery A novel thiol antioxidant that crosses the blood brain bar-
that MPTP could induce parkinsonism was the first sub- rier protectsdopaminergic neurons in experimental models
stantial evidence that this complex disease could result of Parkinsons disease. Eur J Neurosci 21: 637646.
Baldi I, Lebailly P, Mohammed-Brahim B et al. (2003). Neu-
from a single toxin. Evidence from multiple epidemio-
rodegenerative diseases and exposure to pesticides in the
logical studies emphasizes the roles of environmental
elderly. Am J Epidemiol 157: 409414.
risk factors. New findings arising from toxin-induced Ballard PA, Tetrud JW, Langston JW (1985). Permanent human
animal models of PD highlight the possibility that parkinsonism due to 1-methyl-4-phenyl-1,2,3,6-tetrahydro-
selective injury to the striatonigral dopaminergic sys- pyridine (MPTP): seven cases. Neurology 35: 949956.
tem may arise not only from the distinctive properties Banack SA, Cox PA (2003). Biomagnification of cycad neu-
of the toxin, but also from intrinsic susceptibility of rotoxins in flying foxes: implications for ALS-PDC in
the dopaminergic neurons. Guam. Neurology 61: 387389.
The dopaminergic neurons are more vulnerable Ben-Shachar D, Youdim MB (1991). Intranigral iron injec-
than other cell populations to mitochondrial complex tion induces behavioral and biochemical parkinsonism
I inhibition and oxidative stress and several toxins in rats. J Neurochem 57: 21332135.
Berg D, Gerlach M, Youdim MB et al. (2001). Brain iron
implicated in PD were demonstrated to act through
pathways and their relevance to Parkinsons disease.
these mechanisms. These two mechanisms may be
J Neurochem 79: 225236.
interrelated, since inhibition of complex I results in Betarbet R, Sherer TB, MacKenzie G et al. (2000). Chronic sys-
augmented production of ROS. temic pesticide exposure reproduces features of Parkinsons
Combinations of environmental risk factors may disease. Nat Neurosci 3: 13011306.
result in more profound nigrostriatal damage, as Betarbet R, Sherer TB, Greenamyre JT (2002). Animal mod-
was shown in animal and in vitro studies. Early-life els of Parkinsons disease. Bioessays 24: 308318.
TOXIC CAUSES OF PARKINSONISM 395

Bhatt MH, Elias MA, Mankodi AK (1999). Acute and rever- Dexter DT, Jenner P, Schapira AH et al. (1992). Alterations in
sible parkinsonism due to organophosphate pesticide levels of iron, ferritin, and other trace metals in neurode-
intoxication: five cases. Neurology 52: 14671471. generative diseases affecting the basal ganglia. The Royal
Brooks AI, Chadwick CA, Gelbard HA et al. (1999). Kings and Queens Parkinsons Disease Research Group.
Paraquat elicited neurobehavioral syndrome caused by Ann Neurol 32: S94S100.
dopaminergic neuron loss. Brain Res 823: 110. Di Monte DA (2001). The role of environmental agents in
Burn DJ, Mark MH, Playford ED et al. (1992). Parkinsons Parkinsons disease. Clin Neurosci Res 1: 419426.
disease in twins studied with 18F-dopa and positron emis- Double KL, Gerlach M, Youdim MB et al. (2000). Impaired
sion tomography. Neurology 42: 18941900. iron homeostasis in Parkinsons disease. J Neural Transm
Burns RS, Chiueh CC, Markey SP et al. (1983). A primate Suppl 60: 3758.
model of parkinsonism: selective destruction of dopami- Durlach J, Bac P, Durlach V et al. (1997). Are age-related
nergic neurons in the pars compacta of the substantia nigra neurodegenerative diseases linked with various types of
by N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc magnesium depletion? Magnes Res 10: 339353.
Natl Acad Sci USA 80: 45464550. Duvoisin RC, Eldridge R, Williams A et al. (1981). Twin
Caparros-Lefebvre D, Elbaz A (1999). Caribbean study of Parkinson disease. Neurology 31: 7780.
Parkinsonism Study Group. Possible relation of atypical Elkon H, Melamed E, Offen D (2004). Oxidative stress,
parkinsonism in the French West Indies with consumption induced by 6-hydroxydopamine, reduces proteasome
of tropical plants: a case-control study. Lancet 354: activities in PC12 cells: implications for the pathogenesis
281286. of Parkinsons disease. J Mol Neurosci 24: 387400.
Caparros-Lefebvre D, Sergeant N, Lees A et al. (2002). Guade- Felletschin B, Bauer P, Walter U et al. (2003). Screening for
loupean parkinsonism: a cluster of progressive supranuclear mutations of the ferritin light and heavy genes in Parkinsons
palsy-like tauopathy. Brain 125: 801811. disease patients with hyperechogenicity of the substantia
Chaudhuri KR, Hu MTM, Brooks DJ (2000). Atypical par- nigra. Neurosci Lett 352: 5356.
kinsonism in Afro-Caribbean and Indian origin immi- Finkelstein Y, Vardi J (2002). Progressive parkinsonism in a
grants to the UK. Mov Disord 15: 1823. young experimental physicist following long-term expo-
Choi IS, Cheon HY (1999). Delayed movement disorders sure to methanol. Neurotoxicology 23: 521525.
after carbon monoxide poisoning. Eur Neurol 42: 141144. Fleming L, Mann JB, Bean J et al. (1994). Parkinsons dis-
Corasaniti MT, Strongoli MC, Rotiroti D et al. (1998). Para- ease and brain levels of organochlorine pesticides. Ann
quat: a useful tool for the in vivo study of mechanisms of Neurol 36: 100103.
neuronal cell death. Pharmacol Toxicol 83: 17. Fornai F, Schluter OM, Lenzi P et al. (2005). Parkinson-
Corrigan FM, Wienburg CL, Shore RF et al. (2000). Orga- like syndrome induced by continuous MPTP infusion:
nochlorine insecticides in substantia nigra in Parkinsons convergent roles of the ubiquitinproteasome system
disease. J Toxicol Environ Health A 59: 229234. and alpha-synuclein. Proc Natl Acad Sci USA 102:
Cory-Slechta DA, Thiruchelvam M, Barlow BK et al. (2005). 34133418.
Developmental pesticide models of the Parkinson disease Forte G, Bocca B, Senofonte O et al. (2004). Trace and
phenotype. Environ Health Perspect 113: 12631270. major elements in whole blood, serum, cerebrospinal fluid
Cox PA, Sacks OW (2002). Cycad neurotoxins, consumption and urine of patients with Parkinsons disease. J Neural
of flying foxes, and ALS-PDC disease in Guam. Neurol- Transm 111: 10311040.
ogy 58: 956959. Gao HM, Hong JS, Zhang W et al. (2002). Distinct role for
Curtius HC, Wolfensberger M, Steinmann B et al. (1974). Mass microglia in rotenone-induced degeneration of dopaminer-
fragmentography of dopamine and 6-hydroxydopamine. gic neurons. J Neurosci 22: 782790.
Application to the determination of dopamine in human Gao HM, Hong JS, Zhang W et al. (2003a). Synergistic
brain biopsies from the caudate nucleus. J Chromatogr 99: dopaminergic neurotoxicity of the pesticide rotenone and
529540. inflammogen lipopolysaccharide: relevance to the etiology
Davis GC, Williams AC, Markey SP et al. (1979). Chronic of Parkinsons disease. J Neurosci 23: 12281236.
Parkinsonism secondary to intravenous injection of meper- Gao HM, Liu B, Hong JS (2003b). Critical role for micro-
idine analogues. Psychiatry Res 1: 249254. glial NADPH oxidase in rotenone-induced degeneration
Davis LE, Adair JC (1999). Parkinsonism from methanol of dopaminergic neurons. J Neurosci 23: 61816187.
poisoning: benefit from treatment with anti-Parkinson Gerber GB, Leonard A, Hantson P (2002). Carcinogenicity,
drugs. Mov Disord 14: 520522. mutagenicity and teratogenicity of manganese compounds.
Del Zompo M, Piccardi MP, Ruiu S et al. (1993). Selective Crit Rev Oncol Hematol 42: 2534.
MPP uptake into synaptic dopamine vesicles: possible Gerlach M, Double KL, Ben-Shachar D et al. (2003). Neuro-
involvement in MPTP neurotoxicity. Br J Pharmacol melanin and its interaction with iron as a potential risk
109: 411414. factor for dopaminergic neurodegeneration underlying
Dexter DT, Carayon A, Javoy-Agid F et al. (1991). Alterations Parkinsons disease. Neurotox Res 5: 3544.
in the levels of iron, ferritin and other trace metals in Gobe GC (1994). Apoptosis in brain and gut tissue of mice
Parkinsons disease and other neurodegenerative diseases fed a seed preparation of the cycad Lepidozamia peroffs-
affecting the basal ganglia. Brain 114: 19531975. kyana. Biochem Biophys Res Commun 205: 327333.
396 N. LEV ET AL.

Goldman SM, Tanner CM, Olanow CW et al. (2005). Occu- selective toxicity. Proc Natl Acad Sci USA 82:
pation and parkinsonism in three movement disorders 21732177.
clinics. Neurology 65: 14301435. Jellinger K, Linert L, Kienzl E et al. (1995). Chemical evi-
Gorell JM, Johnson CC, Rybicki BA et al. (1997). Occupa- dence for 6-hydroxydopamine to be an endogenous toxic
tional exposures to metals as risk factors for Parkinsons factor in the pathogenesis of Parkinsons disease. J Neural
disease. Neurology 48: 650658. Transm 46: 297314.
Gorell JM, Rybicki BA, Cole Johnson C et al. (1999). Occupa- Jenner P (1998). Oxidative mechanisms in nigral cell death
tional metal exposures and the risk of Parkinsons disease. in Parkinsons disease. Mov Disord 13: 2443.
Neuroepidemiology 18: 303308. Jenner P (2001). Parkinsons disease, pesticides and mito-
Gorell JM, Peterson EL, Rybicki BA et al. (2004). Multiple chondrial dysfunction. Trends Neurosci 24: 245246.
risk factors for Parkinsons disease. J Neurol Sci 217: Jenner P (2003). The contribution of the MPTP-treated pri-
169174. mate model to the development of new treatment strate-
Grandinetti A, Morens DM, Reed D et al. (1994). Prospective gies for Parkinsons disease. Parkinsonism Relat Disord
study of cigarette smoking and the risk of developing 9: 131137.
idiopathic Parkinsons disease. Am J Epidemiol 139: Klaidman LK, Adams JD Jr, Leung AC et al. (1993). Redox
11291138. cycling of MPP: evidence for a new mechanism invol-
Grunblatt E, Mandel S, Jacob-Hirsch J et al. (2004). Gene ving hydride transfer with xanthine oxidase, aldehyde
expression profiling of parkinsonian substantia nigra pars dehydrogenase, and lipoamide dehydrogenase. Free Radic
compacta; alterations in ubiquitin-proteasome, heat shock Biol Med 15: 169179.
protein, iron and oxidative stress regulated proteins, cell Kushnareva Y, Murphy AN, Andreyev A (2002). Complex I-
adhesion/cellular matrix and vesicle trafficking genes. mediated reactive oxygen species generation: modulation
J Neural Transm 111: 15431573. by cytochrome c and NAD(P) oxidation-reduction state.
Hageman G, van der Hoek J, van Hout M et al. (1999). Par- Biochem J 368: 545553.
kinsonism, pyramidal signs, polyneuropathy, and cognitive Laihinen A, Ruottinen H, Rinne JO et al. (2000). Risk for
decline after long-term occupational solvent exposure. Parkinsons disease: twin studies for the detection of
J Neurol 246: 198206. asymptomatic subjects using [18F]6-fluorodopa PET.
Hara S, Mukai T, Kurosaki K et al. (2002). Modification of J Neurol 247: II110II113.
the striatal dopaminergic neuron system by carbon monox- Landrigan PJ, Sonawane B, Butler RN et al. (2005). Early
ide exposure in free-moving rats, as determined by in vivo environmental origins of neurodegenerative disease in
brain microdialysis. Arch Toxicol 76: 596605. later life. Environ Health Perspect 113: 12301233.
Hernan MA, Zhang SM, Rueda-deCastro AM et al. (2001). Langston JW, Ballard P, Tetrud JW et al. (1983). Chronic
Cigarette smoking and the incidence of Parkinsons dis- Parkinsonism in humans due to a product of meperidine-
ease in two prospective studies. Ann Neurol 50: 780786. analog synthesis. Science 219: 979980.
Hirsch EC, Brandel JP, Galle P et al. (1991). Iron and alu- Langston JW, Forno LS, Tetrud J et al. (1999). Evidence of
minum increase in the substantia nigra of patients with active nerve cell degeneration in the substantia nigra of
Parkinsons disease: an X-ray microanalysis. J Neurochem humans years after 1-methyl-4-phenyl-1,2,3,6-tetrahydro-
56: 446451. pyridine exposure. J Ann Neurol 46: 598605.
Hirsch EC, Hoglinger G, Rousselet E et al. (2003). Animal Lannuzel A, Michel PP, Oglinger GUH et al. (2003). The
models of Parkinsons disease in rodents induced by tox- mitochondrial complex I inhibitor annonacin is toxic to
ins: an update. J Neural Transm Suppl 65: 89100. mesencephalic dopaminergic neurons by impairment of
Holthoff VA, Vieregge P, Kessler J et al. (1994). Discordant energy metabolism. Neuroscience 121: 287296.
twins with Parkinsons disease: positron emission tomo- Lee MS, Marsden CD (1994). Neurological sequelae following
graphy and early signs of impaired cognitive circuits. carbon monoxide poisoning clinical course and outcome
Ann Neurol 36: 176182. according to the clinical types and brain computed tomogra-
Huang CC, Weng YH, Lu CS et al. (2003). Dopamine trans- phy scan findings. Mov Disord 9: 550558.
porter binding in chronic manganese intoxication. J Neu- Ley CO, Gali FG (1983). Parkinsonian syndrome after
rol 250: 13351339. methanol intoxication. Eur Neurol 22: 405459.
Hudnell HK (1999). Effects from environmental Mn expo- Liao PC, Kuo YM, Chang YC et al. (2003). Striatal formation
sures: a review of the evidence from non-occupational of 6-hydroxydopamine in mice treated with pargyline, pyro-
exposure studies. Neurotoxicology 20: 379397. gallol and methamphetamine. J Neural Trans 110: 487494.
Indakoetxea B, Lopez de Munain A, Marti-Masso JF et al. Linert W, Herlinger E, Jameson RF et al. (1996). Dopamine,
(1990). Parkinsonism after methyl alcohol poisoning. Neu- 6-hydroxydopamine, iron, and dioxygentheir mutual
rologia 5: 238241. interactions and possible implication in the development of
Javitch JA, DAmato RJ, Strittmatter SM et al. (1985). Parkinsons disease. Biochim Biophys Acta 1316: 160168.
Parkinsonism-inducing neurotoxin, N-methyl-4-phenyl- Liou HH, Tsai MC, Chen CJ et al. (1997). Environmental
1,2,3,6-tetrahydropyridine: uptake of the metabolite risk factors and Parkinsons disease: a case-control study
N-methyl-4-phenylpyridine by dopamine neurons explains in Taiwan. Neurology 48: 15831588.
TOXIC CAUSES OF PARKINSONISM 397

Liu B, Hong JS (2003). Role of microglia inflammation- Naoi M, Maruyama W, Kasamatsu T et al. (1998). Oxidation
mediated neurodegenerative diseases: mechanisms and of N-methyl(R)salsolinol: involvement to neurotoxicity
strategies for therapeutic intervention. J Pharmacol Exp and neuroprotection by endogenous catechol isoquino-
Ther 304: 17. lines. J Neural Transm Suppl 52: 125138.
Liu B, Gao H, Hong J (2003). Parkinsons disease and expo- Naoi M, Maruyama W, Takahashi T et al. (2000). Involve-
sure to infectious agents and pesticides and the occurrence ment of endogenous N-methyl(R)salsolinol in Parkinsons
of brain injuries: role of neuroinflammation. Environ disease: induction of apoptosis and protection by deprenyl.
Health Perspect 111: 10651073. J Neural Transm Suppl 58: 111121.
Maharaj H, Sukhdev Maharaj D, Scheepers M et al. (2005). Naoi M, Maruyama W, Akao Y et al. (2002). Dopamine-
l-DOPA administration enhances 6-hydroxydopamine derived endogenous N-methyl-(R)-salsolinol: its role in
generation. Brain Res 1063: 180186. Parkinsons disease. Neurotoxicol Teratol 24: 579591.
Manning-Bog AB, McCormack AL, Li J et al. (2002). The Nicklas WJ, Youngster SK, Kindt MV et al. (1987). MPTP,
herbicide paraquat causes up-regulation and aggregation MPP and mitochondrial function. Life Sci 40: 721729.
of alpha-synuclein in miceparaquat and alpha-synu- Olanow CW (2004). Manganese-induced parkinsonism and
clein. J Biol Chem 277: 16411644. Parkinsons disease. Ann NY Acad Sci 1012: 209223.
Marttila RJ, Kaprio J, Koskenvuo M et al. (1988). Parkinsons Orr CF, Rowe DB, Halliday GM (2002). An inflammatory
disease in a nationwide twin cohort. Neurology 38: review Parkinsons disease. Prog Neurobiol 68: 325340.
12171219. Paik SR, Shin HJ, Lee JH et al. (1999). Copper(II)-induced
Maruyama W, Abe T, Tohgi H et al. (1999). An endogenous self-oligomerization of alpha-synuclein. Biochem J 340:
MPTP-like dopaminergic neurotoxin, N-methyl(R)salsoli- 821828.
nol, in the cerebrospinal fluid decreases with progression Panov A, Dikalov S, Shalbuyeva N et al. (2005). Rotenone
of Parkinsons disease. Neurosci Lett 262: 1316. model of Parkinson disease: multiple brain mitochondria
Maruyama W, Strolin-Benedetti M, Naoi M (2000). N- dysfunctions after short term systemic rotenone intoxica-
methyl(R)salsolinol and a neutral N-methyltransferase as tion. J Biol Chem 280: 4202642035.
pathogenic factors in Parkinsons disease. Neurobiology Parkinson RB, Hopkins RO, Cleavinger HB et al.
8: 5568. (2002). White matter hyperintensities and neuropsycholo-
Mayer RA, Kindt MV, Heikkila RE (1986). Prevention of gical outcome following carbon monoxide poisoning.
the nigrostriatal toxicity of 1-methyl-4-phenyl-1,2,3,6-tet- Neurology 58: 15251532.
rahydropyridine by inhibitors of 3,4-dihydroxyphenylethy- Perese DA, Ulman J, Viola J et al. (1989). A 6-hydroxydopa-
lamine transport. J Neurochem 47: 10731079. mine-induced selective parkinsonian rat model. Brain Res
McCormack AL, Thiruchelvam M, Manning-Bog AB et al. 494: 285293.
(2002). Environmental risk factors and Parkinsons dis- Piccini P, Burn DJ, Ceravolo R et al. (1999). The role of
ease: selective degeneration of nigral dopaminergic neu- inheritance in sporadic Parkinsons disease: evidence from
rons caused by the herbicide paraquat. Neurobiol Dis 10: a longitudinal study of dopaminergic function in twins.
119127. Ann Neurol 45: 577582.
McLean DR, Jacobs H, Mielke BW (1980). Methanol poi- Priyadarshi A, Khuder SA, Schaub EA et al. (2001). Environ-
soning: a clinical and pathological study. Ann Neurol 8: mental risk factors and Parkinsons disease: a metaanaly-
161167. sis. Environ Res 86: 122127.
McNaught KS, Mytilineou C, Jnobaptiste R et al. Prockop LD (2005). Carbon monoxide brain toxicity: clini-
(2002). Impairment of the ubiquitin-proteasome system cal, magnetic resonance imaging, magnetic resonance
causes dopaminergic cell death and inclusion body forma- spectroscopy, and neuropsychological effects in 9 people.
tion in ventral mesencephalic cultures. J Neurochem 81: J Neuroimaging 15: 144149.
301306. Przedborski S, Vila M (2003). The 1-methyl-4-phenyl-
Meco G, Bonifati V, Vanacore N et al. (1994). Parkinsonism 1,2,3,6-tetrahydropyridine mouse model: a tool to explore
after chronic exposure to the fungicide maneb (manganese the pathogenesis of Parkinsons disease. Ann NY Acad Sci
ethylene-bis-dithiocarbamate). Scand J Work Environ 991: 189198.
Health 20: 301305. Przedborski S, Levivier M, Jiang H et al. (1995). Dose-
Mergler D, Baldwin M (1997). Early manifestations of man- dependent lesions of the dopaminergic nigrostriatal path-
ganese neurotoxicity in humans: an update. Environ Res way induced by intrastriatal injection of 6-hydroxydopa-
73: 92100. mine. Neuroscience 67: 631647.
Mizuno Y, Sone N, Saitoh T (1987). Effects of 1-methyl-4- Przedborski S, Jackson-Lewis V, Naini AB et al. (2001). The
phenyl-1,2,3,6-tetrahydropyridine and 1-methyl-4-phenyl- parkinsonian toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydro-
pyridinium ion on activities of the enzymes in the electron pyridine (MPTP): a technical review of its utility and
transport system in mouse brain. J Neurochem 48: safety. J Neurochem 76: 12651274.
17871793. Racette BA, Antenor JA, McGee-Minnich L et al. (2005).
Nagatsu T (1997). Isoquinoline neurotoxins in the brain and [18F]FDOPA PET and clinical features in parkinsonism
Parkinsons disease. Neurosci Res 29: 99111. due to manganism. Mov Disord 20: 492496.
398 N. LEV ET AL.

Rasia RM, Bertoncini CW, Marsh D et al. (2005). Structural Thiruchelvam M, Richfield EK, Goodman BM et al. (2002).
characterization of copper(II) binding to a-synuclein: Developmental exposure to the pesticides paraquat and
insights into the bioinorganic chemistry of Parkinsons maneb and the Parkinsons disease phenotype. Neurotoxi-
disease. Proc Natl Acad Sci USA 102: 42944299. cology 23: 621633.
Rodriguez VM, Thiruchelvam M, Cory-Slechta DA (2005). Thiruchelvam M, McCormack A, Richfield EK et al. (2003).
Sustained exposure to the widely used herbicide atrazine: Age-related irreversible progressive nigrostriatal dopami-
altered function and loss of neurons in brain monoamine nergic neurotoxicity in the paraquat and maneb model of
systems. Environ Health Perspect 113: 708715. the Parkinsons disease phenotype. Eur J Neurosci 18:
Rybicki BA, Johnson CC, Uman J et al. (1993). Parkinsons 589600.
disease mortality and the industrial use of heavy metals in Uversky VN (2004). Neurotoxicant-induced animal models
Michigan. Mov Disord 8: 8792. of Parkinsons disease: understanding the role of rotenone,
Schober A (2004). Classic toxin-induced animal models of maneb and paraquat in neurodegeneration. Cell Tissue Res
Parkinsons disease: 6-OHDA and MPTP. Cell Tissue 318: 225241.
Res 318: 215224. Uversky VN, Li J, Fink AL (2001). Metal-triggered struc-
Shaw CA, Wilson JM (2003). Analysis of neurological disease tural transformations, aggregation, and fibrillation of
in four dimensions: insight from ALS-PDC epidemiology human alpha-synuclein: a possible molecular link between
and animal models. Neurosci Biobehav Rev 27: 493505. Parkinsons disease and heavy metal exposure. J Biol
Sherer TB, Kim J-H, Betarbet R et al. (2003). Subcutaneous Chem 276: 4428444296.
rotenone exposure causes highly selective dopaminergic Verslegers W, Van den Kerchove M, Crols R et al. (1988).
degeneration and alpha-synuclein aggregation. Exp Neurol Methanol intoxication. Parkinsonism and decreased Met-
179: 916. enkephalin levels due to putaminal necrosis. Acta Neurol
Belg 88: 163171.
Smeyne M, Jiao Y, Shepherd KR et al. (2005). Glia cell
Vieregge P, Schiffke KA, Friedrich HJ et al. (1992). Parkinsons
number modulates sensitivity to MPTP in mice. Glia 52:
disease in twins. Neurology 42: 14531461.
144152.
Vieregge P, Hagenah J, Heberlein I et al. (1999). Parkinsons
Sohn YH, Jeong Y, Kim HS et al. (2000). The brain lesion
disease in twins: a follow-up study. Neurology 53: 566572.
responsible for parkinsonism after carbon monoxide poi-
Vila M, Vukosavic S, Jackson-Lewis V et al. (2000). Alpha-
soning. Arch Neurol 57: 12141218.
synuclein up-regulation in substantia nigra dopaminergic
Spencer PS, Palmer VS, Ludolph AC (2005). On the decline and
neurons following administration of the parkinsonian toxin
etiology of high-incidence motor system disease in West
MPTP. J Neurochem 74: 721729.
Papua (southwest New Guinea). Mov Disord 20: S119S126.
Ward CD, Duvoisin RC, Ince SE et al. (1983). Parkinsons
Stern Y (1990). MPTP-induced parkinsonism. Prog Neurobiol
disease in 65 pairs of twins and in a set of quadruplets.
34: 107114.
Neurology 33: 815824.
Storch A, Kaftan A, Burkhardt K et al. (2000). 1-Methyl-
Wesemann W, Blaschke S, Solbach M et al. (1994). Intrani-
6,7-dihydroxy-1,2,3,4-tetrahydroisoquinoline (salsolinol)
gral injected iron progressively reduces striatal dopamine
is toxic to dopaminergic neuroblastoma SH-SY5Y cells
metabolism. J Neural Transm Park Dis Dement Sect 8:
via impairment of cellular energy metabolism. Brain Res
209214.
855: 6775.
Wirdefeldt K, Gatz M, Schalling M et al. (2004). No evidence
Storch A, Ott S, Hwang YI et al. (2002). Selective dopami-
for heritability of Parkinson disease in Swedish twins.
nergic neurotoxicity of isoquinoline derivatives related to
Neurology 63: 305311.
Parkinsons disease: studies using heterologous expression
Wirdefeldt K, Gatz M, Pawitan Y et al. (2005). Risk and pro-
systems of the dopamine transporter. Biochem Pharmacol
tective factors for Parkinsons disease: a study in Swedish
63: 909920.
twins. Ann Neurol 57: 2733.
Tanner CM, Chen B, Wang W et al. (1989). Environmental
Zayed J, Ducic S, Campanella G et al. (1990). Environmental
factors and Parkinsons disease: a case-control study in
factors in the etiology of Parkinsons disease. Can J Neurol
China. Neurology 39: 660664.
Sci 17: 286291.
Tanner CM, Ottman R, Goldman SM et al. (1999). Parkinson
disease in twins: an etiologic study. JAMA 281: 341346.
Thiruchelvam M, Richfield EK, Baggs RB et al. (2000). The
Further Reading
nigrostriatal dopaminergic system as a preferential target
of repeated exposures to combined paraquat and maneb: Di Monte DA (2003). The environment and Parkinsons dis-
implications for Parkinsons disease. J Neurosci 20: ease: is the nigrostriatal system preferentially targeted by
92079214. neurotoxins? Lancet Neurol 2: 531538.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 51

Drug-induced parkinsonism

FEDERICO EDUARDO MICHELI* AND MARIA GRACIELA CERSOSIMO

University of Buenos Aires, Buenos Aires, Argentina

51.1. Introduction Interestingly enough, despite the wide interest


neurologists have shown in this subject in the last
Parkinsonism encompasses a heterogeneous group of 15 years, the cause of these movement disorders is
movement disorders, featuring resting tremor, rigidity, apt to be overlooked, unless the physician has a good
bradykinesia and postural instability (Quinn, 1995), understanding of the possible offending medications.
which have a significant impact on an individuals Therefore, the risks and benefits of drug therapy need
quality of life (Marinus et al., 2002). Whereas primary to be known and carefully considered by physicians.
parkinsonism includes progressive, neurodegenerative Patients and their families should be duly informed
disorders, Parkinsons disease (PD) being the most about them.
common, it is known that secondary parkinsonism can
be induced by drugs or caused by toxins or metabolic 51.2. History
disorders.
Drug-induced movement disorders (DIMD), most The modern history of DIMD starts in the early 1950s
of them induced by antidopaminergic drugs, are some when chlorpromazine was developed in France and
of the most frequent causes of secondary movement acknowledged as a breakthrough in the treatment of
disorders, often causing severe psychosocial distur- psychosis (Delay et al., 1952; Delay and Deniker,
bance in both non-psychotic patients and patients with 1956). However, initial enthusiasm was soon tempered
schizophrenia (Caligiuri et al., 2000). In addition, by its side-effects, including acute reactions such as
DIMD are a major cause of poor compliance with akathisia, dystonia and drug-induced parkinsonism
treatment, which in turn is associated with relapses, (DIP), which were soon documented (Steck, 1954;
hospitalization and morbidity (Casey, 1991). Ayd, 1961).
DIMD encompasses a broad spectrum of syndromes, The psychomotor apathy syndrome was described
diverse in time of onset and in their clinical mani- after initial clinical trials with chlorpromazine in psy-
festations. Acute-onset syndromes develop in hours to chiatric patients and was thought to be specific for this
days and include acute akathisia, acute dystonic drug. Later on, this finding was related to the akinesia
reactions and neuroleptic malignant syndrome; parkin- syndrome without hypertonia which had been pre-
sonism develops over weeks and the tardive syndromes viously described as the sequelae of encephalitis
typically develop after months or even years of expo- lethargica by Lhermitte in 1923. At that time reser-
sure to neuroleptic or other antidopaminergic drugs. pine, a dopamine-depleting drug, known to induce an
Although some types of DIMD can be successfully akinetic state in animals (Carlsson, 1959) was noted
treated, at present, no consistently effective treatment is to cause a parkinsonian state in humans. This observa-
available for others which may become irreversible even tions, coupled with the known histopathology of PD,
if the causative drug is discontinued, highlighting that led to the discovery that dopamine is severely depleted
certain drugs are apt to cause persisting, if not permanent, in PD (Ehringer and Hornykiewicz, 1960).
central nervous system (CNS) changes. Thus, attention In 1954 Steck reported parkinsonism in patients trea-
should focus on close monitoring and prevention. ted with chlorpromazine and reserpine. He highlighted

*Correspondence to: Professor Federico Micheli, University of Buenos Aires, Hospital de Clnicas Jose de San Martn, Buenos
Aires, Argentina. E-mail: fmicheli@fibertel.com.ar
400 F. E. MICHELI AND M. G. CERSOSIMO
that these manifestations were reversible and that they often pose a dilemma as to the cause of their
rather resembled postencephalitic parkinsonism, but movement disorder and drug therapy must always be
not PD. He also reported akathisia, a common feature considered in the differential diagnosis. Which particu-
in postencephalitic parkinsonism but not in PD, in his lar patient with parkinsonism will turn to be a case of
cases. DIP or even PD aggravated or unmasked by drugs?
More persistent or even permanent dyskinesias Clinicians need to answer these questions as accurately
were first recognized in the late 1960s (Uhnbrand as possible since a mistaken diagnosis may lead to
and Faurbye, 1969). unnecessary therapies, often associated with unwanted
Currently, it is widely acknowledged that all dopa- effects and an emotional and economic burden for
mine antagonists as well as other drugs that interfere patients and their families. In brief, the wide range
with the synthesis or release of dopamine have the and increasing list of drugs implicated in the produc-
potential to produce DIP in predisposed individuals. tion of DIP pose a challenging task for the clinician
As DIP was initially recognized as secondary to (Friedman, 1989).
neuroleptics (from the Greek: which grip the nerves),
it almost became synonymous with neuroleptic- 51.3. Prevalence
induced parkinsonism. The use of antipsychotic drugs
in the treatment of psychosis led to the common occur- DIP has been claimed to represent one of the most
rence of DIP in this population (Freyhan, 1959), which frequent causes of secondary parkinsonism in clinical
was most likely underestimated in clinical practice practice (Teive et al., 2004), if not the most frequent
(Marti Masso et al., 1993). cause (Nguyen et al., 2004) in several countries, to
For some time, it was suggested that appropriate the point that many authors have suggested that DIP
control of psychosis could only be accomplished when should always be suspected when parkinsonian symp-
DIP occurred, but this has been clearly shown to be toms rapidly develop or worsen in a patient treated
false, leaving DIP as an unwanted adverse effect of with certain medications, particularly those with
neuroleptic drugs. The new class of antipsychotics, antidopaminergic properties.
denominated atypical neuroleptics, is at least as The estimates for DIP differ widely among differ-
effective as the older antipsychotics but is much less ent series. Whereas PD is likely to account for up to
likely to cause movement disorders. 60% of patients with parkinsonism, the second leading
Among the various types of DIMD secondary to cause is DIP, detected in 20% of patients (Cardoso,
neuroleptics, DIP is one of the most frequent but is 1995). However, regional estimates are extremely
often poorly recognized, and is frequently indistin- variable.
guishable from idiopathic PD which purportedly results A study of a population of 208 000 in a district
from the blockade of striatal dopamine receptors. located in the English Midlands showed a PD preva-
It is now well known that drugs, with a wide range lence of 108.4 per 100 000, which is roughly compar-
of applications in medicine, beyond the treatment of able with figures from Carlisle, England; Rochester,
psychiatric illnesses and of diverse chemical nature, Minnesota; and south-west Finland. In this population,
may induce or exacerbate parkinsonism. In addition DIP was very uncommon (Aronson, 1985).
to neuroleptics, selective serotonin reuptake inhibitors Conversely, a door-to-door, two-phase approach
(SSRIs), lithium, valproic acid, calcium channel block- study of parkinsonism prevalence in three elderly (over
ers, antiarrhythmics, cholinergics, chemotherapeutics, 65 years of age) population groups of central Spain
amphotericin B, estrogens and others have been impli- showed that, out of 118 subjects with parkinsonism,
cated and it seems that an ever-growing list will add 81 had PD (68.6%), 26 DIP (22.0%), 6 parkinsonism/
new potential causes for DIP. dementia (5.1%), 3 vascular parkinsonism (2.5%) and
Currently, neuroleptic drugs, including substituted 2 unspecified parkinsonism (1.7%). The prevalence
benzamides and calcium channel blockers, not always was 2.2% (95% confidence interval (CI), 1.82.6) for
used as antipsychotics, are probably the drugs most all types of parkinsonism and 1.5% (95% CI, 1.21.8)
commonly involved in DIP. It is known that the elderly for PD (Benito-Leon et al., 1998).
population is at an increased risk of DIMD, including To support the above, DIP has also been considered
DIP, due to intrinsic factors and because such patients the second leading cause of parkinsonism, only after
are often treated with several drugs, including those PD, and representing 1030% of all patients with
from self-medication (Gershanik, 1994). parkinsonism in Spain (Errea-Abad et al., 1998).
Psychiatric patients treated with typical neurolepics The incidence of parkinsonism and its specific types
are well known to be at risk to develop DIP; however, was studied among residents of Olmsted County, USA,
non-psychotic outpatients developing parkinsonism between 1976 and 1990. They found 364 incident cases
DRUG-INDUCED PARKINSONISM 401
of parkinsonism: 154 with PD (42%), 72 with DIP Bradykinesia is often the initial, most frequent
(20%), 61 unspecified (17%), 51 with parkinsonism in and often the only manifestation of DIP, featuring
dementia (14%) and 26 with other causes (7%). PD hypomimia, loss of associated movements and speech
was the most common type of parkinsonism, followed disturbances.
by parkinsonism in dementia in men and DIP in women Symmetric clinical signs are felt to be the rule in
(Bower et al., 1999). PD and often a clue to a correct diagnosis. However,
Similar findings have been reported in Japan, where this may not always be the case and, as in PD, sub-
DIP is currently the second leading cause of parkin- groups seem to exist. Caligiuri et al. (1989) studied
sonism together with idiopathic PD. The ratio of the 26 treated schizophrenic patients and 14 normal con-
incidence of DIP to PD has been reported to be trols for the presence and asymmetry of neuroleptic-
between 1:2 and 1:5 (Kuzuhara, 1997). induced rigidity, to evaluate the sensitivity, reliability
In Italy the figures for DIP seem to be lower. A and validity of a quantitative procedure, showing that
population-based survey performed in eight Italian 65% of the patients exhibited pathological rigidity.
municipalities detected 42 PD (62%), 8 parkinson- Moreover, 76% of these patients displayed asymmetric
ism/dementia (12%), 8 vascular parkinsonism (12%), rigidity, and the remaining 24% exhibited bilateral
7 DIP (10%) and 3 cases of unclassified parkinsonism symptoms. Newly treated patients exhibited greater
(5.8%) (Baldereschi et al., 2000). rigidity on the right side as compared to patients who
had been consistently treated for at least 3 months.
51.4. Symptoms They speculated that striatal dopaminergic activity
may be asymmetric and more marked in the right stria-
The differential diagnosis from PD on clinical grounds tum among recently treated patients (Caligiuri et al.,
is often troublesome as the clinical features of DIP 1989). Even though this is a drug-induced syndrome,
mimic those of PD, except for the rather rapid progres- signs may be asymmetric in half of the patients, as
sion of symptoms in the latter (Kuzuhara, 1997). illustrated by findings in a small number of patients
Bradykinesia, rigidity, impaired postural reflexes (Hardie and Lees, 1988; Sethi and Zamrini, 1990). In
and tremor may occur. addition PD may present a symmetrical onset in 4%
However, some subtle differences may be of cases (Rajput et al., 1993).
observed (Table 51.1): for example, DIP is often The complete triad of tremor, rigidity and bradyki-
associated with tardive dyskinesias (TDKs) and invol- nesia is only seen in a limited number of cases, as illu-
vement may be symmetrical. Rigidity and impaired strated by the 25% observed in the series from Llau
arm-swing are the most common early signs, with tre- et al. (1994).
mor marking the onset in only about one-third of Gait abnormalities are frequent manifestations of
cases (Ayd, 1961). The latter is often not relevant DIP (Akbostanci et al., 1999) but a very uncommon
and may even be absent (Akbostanci et al., 1999). initial manifestation of PD. Interestingly enough, freez-
This could be due to either the younger age of most ing is frequently observed in patients with vascular
patients with DIP or to differences between the two parkinsonism (57%), normal-pressure hydrocephalus
conditions. (56%) and generally in patients with neurodegenerative
Table 51.1
Features that characterize drug-induced parkinsonism and Parkinsons disease

Drug-induced parkinsonism Parkinsons disease

Symptoms at onset Bilateral and symmetric or asymmetric Unilateral or asymmetric


Onset Acute or subacute Chronic
Course with appropriate treatment Regressive Progressive
Response to anticholinergic drugs Evident Mild to moderate
Response to levodopa Poor Marked
Akathisia Present Absent
Other associated features Orobuccal dyskinesia and other choreic
manifestations, rabbit syndrome
Incidence of rest tremor < >
Gender > Females > Males
Freezing Uncommon Common
Seborrheic dermatitis > <
402 F. E. MICHELI AND M. G. CERSOSIMO
parkinsonism, including progressive supranuclear sive need to move and objective motor manifestations
palsy, multiple system atrophy and corticobasal gang- such as shifting of the body weight from foot to foot
lionic degeneration (45%). However, freezing seems while standing or crossing and uncrossing the legs
to be rare in patients with DIP who have a very low when sitting. It is a typical side-effect of neuroleptics
risk for developing freezing (P < 0.00001; OR, 0.1) and often coexists with DIP (Kim and Byun, 2003).
compared to patients with other types of parkinsonism Akathisia is extremely uncommon in PD, although
(Giladi et al., 1997). it has been documented in parkinsonian patients trea-
A significant number of patients treated with ted with neuroleptics (Prueter et al., 2003).
phenothiazine or butyrophenone neuroleptic drugs with Rabbit syndrome features a rapid orofacial tremor,
no previous movement disorders develop abnormally frequently occurring as a long-term side-effect of
low tremor frequencies. As low-frequency tremors are neuroleptic treatment. However, response to anticholi-
often associated with PD, it has been speculated that nergic medication and the frequent association with
they probably represent early signs of DIP (Arblaster DIP suggest that the underlying mechanism of rabbit
et al., 1993). syndrome is similar to that of acute forms of DIP
An increased prevalence of seborrheic dermatitis is (Wada and Yamaguchi, 1992).
a well-known feature of idiopathic PD and postence- Tardive tremor is defined according to Jankovic
phalitic parkinsonism. Evaluation of 42 hospitalized (1995) as a high-amplitude, 48 Hz rest and postural
patients with DIP and 47 hospitalized psychiatric tremor developing after prolonged exposure to a drug.
patients with movement disorders showed a statisti- Tardive tremor has been associated with neuroleptics
cally significant higher prevalence of clinically diag- (Stacy and Jankovic, 1992) and other drugs, including
nosed seborrheic dermatitis in the DIP group (59.5% calcium channel blockers (Garcia-Ruiz et al., 1992).
versus 15%) (Binder and Jonelis, 1983). Tardive dyskinesia (TDK) comprises hyperkinetic
As in PD, dysphagia may be a prominent and even involuntary movements, varying in localization and
an early sign of DIP. A 74-year-old woman, without appearance. The most common presentation involves
prior symptoms of parkinsonism or dysphagia, pre- the oral and facial regions and is called bucco-linguo-
sented the association of both after the administration masticatory syndrome; however, in many cases finger
of trifluoperazine hydrochloride (Bashford and Bradd, arm or toeleg movements are evident. Thoracic
1996). Another patient complained of difficulty chew- movements (respiratory dyskinesias) and pelvic invol-
ing and swallowing as the initial manifestation of DIP. vement are also frequent. TD and DIP have been
The evaluation of dysphagia demonstrated abnormal- hypothesized to reflect opposing states of dopamine
ities during all stages of ingestion. However, the function as DIP TD occurs more frequently in the
prepharyngeal stages were disproportionately affected elderly population (Crane and Smeets, 1974).
when compared with those patients with PD and Tardive dystonia (TD) features sustained involun-
similar levels of parkinsonian functional disability tary contractions of antagonist muscles causing slow
(Leopold, 1996). Dysphagia is a potentially life- twisting movements or sustained postures. Dystonia
threatening complication of DIP. Its early recognition is more often localized in the oral region, causing oro-
is of paramount importance as it enables treatment by mandibular involvement with either forced jaw open-
simple medical, physical and dietary manipulations. ing and tongue protrusion or diurnal bruxism
(Micheli et al., 1993); in the facial areas, producing
51.5. Associated features blepharospasm; in the neck, with several types of cer-
vical dystonia; and in the trunk, among others.
DIP can be associated with other movement disorders
which are known to be typically induced by drugs. 51.6. Course
Quite often such movements require therapeutic
approaches which are different from, if not opposed DIP usually develops gradually within the first 3
to, those needed for the treatment of parkinsonism. months after the patient is exposed to the causative
Nevertheless, the presence of such movements is a drug (Friedman, 1989), although it may take several
clue to the correct diagnosis of DIP. Although some months for the full-blown picture to manifest.
of them are acute (akathisia), tardive movement On rare occasions DIP can be caused by neuroleptic
disorders appear after long-term treatment with the withdrawal and has been labeled as withdrawal
causative drug and moreover treatment poses a real emergent parkinsonism (Inoue and Janikowski,
challenge. 1981), but no adequate explanation for this phenom-
Akathisia is a complex syndrome featuring a enon has been provided. A possible clue, in some
subjective sense of inner restlessness with a compul- cases treated concomitantly with neuroleptics and
DRUG-INDUCED PARKINSONISM 403
anticholinergics, where both drugs are discontinued at should immediately alert the physician to consider DIP
the same time, could be the different half-lives of these as the first possible diagnosis. However, in non-
drugs as well as the dissimilar duration of their action psychotic patients, the awareness of the clinician of
on the CNS. the harmful effects of the offending drugs is often insuf-
It is commonly acknowledged that DIP is usually a ficient to lead to a correct diagnosis, which is then
benign condition that resolves rapidly after the causative unfortunately delayed. Frequently DIMD, including
drug is withdrawn. Usually parkinsonian symptoms begin DIP, are misdiagnosed (Weiden et al., 1987).
to improve in several weeks and patients are relieved Symptoms may be misleading as illustrated by
from the symptoms usually within several months (Kuzu- Hansen et al. (1992), who reported on the occurrence
hara, 1997), although complete resolution may take over of TDK and DIP in 101 inpatients, documenting mis-
a year (Marti-Masso and Poza, 1996, 1998). recognition of both disorders by resident physicians,
Occasional cases with extremely unusual duration of that determined a DIP prevalence of 11% versus a
symptoms after discontinuance of the offending 26% rate found by the researchers. Residents tended
drug have been described (Klawans et al., 1973). to miss DIP in younger patients and in patients with
Physicians should be acquainted with the potential long affective disorders in whom a differential diagnosis
duration of DIP in order to avoid diagnostic errors. with DIP may be troublesome.
Despite the lack of clear-cut evidence, it is often Although radiological procedures, including posi-
acknowledged that tolerance develops to DIP. This tron emission tomography (PET) scans, have proven
assumption is supported by the observation that abnormal in PD, they are not commercially avail-
withdrawal of anticholinergics co-administered with able and the diagnosis is currently based on clinical
neuroleptics is followed by rare cases of DIP. evaluation. Functional neuroimaging using (123I)
Conversely, Kennedy et al. (1971) reported that 27 out beta-carboxymethyoxy-3-beta-(4-iodophenyl) tropane
of 63 patients treated with trifluoperazine for more than (CIT) and single-photon emission computed tomogra-
3 years continued to have tremor and 24 had rigidity. phy (SPECT) provide information on the integrity of
However, a small number of patients may have the dopaminergic system in vivo and are promising
persistent parkinsonism. Whether this is an aging- diagnostic tools in early PD, and particularly in the
related effect remains unclear (Jeste et al., 1998). differential diagnosis between parkinsonism and
DIP is reversible, with the possible exception of a non-organic causes of similar symptoms.
small percentage of elderly patients. Melamed et al. Performing (123I) beta-CIT and SPECT imaging at
(1991) described 2 young patients who developed DIP baseline appears to be a useful diagnostic approach to
during chronic treatment with neuroleptics for a psycho- detect patients thought to have parkinsonism at baseline
tic disorder. Parkinsonism not only persisted but even but who, after follow-up, do not have parkinsonism (Jen-
progressed after discontinuation of the antipsychotic nings et al., 2004). These findings may be particularly
drugs. One patient experienced tremor-predominant useful in the differential diagnosis of negative symptoms
parkinsonism whereas the other presented an akinetic- of psychotic patients mimicking parkinsonism.
rigid syndrome. Neither had TDK and they both bene- Other symptoms present in PD, such as depression,
fited from levodopa therapy. Such persistent DIP cases asthenia, sense of weakness, sedation and effects
have been interpreted as pre-existent subclinical PD of understimulation, as have been reported in pati-
cases, uncovered by drug therapy. In theory, these drugs ents institutionalized for long periods of time
may precipitate the degeneration of vulnerable, nigros- (Fleischhacker, 2000), may overlap with the underly-
triatal neurons by generating cytotoxic free radicals or ing psychiatric illness. There may also be drug-related
by attrition, due to accelerated neuronal firing rates. side-effects that are unrelated to the parkinsonism.
Total disappearance of the clinical signs occurred The striking similarity between the pattern of visual
in 74% of the patients, whereas, in 15% of cases, contrast sensitivity loss in DIP and that in PD suggests that
DIP led to the diagnosis of underlying idiopathic a widespread dopaminergic deficiency, regardless of the
PD in the series of Llau et al. (1994). This study cause, may similarly affect vision (Bulens et al., 1989).
showed that almost 80% of DIP cases are due to
two pharmacological classes of agents: antidopami- 51.8. Pathology
nergic agents and calcium channel blockers.
Postmortem reports of DIP cases are limited and the
51.7. Diagnosis results are at odds. Bathia et al. (1993) reported a
patient with cranial dystonia who also had rest tremor
In the setting of a psychiatric patient receiving neuro- in one arm and who developed DIP while under
leptic drugs, the appearance of parkinsonian symptoms treatment. The patients brain was normal on autopsy.
404 F. E. MICHELI AND M. G. CERSOSIMO
Brown et al. (1996) examined caudate nuclei areas in ethanethiolato(3-)-N2,N20 ,S2,S20 ]oxo-[1R-(exo-exo)],
10 schizophrenic subjects with DIP and 25 schizophrenic TRODAT) may give a measure of presynaptic neuronal
subjects without parkinsonian symptoms. The subjects degeneration. Striatal uptake has been shown to correlate
with parkinsonian symptoms were found to have statisti- with disease severity, in particular with bradykinesia and
cally significant smaller right caudate nuclei and a trend rigidity, and could be useful to monitor disease progres-
towards smaller left caudates. Cortical measurements sion in PD. Dopamine deficiency largely antedates clini-
did not differ between the groups. The authors conclude cal manifestations and is always present, even in the
that the results support the contention that antipsychotic earliest clinical presentations of PD; a normal scan
drugs exert some CNS neurotoxic effects. suggests an alternative diagnosis such as DIP or essential
Rajput et al. (1982) reported 2 patients who developed tremor, vascular parkinsonism (unless there is focal
parkinsonism after treatment with neuroleptic drugs. basal ganglia infarction) or psychogenic parkinsonism.
Clinical manifestations remitted completely when the Additional applications of this method include character-
offending drugs were discontinued. Surprisingly, when izing dementia with parkinsonian features (abnormal
these cases came to autopsy, histological examination results in dementia with Lewy bodies, common in
in each patient disclosed abnormalities characteristic of Alzheimers disease); and differentiating juvenile-onset
PD. Levels of homovanillic acid were low in both cases PD (abnormal DAT) from dopa-responsive dystonia
and dopamine levels were reduced in the striatum in 1 (normal DAT) (Marshall and Grosset, 2003).
case. It is postulated that these 2 patients had subclinical Recently it has been shown that striatal morphology
PD that was unmasked by neuroleptics. on a three-dimensional display of 123I-beta-CIT SPECT
Bower et al. (2002) correlated the clinical features data provides information that is of diagnostic signifi-
with pathological findings in an autopsy series of cases cance for PD. This morphometry can be done without
of parkinsonism in Olmsted County, MN, for the years technically demanding region of interest analysis and
19761990. They came across 364 incidental cases of thus this technique may be suitable for routine clinical
parkinsonism, of which 235 were deceased at the time use to study the integrity of the dopaminergic nigrostria-
of the study; 39 autopsied brains were available for tal system (Ichise et al., 1999). Moreover, it has been
analysis (17% of deceased cases). Of the 8 cases given reported to characterize the different types of parkinson-
the clinical diagnosis of DIP, 6 were found to have ism (Lorenzo Bosquet et al., 2004).
basal ganglia pathology. Negative symptoms in psychotic patients have been
correlated with parkinsonian symptoms, some vegeta-
51.9. Differential diagnosis tive features of depression and with anticholinergic
doses, but not with negative symptoms and cognitive
A comprehensive drug history is essential when features of depression or neuroleptic drugs. These
evaluating a patient with parkinsonian signs. findings suggest that the assessment criteria for nega-
Very few disorders can mimic DIP, and when the tive symptoms, depression and DIP overlap in treated
features of parkinsonism and a drug history are clear, schizophrenic patients (Prosser et al., 1987).
the diagnosis of DIP is straightforward and the most A retrospective evaluation of the premorbid status of
troublesome question that arises is whether the patient patients within their first episode of psychosis showed
has DIP only or subclinical PD uncovered by drug that lower sociability and withdrawal scores correlated
exposure. Answering this question is of paramount with increased time to treatment response, more severe
importance to both patients and doctors as the negative symptoms, deterioration of premorbid function-
prognosis of these disorders is quite different. ing and increased DIP. These findings suggest that, prior
Unfortunately, a definitive diagnosis is often delayed; to acute psychosis onset, certain behavioral precursors
meanwhile the patient is under careful observation. reflecting premorbid functioning may predict subsequent
Functional imaging of the dopamine transporter illness manifestations (Strous et al., 2004).
(DAT) is useful to check the integrity of the dop- Psychogenic parkinsonism is extremely rare in
aminergic system and is apt to be used in patients clinical practice. Tolosa et al. (2003) reviewed the
with mild, incomplete or uncertain parkinsonism. clinical features and results of DAT imaging in DIP
Imaging with various specific SPECT ligands for DAT and psychogenic parkinsonism. These two conditions
(N-omega-fluoropropyl-2-beta-carbomethoxy-3 beta- normally give normal striatal DAT imaging results;
(4-iodophenyl)nortropane, FP-CIT; 2-beta-carbometh- an abnormal result in either case could rule out both
oxy-3 beta-(4-iodophenyl)tropane [123I], beta-CIT; conditions, corroborating a diagnosis of organic
N-((E)-3-iodopropen-2-yl)-2 beta-carbomethoxy-3 parkinsonism in uncertain cases.
beta-(4-chlorophenyl)tropane [123I], IPT; [2-[[2- Severe DIP cases should also be differentiated from
[[[3-(4-chlorophenyl)-8-methyl-8-azabicyclo[3,2,1] catatonia, a probably underdiagnosed disorder (Van
oct-2-yl]methyl](2-mercaptoethyl)amino]ethyl]amino] der Heijden et al., 2005) in which tremor is absent.
DRUG-INDUCED PARKINSONISM 405
Unfortunately the two conditions may coexist as some become rigid. Patients with familial essential tremor
patients with catatonia can be under treatment with have normal, whereas those with isolated rest tremor
neuroleptics. Assessment of the patients mental state have consistently low, putamen 18F-dopa uptake. DIP
can make the difference clear. Symptoms like waxy is infrequently associated with underlying nigral
flexibility and muteness are clues for the diagnosis of pathology (Brooks, 1991).
catatonia. As lorazepam and electroconvulsive therapy
(ECT) are effective in treating catatonia, an early 51.12. Transcranial ultrasound (TCS)
diagnosis is required (Huang, 2005).
Among the various causes of parkinsonism other than Applying TCS, Berg et al. (1999) found that approxi-
PD that must be considered in the differential diagnosis mately 9% of healthy individuals exhibit an increased
of DIP, those degenerative, toxic, tumors or hydrocepha- substantia nigra (SN) echogenicity, a sonographic
lus that combine mental changes, often requiring drug feature that is often found in PD (Becker et al.,
therapy and movement disorders, are on the first line. 1995). PET studies in 10 of these otherwise healthy
In young patients, both Wilsons and Huntingtons subjects with hyperechogenicity of the SN showed
disease cause parkinsonism and psychosis. Wilsons dis- reduced [18F]-dopa uptake of the striatum, suggesting
ease typically causes a prominent resting and postural that elevated echogenicity of the SN in healthy adults
tremor as well as dysarthria. The peculiar dystonic facies can be associated with subclinical dysfunction of the
of Wilsons disease are strongly suggestive of this disor- dopaminergic nigrostriatal system (Berg et al., 1999).
der. Huntingtons disease usually causes chorea in adults As these patients could be at increased risk of developing
and parkinsonism in children, but a parkinsonian (West- DIP, Berg et al. (2001) compared the echo pattern of
phal) variant can occur in young adults. In this case a patients on neuroleptics who developed DIP with those
positive family history for chorea is a rule. who did not show evidence of parkinsonian symptoms.
Parkinsonism can also be a feature of normal- In a second study they compared the echo pattern with
pressure hydrocephalus, often associated with a gait the severity of the parkinsonian signs. Findings demon-
disorder that is rather atypical for DIP, urinary inconti- strated that patients with more extended hyperechogenic
nence and cognitive imapirment. Brain computed tomo- areas at the SN were more susceptible to DIP, which was
graphy (CT) scans or magnetic resonance imaging more severe contralateral to the side of the more echo-
(MRI) scans are useful when considering this diagnosis. genic SN (Berg et al., 2001).
In the last few years brain imaging has emerged
as a potentially useful tool to study the activity of 51.13. Risk factors
the basal ganglia as well as help in the diagnosis
of different types of movement disorder. Unfor- Studies addressing the identification of risk factors for
tunately they are not yet widely available in clinical the development of DIP highlighted the noteworthy indi-
practice. vidual susceptibility to develop DIP. It has been accepted
that both the drug potency and dose play a role in the
51.10. Magnetic resonance spectroscopy development of DIP, but this may not always be the case.
Although not all risk factors have been universally
MR spectroscopy has been described as a useful tool accepted and some have even been challenged, we list
to monitor the neurobiological correlates of DIP and here the most frequently cited.
other DIMD. The severity of DIP assessed by the
SimpsonAngus Scale significantly correlates with 51.13.1. Age/dementia
the higher choline (Cho) concentration and moreover
tends to correlate with the higher N-acetyl-aspartate Elderly patients and particularly those with associated
(NAA) concentration in a group of patients (Yamasue dementia are at greater risk than patients without
et al., 2003). dementia for persistent drug-induced extrapyramidal
side-effects (Marti Masso and Poza, 1996; Caligiuri
51.11. Positron emission tomography et al., 2000).
Moghal et al. (1995) reported the presence of move-
Putamen 18F-dopa uptake of PD patients is reduced by ment disorders in an institutionalized elderly population
at least 35% at onset of symptoms; therefore, PET of Saskatchewan. Movement disorder was detected in
scans can be used to detect preclinical disease in clini- 19% of cases. Most of these cases (16%) were female;
cally unaffected twins and relatives of patients with 10% had essential tremor, 6% PD and 2.3% had DIP.
PD. PET scans can be used to detect underlying nigral In a community incidence study, 7.2% had DIP (Rajput
pathology in patients with isolated tremor and patients et al., 1984) and a prevalence study reported 8.8% cases
taking dopamine receptor-blocking agents who of parkinsonism being DIP (Bharucha et al,. 1988).
406 F. E. MICHELI AND M. G. CERSOSIMO
To examine the natural history and pathogenesis of cause DIP. Several authors have shown that findings
parkinsonism in Alzheimers disease, 44 subjects with suggest that DIP is associated with an increased risk
clearly established senile dementia of the Alzheimer for PD (Chabolla et al., 1998).
type were studied for 66 months. Sixteen subjects
(36%) developed idiopathic parkinsonism and 12 sub- 51.13.3. Cigarette smoking
jects (27%) developed DIP; the chief clinical features
of both types were bradykinesia and rigidity, but not Several studies have shown that cigarette smoking
resting tremor. The presence of parkinsonism was may have a protective effect as regards the risk for
associated with global (rather than selective) cognitive PD (Allam et al., 2004).
impairment, as determined by psychometric testing Similarly, according to one study, schizophrenic
and with a more rapid progression to advanced stages patients who smoke have significantly less cognitive
of dementia (Morris et al., 1989). impairment (P < 0.02) and a lower prevalence of
The association between parkinsonism and Alzhei- DIP as compared to non-smokers (Sandyk, 1993).
mers disease has received great attention in recent Based on these findings, the author suggests that cigar-
years. It has been speculated that the occurrence of par- ette smoking may protect against the development of
kinsonism and Alzheimers disease is not a chance dementia and DIP in schizophrenia.
association.
The pathological correlates of parkinsonism were 51.13.4. Genetics
found to be heterogeneous in the postmortem examina-
tion of subjects with Alzheimers disease. Some of The results of epidemiological studies indicate that
them exhibited pathological features of PD, whereas there is a hereditary predisposition to developing
others had non-specific nigral lesions; and even in some DIP. Metzer et al. (1989) investigated the human
cases neither histological changes nor reduced neuronal leukocyte antigen (HLA) antigen prevalence rates in
densities in the SN were observed (Morris et al., 1989). patients with neuroleptic DIP. They evaluated
The incidence and prevalence of DIP and TD are neuroleptic-treated, chronic inpatients with Diagnostic
significantly greater among elderly patients, whereas and Statistical Manual of Mental Disorders III
akathisia seems to occur evenly across the age range (DSM-III)-diagnosed schizophrenia, 29 with DIP and
and dystonia is more common among younger patients 23 without parkinsonian signs. They were tested for 23
(Errea-Abad et al., 1998; Caligiuri et al., 2000). type A, 43 type B, 4 type C and 10 type DR
Neuroleptic use is a common cause of DIP among HLA antigens and it was found that, in DIP patients,
the elderly and this side-effect is frequently treated HLA-B44 was significantly more prevalent, suggesting
by adding an anticholinergic or dopaminergic drug to that this HLA antigen could play a role in genetic or
the regimen. The use of anticholinergic drugs presents immunologic susceptibility to develop DIP in some
risks of additional drug side-effects; the use of dopa- patients.
minergic drugs, generally not appropriate for DIP, A higher occurrence of a positive family history of
suggests that extrapyramidal neuroleptic side-effects PD and/or essential tremor and of higher frequency
may often be mistaken for idiopathic PD in older of secondary cases with PD and/or essential tremor
patients (Avorn et al., 1995). among close relatives of patients with parkinsonism
induced by cinnarizine and flunarizine as compared
51.13.2. Pre-existing movement disorders to age-matched controls suggests the involvement
of a genetic susceptibility in developing this drug-
There is general agreement that pre-existing extrapyr- induced disorder. DIP could be regarded as a
amidal signs increase the patients vulnerability to multifactorial disease process resulting from potential
develop significant DIMD (Caligiuri et al., 2000). To neurotoxicity of drugs on a background of inherited
support this, previous studies suggest that many predisposition (Negrotti et al., 1992).
untreated schizophrenic patients exhibit motor distur-
bances. On the basis of these findings, the authors 51.13.5. Gender
hypothesized that pre-existing extrapyramidal move-
ment disorders may increase the risk of developing The fact that women are at increased risk of develop-
DIP. ing DIP has been consistently reported by many inves-
A number of studies have shown that in a variable tigators (Klawans et al., 1973; Llau et al., 1994; Marti
number of cases DIP is only the expression of subcli- Masso and Poza, 1996; Errea-Abad et al., 1998).
nical PD unmasked by the action of neuroleptics, In this regard a retrospective study carried out by Llau
calcium channel blockers or other drugs known to et al. (1994) investigated the characteristics of DIP
DRUG-INDUCED PARKINSONISM 407
notified to the Midi-Pyrenees Pharmacovigilance Centre, had an enhancing lesion in the midbrain at the T1-
France between 1983 and 1992 found that, among 3923 weighted MRI, which correlated with a homolateral
side-effects spontaneously reported between 1983 and ophthalmoplegia and contralateral parkinsonism.
1992 to the center, 53 (1.4%) were DIP cases. Mean The authors concluded that parkinsonism was probably
age was 65  2 (sem) years (range 2188 years). DIP secondary to HIV in 12, to metoclopramide in 1 and to
appeared after a mean treatment duration of 473  142 neurotoxoplasmosis in another 1, and that a diagnosis
days (range 1 day to 15 years) and occurred most fre- of HIV should always be considered in young patients
quently in women (63%) (Llau et al., 1994). Estrogen- with secondary parkinsonism:
related dopamine receptor blockade has been suggested By using stereological techniques, Reyes et al.
as a possible explanation for female preponderance (1991) estimated the volume density of melanin and
(Glazer et al., 1983). However, others have not found a counted the number of pigmented and non-pigmented
gender preponderance (Moleman et al., 1982). neuronal cell bodies in the pars compacta of the SN
of autopsied AIDS cases without any evidence of PD
51.13.6. Schizophrenia subtype or inflammation or necrosis of the midbrain. They
found that the total number of neuronal cell bodies
It is possible that DIP may relate to negative symptoms, was 25% lower in AIDS patients than in age-matched
whereas TDK in schizophrenia may be a covariate of controls. The histopathological examination showed
positive symptoms. Sandyk and Kay (1992) studied nigral degeneration with neuronal loss, small neuronal
the relationship of TDK and DIP with psychopathology cell bodies packed with melanin, reactive astrocytosis
clusters rated on the Positive and Negative Syndrome and extracellular melanin in the AIDS patients but not
Scale in schizophrenic patients and found that involun- in the controls, showing that a subclinical nigral degen-
tary movements of TDK were significantly associated eration is common in AIDS and could possibly explain
with the activation cluster (P < 0.01), whereas DIP the higher susceptibility of some patients to DIP.
was significantly associated with the anergia cluster
(P < 0.01). However, it has been speculated that the 51.13.9. Drugs that may cause drug-induced
positive correlation of DIP and negative symptoms parkinsonism
stems from the similarity between masked facies of
DIP and blunted affect of schizophrenics (Hoffman It is accepted that roughly half of the parkinsonism
et al., 1991). cases seen in a neurology practice are drug-induced
or aggravated, generally by psychotropic drugs
51.13.7. Ventricular/brain ratio (Marti Masso and Poza, 1996). Llau et al. (1995)
reported on the characteristics and drugs inducing
It has been shown that the magnitude of DIP is unwanted movement disorders notified to a Regional
positively correlated with ventricular/brain ratio and Drug Surveillance Centre between 1989 and 1993.
the severity of negative symptoms of schizophrenia Of a total of 4000 general side-effects, 122 were
(Hoffman et al., 1991). On the basis of these findings movement disorders, the most common of which was
it is suggested that a large lateral ventricular size DIP (40% of cases). They were mostly caused by neu-
may be associated with increased vulnerability to roleptics, followed by antiemetics and calcium channel
develop DIP (Luchins et al., 1983). blockers. In this regard flunarizine and cinnarizine are
some of the most common drugs causing DIP in the
51.13.8. Human immunodeficiency virus (HIV) countries where they are available (Teive et al., 2004).
This is probably the rule in most countries,
HIV infection is a risk factor for developing DIP and although there may be some intracountry regional
acquired immunodeficiency syndrome (AIDS) patients variations over time.
are more prone to develop this side-effect when exposed Marti Masso and Poza (1996) performed a retro-
to drugs liable to induce movement disorders. Pitagoras spective study of DIP patients seen between January
de Mattos et al. (2002) reported on their 14 years experi- 1981 and December 1993 in Spain. Of the 306 cases
ence (19861999) treating 2460 HIV-positive inpati- of parkinsonism seen, 56.8% were induced or aggra-
ents; 1053 (42.8%) presented at least one neurological vated by drugs. The drugs implicated most often were
manifestation and 28 (2.7%) had involuntary move- cinnarizine, sulpiride and flupentixol. Some patients
ments. Half of them had parkinsonism and, surprisingly, were on two of these drugs and a minority on three.
7 had enlarged ventricles, confirmed on CT scans. One The number of DIP cases seen increased after 1986
patient had a hypodense area with mass effect, sugges- and then remained stable through 1993. Between
tive of toxoplasmosis of the right basal ganglia. One 1981 and 1988, the most often implicated drug in
408 F. E. MICHELI AND M. G. CERSOSIMO
DIP was cinnarizine, though its relative impact of drugs capable of inducing DIP is growing rapidly
decreased in later years (Errea-Abad et al., 1998). This and seems to be endless (Marti Masso et al., 1993).
is most likely due to the better knowledge of the pro- Table 51.2 lists drugs known to induce or aggravate
file of side-effects of these drugs. Nevertheless, the list parkinsonian symptoms.

Table 51.2
Drugs that may induce or aggravate parkinsonism

Acetophenazine Meperidine
Alcohol withdrawal Mesoridazine
Alizapride 3,4-Methylenedioxymethamphetamine
Alpha-methyldopa Metoclopramide
Amiodarone Mexiletine
Amoxapine Molindone
Amphotericin b Naproxen
Antiacids Nefazodone
Aprindine Nifedipine
Bethanechol Olanzapine
Bupivacaine Paclitaxel
Bupropion Papaverine
Buspirone Paroxetine
Captopril Pentoxifylline
Carboplatin Perfenazine
Cephaloridine Perhexiline
Ciclosporin Perphenazine
Cimetidine Pethidine
Citalopram Phenobarbital
Cyclophosphamide Phenytoin
Cytosine arabinoside Pimozide
Chloroquine Piperazine
Chlorpromazine Pirlindol
Chlorprothixene Procainamide
Cinnarizine Procaine
Cisapride Prochlorperazine
Clebopride Promethazine
Diazepam Propiverine
Diltiazem Pyridoxine
Disulfiram Remoxipride
Domperidone Reserpine
Droperidol Risperidone
Ecstasy Sertraline
Estrogen and oral contraceptives Sodium valproate
Ethinyl Sufentanil
Fentanyl Sulindac
Flunarizine Sulpiride
Fluoxetine Tacrina
Flupentixol Tetrabenazine
Fluphenazine Thiethylperazine
Flurbiprofen Tiapride
Gemcitabine Trifluoperazine
Gold Triflupromazine
Haloperidol Trimeprazine
Halothane Thioridazine
Isoflurane Thiothixene
Levopromazine Triperidol
Lithium Veralipride
Lorazepam Verapamil
Loxapine Ziprasidone
DRUG-INDUCED PARKINSONISM 409
51.13.10. Neuroleptics Consistent with the suggested oxidative stress activity
of haloperidol, chronic treatment with the drug is accom-
Neuroleptics are at present the most effective treatment panied by the formation of a neurotoxic pyridinium meta-
for schizophrenia (Lyne et al., 2004), but non- bolite (high-performance planar chromatography
compliance is a major problem, contributing to the HPPC), a tetra hydropyridine analog resembling those
discontinuation of therapy and leading to relapses. of the pyridinium metabolite 1-methyl-4-phenyl-
Though several factors play a role in this regard, one 1,2,3,6-tetrahydropyridine (MPTP), which induces par-
of the most common side-effects is the development kinsonian symptoms (Bloomquist et al., 1994; Rollema
of movement disorders induced by these drugs (Borison et al., 1994). Gil-ad et al. (2001) studied the neurotoxic
et al., 1983). These unwanted effects led to the develop- activity of different neuroleptics (typical and atypical)
ment of a new generation of neuroleptics devoid of in order to correlate these effects with DIMD. With the
extrapyramidal side-effects, also known as atypical exception of clozapine, a correlation was observed
neuroleptics, the prototype of which is clozapine (Awad between the drug-induced neurotoxicity and its known
and Voruganti, 2004). associated movement disorders, suggesting a possible
The majority of prescriptions for antipsychotics in functional relationship between the two phenomena.
developed countries in the past years have been for Neurotoxic activity of neuroleptics was found in both
atypical drugs and the tendency shows a progressive the NB cell line and the primary mouse embryo-selected
increment. However, although most atypical neurolep- neuronal culture. Their results suggest that neuroleptic-
tics rarely induce movement disorders in young induced neurotoxicity is independent of dopamine. Pre-
patients, most are liable to cause them in susceptible vious studies (Chiueh et al., 1993; Offen et al., 1995;
populations, including elderly patients or those with Ziv et al., 1995) have shown that dopamine alone pos-
previous movement disorders, including subclinical sesses a neurotoxic and apoptotic activity at high concen-
PD. In addition, most cause akathisia (Kurz et al., 1995). trations in different neuronal cultures. This effect was
Even though investigators agree on the clinical attenuated by inhibitors of apoptosis and by some antiox-
manifestations and diagnosis of DIP, the mechanism idants (vitamin E and N-acetylcysteine), suggesting an
by which drugs induce symptoms and the identifica- oxidative stress mechanism in the drug-induced toxicity
tion of the individuals at risk are still a matter of which leads to apoptotic cell death.
conjecture. The expression of c-fos, an immediate early gene,
Since the description of the first cases, it has been correlates with neuronal activation. Thus, Fos immu-
obvious that a clear doseresponse correlation was nohistochemistry has turned out to be a valuable
absent and a discrepancy between the amount of method for mapping pathways of the CNS (Curran
neuroleptics taken and the development of parkinson- and Morgan, 1995). It has been reported that acute
ism was evident (Hall et al., 1956) suggesting that sim- administration of a typical neuroleptic, haloperidol,
ply the blockade of dopamine receptors was not the produces a large increase in the number of Fos-posi-
cause of DIP. Neuroimaging studies have suggested tive neurons in the striatum, the nucleus accumbens
that parkinsonism occurs after D2-receptor occupancy and the lateral septal nucleus in the brain of rats
exceeds 80% (Farde et al., 1992). However, the obser- (Robertson and Fibiger, 1992). By contrast, com-
vation of parkinsonism at or above 60% occupancy pounds that are not likely to cause movement disorders
levels is very consistent with prior pathological fail to increase Fos in the dorsolateral striatum or pro-
(Jellinger, 1986; Gibb, 1992) and neuroimaging duce only minor elevations (Robertson and Fibiger,
studies (Frost et al., 1993; Marek et al., 1996) of 1992). Propranolol has been reported to attenuate Fos
patients with PD that suggest symptoms become clini- expression in regions of the rat brain that are likely
cally manifest following the loss of as little as 50% of to be responsible for akathisia (Ohashi et al., 1998).
nigrostriatal neurons. In addition, plasma neuroleptic Benzamide derivatives, including sulpiride and
levels fail to correlate with DIP severity. metoclopramide, have been the main cause of DIP in
The possibility that the neurotoxic activity of recent years in Japan (Kuzuhara, 2000; Naito and
neuroleptics is related to their induced movement dis- Kuzuhara, 2004).
orders has been previously suggested. In two in vivo
studies, Ben Shachar et al. (1993, 1994) found that 51.13.11. Flunarizine and cinnarizine
haloperidol and phenotiazines induced a marked
increase in iron transport across the bloodbrain barrier Both drugs are calcium channel blockers but
in rats and mice, resulting in increased concentration of flunarizine is 2.515 times more potent. Cinnarizine
iron in basal ganglia, which in turn triggers in vivo was developed first and flunarizine is a cinnarizine
generation of hydroxyl radicals and oxidative stress. derivative and differs since it includes a piperazine
410 F. E. MICHELI AND M. G. CERSOSIMO
derivative in its molecule, like some neuroleptics and selective SSRIs. Symptoms include dystonia, akathisia
antihistamine drugs (Chouza et al., 1986; Micheli and parkinsonism (Leo, 1996; Spigset, 1999; Pina
et al., 1987, 1989; Fernandez Pardal et al., 1988; Latorre et al., 2001).
Negrotti and Calzetti, 1997; Teive et al., 2004). These Serotonin modulates dopamine in the basal ganglia
drugs have been widely employed in Spain and Latin by inhibiting its production and release. Thus, an
America in the treatment of dizziness, cerebrovascular increase in serotonergic transmission may cause parkin-
disorders and cognitive impairment. More recently, sonism, particularly in susceptible patients, including
they have also been used in the prevention of migraine those with subclinical PD and elderly patients.
attacks (Lucetti et al., 1998) and as adjunctive treatment Ectasy and related derivatives which share a basic
for epilepsy (Chaisewikul et al., 2001). Movement amphetamine structure and exert stimulatory effects
disorders secondary to these calcium antagonists were in humans have been found to have a possible antipar-
first recognized by De Melo-Souza in Brazil and kinsonian effect in animal experimental models
reported at the IX Brazilian Congress of Neurology. (Schmidt et al., 2002). Curiously, ecstasy has recently
Subsequently they were widely recognized in most of been suspected to cause DIP (OSuilleabhain and
the countries where these drugs are available (Chouza Giller, 2003). For the last 20 years 3,4-methylenediox-
et al., 1986; Micheli et al., 1987, 1989; Fernandez Par- ymethamphetamine (MDMA: also known as ecstasy)
dal et al., 1988; Marti Masso and Poza, 1996; Negrotti has become a widely used recreational drug. Metham-
and Calzetti, 1997; Teive et al., 2004). Curiously, most phetamine binds to the DAT in presynaptic terminals
of the reported cases have involved elderly women and and reverses dopamine transport (Sulzer et al., 1993).
many of them showed depression as a prominent Loss of dopaminergic neuronal terminals in human
feature. Recovery after withdrawal of flunarizine or postmortem tissue has been documented in chronic
cinnarizine usually takes several months to complete methamphetamine users and in experimental animals
(Marti Masso and Poza, 1998). treated with methamphetamine. Based on such evi-
The mechanism by which flunarizine and cinnari- dence, there is concern that loss of neuronal markers
zine cause DIP and other movement disorders is not may represent terminal degeneration, predisposing to
completely understood but it is plausible that a presy- parkinsonism as the individual becomes older. Recent
naptic as well as a postsynaptic mechanism contribute studies suggest that MDMA can cause toxicity to
to the dopaminergic dysfunction. Mena et al. (1995) dopamine-containing neurons in monkeys (OShea
investigated the effects of calcium channel antagonists and Colado, 2003).
on the pharmacology of dopamine systems in vivo and
in vitro and demonstrated that flunarizine, cinnarizine 51.14. Treatment
and diltiazem reduce the viability of dopamine-rich
human neuroblastoma cells in vitro. They concluded Particular care should be taken in treating patients who
that all calcium channel antagonists tested reduced are at risk of developing DIP or other DIMD with
dopamine neurotransmission in vitro and in vivo, but drugs that are known to cause such side-effects. Neu-
the evidence of toxicity for dopamine cells in vitro is roleptic drugs should be used according to strict cri-
restricted to flunarizine, cinnarizine and diltiazem, teria and during the period when they are necessary.
the latter only seldom associated with DIP (Dick and The benefit should be weighed against the possible
Barold, 1989). side-effects before the decision to treat a patient with
Based on animal models and laboratory studies, flu- a potential harmful drug is reached. When more than
narizine and cinnarizine have been shown to inhibit one drug is apt to benefit the patient, the less harmful
the energy-dependent vesicular dopamine uptake drug obviously has to be prescribed. When the offend-
(Terland and Flatmark, 1999), block dopamine D2- ing drug is required, as in the case of a psychotic
receptor (Brucke et al., 1995) and inhibit cathecola- patient, the antipsychotic should be replaced by an
mine transport (Vaccari, 1993) and mitochondrial atypical neuroleptic.
complexes I and II (Veitch and Hue, 1994). If prevention fails, the most effective therapy for
DIP is cessation of the offending drug. Most patients
51.13.12. Selective serotonin reuptake inhibitors will become symptom-free after 12 months off the
causative drug but occasional patients require up to a
Regardless of the general claim that SSRIs do not year or even longer for complete resolution to occur.
induce or worsen parkinsonism (DellAgnello et al., A small number of patients never recover completely
2001), in recent years several cases have been reported or partially recover and after some time exhibit signs
of movement disorders as a result of treatment with of parkinsonism again, raising the question as to
DRUG-INDUCED PARKINSONISM 411
whether they had subclinical PD that was unmasked by enhances dopamine-mediated effects and increases
the administration of the drug. The autopsy findings of gamma-aminobutyric acid concentrations in the
typical histologic features of PD in patients who had CNS. Optimal parameters relevant to the antiparkin-
DIP during life supports this notion (Rajput et al., sonism effects of ECT require further study (Faber
1982). Medical therapy is indicated when discontinu- and Trimble, 1991).
ance of the offending drug does not result in improve- Previous investigations reported in the literature
ment or if the patient requires therapy with the have found propranolol to attenuate tremor in PD,
causative drug. but no significant differences in attenuation of DIP
DIP may improve with the use of anticholinergics tremor by propranolol and placebo could be documented
(Saltz et al., 2000) and there seems to be no difference (Metzer et al., 1993).
between the efficacy of the two most popular anticho- The results of a preliminary study suggest that, in
linergics. A single-blind study compared the clinical schizophrenic patients with acute psychosis, the addi-
efficacy of biperiden hydrochloride and benzhexol in tion of vitamin E to neuroleptic medication at the initia-
the treatment of neuroleptic-induced parkinsonism. tion of treatment shows a trend towards a decrease in
Both drugs were highly effective and all patients the severity of acute DIMD. Unfortunately, it does not,
responded to medication. No significant difference however, affect the severity of acute akathisia. Vitamin
was observed between the two treatment groups when E did not interfere with the antipsychotic effect of the
individual symptoms were examined (Rosen, 1963; neuroleptic medication (Dorfman-Etrog et al., 1999).
Jensen and Amdisen, 1964; Magnus, 1980). However, The association between DIP and other movement
this therapy should be avoided in the elderly or demen- disorders, especially TD, poses a challenge as they
ted patients or those with concurrent TD. Side-effects, are responsive to pharmacological agents with antago-
including mouth dryness, constipation, blurred vision nistic properties. Unfortunately, there is no adequate
and essentially memory disturbances and delirium, answer to this question and results are often poor. It
are more prevalent in the elderly. On the other hand, has been proposed that dopamine-depleting drugs like
TD symptoms worsen on anticholinergic therapy. reserpine and tetrabenazine may be of benefit in some
Amantadine, a putative dopaminergic compound cases, improving TD without worsening parkinsonism
known to be therapeutically effective in idiopathic and (Fahn and Mayeux, 1980).
postencephalitic parkinsonism, is an alternative to The issue as to whether treatment should be used to
anticholinergic drugs (Kelly and Abuzzahab, 1971; treat symptoms of parkinsonism or used prophylacti-
Kelly et al., 1974). In a double-blind placebo-controlled cally to prevent the development of DIP has been a mat-
cross-over study of 39 psychiatric inpatients, amanta- ter of conjecture. In clinical practice many
dine and trihexyphenidyl were equally effective in psychiatrists initiate treatment with neuroleptics and
treating DIP, and amantadine produced fewer and less anticholinergics concomitantly. Currently the routine
severe side-effects. The authors suggest that amanta- use of prophylactic anticholinergics is not recom-
dine is an effective alternative to atropine-like agents, mended and is clearly contraindicated in the elderly.
with fewer implications for long-term risk of tardive An individualized riskbenefit assessment is necessary
TD (Fann and Lake, 1976). for the younger patient in whom prophylactic use of
After a few months, most cases of DIP subside and for anticholinergic drugs is considered (Mamo et al., 1999).
that reason a practical approach should include tapering
of these drugs after 23 months to evaluate spontaneous References
improvement (Horiguchi and Nishimatsu, 1992).
An alternative, and practical, approach is to use Akbostanci MC, Atbasoglu EC, Balaban H (1999). Tardive
ECT when appropriate, which transiently ameliorates dyskinesia, mild drug-induced dyskinesia, and drug-
parkinsonism and treats psychosis (Goswami et al., induced parkinsonism: risk factors and topographic
1989). ECT has been claimed to produce some benefit distribution. Acta Neurol Belg 99: 176181.
in occasional patients with DIP. A patient with schi- Allam MF, Campbell MJ, Hofman A et al. (2004). Smoking
and Parkinsons disease: systematic review of prospective
zoaffective disorder and anticholinergic refractory
studies. Mov Disord 19: 614621.
neuroleptic-induced parkinsonism manifested a
Ananth J, Samra D, Kolivakis T (1979). Amelioration of
marked increase of parkinsonian symptoms and dystonia drug-induced Parkinsonism by ECT. Am J Psychiatry
after ECT (Hanin et al., 1995). As DIP, TD and TDK 136 (8): 1094.
have also been shown to improve with ECT adminis- Arblaster LA, Lakie M, Mutch WJ et al. (1993). A study of
tration (Ananth et al., 1979), whereas tic syndromes the early signs of drug induced parkinsonism. J Neurol
have achieved mixed results. In animals, ECT Neurosurg Psychiatry 56 (3): 301303.
412 F. E. MICHELI AND M. G. CERSOSIMO

Aronson TA (1985). Persistent drug-induced parkinsonism. Bower JH, Maraganore DM, McDonnell SK et al. (1999).
Biol Psychiatry 20 (7): 795798. Incidence and distribution of parkinsonism in Olmsted
Avorn J, Bohn RL, Mogun H et al. (1995). Neuroleptic drug County, Minnesota, 19761990. Neurology 52 (6):
exposure and treatment of parkinsonism in the elderly: a 12141220.
case-control study. Am J Med 99 (1): 4854. Bower JH, Dickson DW, Taylor L et al. (2002). Clinical
Awad AG, Voruganti LN (2004). Impact of atypical antipsy- correlates of the pathology underlying parkinsonism: a
chotics on quality of life in patients with schizophrenia. population perspective. Mov Disord 17 (5): 910916.
CNS Drugs 18: 877893. Brooks DJ (1991). Detection of preclinical Parkinsons
Ayd FJ (1961). A survey of drug-induced extrapyramidal disease with PET. Geriatrics 46 (Suppl 1): 2530.
reactions. JAMA 175: 10541060. Brown KW, Wardlaw JM, White T et al. (1996). Caudate
Baldereschi M, Di Carlo A, Rocca WA et al. (2000). Parkin- nucleus area in drug-induced parkinsonism. Acta Psychiatr
sons disease and parkinsonism in a longitudinal study: Scand 94 (5): 348351.
two-fold higher incidence in men. ILSA Working Group. Brucke T, Wober C, Podreka I et al. (1995). D2 receptor
Italian Longitudinal Study on Aging. Neurology 55 (9): blockade by flunarizine and cinnarizine explains extrapyr-
13581363. amidal side effects. A SPECT study. J Cereb Blood Flow
Bashford G, Bradd P (1996). Drug-induced Parkinsonism Metab 15: 513518.
associated with dysphagia and aspiration: a brief report. Bulens C, Meerwaldt JD, van der Wildt GJ et al. (1989).
J Geriatr Psychiatry Neurol 9 (3): 133135. Visual contrast sensitivity in drug-induced Parkinsonism.
Becker G, Seufert J, Bogdahn U et al. (1995). Degeneration J Neurol Neurosurg Psychiatry 52 (3): 341345.
of substantia nigra in chronic Parkinsons disease visua- Caligiuri MP, Bracha HS, Lohr JB (1989). Asymmetry of
lized by transcranial color-coded real-time sonography. neuroleptic-induced rigidity: development of quantitative
Neurology 45: 182184. methods and clinical correlates. Psychiatry Res 30 (3):
Ben Shachar D, Livne E, Spanier I et al. (1993). Iron 275284.
modulates neuroleptics induced effects related to the Caligiuri MR, Jeste DV, Lacro JP (2000). Antipsychotic-
dopaminergic system. Isr J Med Sci 29: 587592. induced movement disorders in the elderly: epidemiology
Ben Shachar D, Livne E, Spanier I et al. (1994). Typical and and treatment recommendations. Drugs Aging 17 (5):
atypical neuroleptics induce alteration in blood brain 363384.
barrier and brain 59Fe c13 uptake. J Neurochem 62: Cardoso F (1995). Treatment of Parkinson disease. Arq Neu-
112118. ropsiquiatr 53 (1): 110.
Benito-Leon J, Bermejo-Pareja F, Rodrguez J et al. (2003). Carlsson A (1959). The occurrence, distribution and physio-
Neurological Disorders in Central Spain (NEDICES) logical role of catecholamines in the nervous system.
Study Group. Prevalence of PD and other types of parkin- Pharmacol Rev 11: 490493.
sonism in three elderly populations of central Spain. Mov Casey DE (1991). Neuroleptic drug-induced extrapyramidal
Disord 18 (3): 267274. syndromes and tardive dyskinesia. Schizophr Res 4:
109120.
Berg D, Becker G, Zeiler B et al. (1999). Vulnerability of the
Chabolla DR, Maraganore DM, Ahlskog JE et al. (1998).
nigrostriatal system as detected by transcranial ultrasound.
Drug-induced parkinsonism as a risk factor for Parkin-
Neurology 53: 10261031.
sons disease: a historical cohort study in Olmsted County,
Berg D, Jabs B, Merschdorf U et al. (2001). Echogenicity of
Minnesota. Mayo Clin Proc 73 (8): 724727.
substantia nigra determined by transcranial ultrasound
Chaisewikul R, Baillie N, Marson AG (2001). Calcium
correlates with severity of parkinsonian symptoms induced
antagonists as an add-on therapy for drug-resistant
by neuroleptic therapy. Biol Psychiatry 50: 463467.
epilepsy. Cochrane Database Syst Rev (4): CD002750.
Bharucha NE, Bharucha EP, Bharucha AE et al. (1988). Pre- Review.
valence of Parkinsons disease in the Parsi community of Chiueh CC, Miyake H, Peng T (1993). Role of dopamine
Bombay, India. Arch Neurol 45 (12): 13211323. autoxidation, hydroxyl radical generation and calcium
Binder RL, Jonelis FJ (1983). Seborrheic dermatitis in neuro- overload in underlying mechanisms involved in MPTP
leptic-induced parkinsonism. Arch Dermatol 119 (6): induced Parkinsonism. Adv Neurol 60: 251258.
473475. Chouza C, Scaramelli A, Caamano JL et al. (1986). Parkin-
Bloomquist J, King E, Wright A et al. (1994). 1-methyl- sonism, tardive dyskinesia, akathisia, and depression
4-phenylpyridinium-like neurotoxicity of a pyridinium induced by flunarizine. Lancet 7 (1): 13031304.
metabolite derived from haloperidol; cell culture and neu- Crane GE, Smeets RA (1974). Tardive dyskinesia and drug
rotransmitter uptake studies. J Pharmacol Exp Ther 270: therapy in geriatric patients. Arch Gen Psychiatry 30:
822830. 341343.
Borison RL, Hitri A, Blowers AJ et al. (1983). Antipsychotic Curran T, Morgan JI (1995). Fos: an immediate-early tran-
drug action: clinical, biochemical, and pharmacological scription factor in neurons. J Neurobiol 26: 403412.
evidence for site specificity of action. Clin Neuropharma- Delay J, Deniker P (1956). Chlorpromazine and neuroleptic
col 6: 137150. treatment in psychiatry. J Clin Exp Psychopathol 17: 1924.
DRUG-INDUCED PARKINSONISM 413

Delay J, Deniker P, Harl JM (1952). Utilisation en therapeu- Garcia-Ruiz PJ, Garcia de Yebenes J, Jimenez-Jimenez
tique psychiatrique dune phenothiazine daction centrale FJ et al. (1992). Parkinsonism associated with calcium
elective (4560 RP). Ann Med Psychol 110: 112117 . channel blockers: a prospective follow-up study. Clin
DellAgnello G, Ceravolo R, Nuti A et al. (2001). SSRIs do Neuropharmacol 15: 1926.
not worsen Parkinsons disease: evidence from an Gershanik OS (1994). Drug-induced parkinsonism in the
open-label, prospective study. Clin Neuropharmacol 24: aged. Recognition and prevention. Drugs Aging 5 (2):
221227. 127132.
Dick RS, Barold SS (1989). Diltiazem-induced parkinson- Gibb WR (1992). Melanin, tyrosine hydroxylase, calbindin
ism. Am J Med 87 (1): 9596. and substance P in human midbrain and substantia nigra
Dorfman-Etrog P, Hermesh H, Prilipko L et al. (1999). The in relation to nigrostriatal projections and differential neu-
effect of vitamin E addition to acute neuroleptic treatment ronal susceptibility in Parkinsons disease. Brain Res 581:
on the emergence of extrapyramidal side effects in schizo- 283291.
phrenic patients: an open label study. Eur Neuropsycho- Gil-ad I, Shtaif B, Shiloh R et al. (2001). Evaluation of the
pharmacol 9: 475477. neurotoxic activity of typical and atypical neuroleptics:
Ehringer H, Hornykiewicz O (1960). Verteilung von relevance to iatrogenic extrapyramidal symptoms. Cell
noradrenalen und dopamine (3-hydroxytyramine) im Mol Neurobiol 21: 705716.
gehirn des menschen und ihr berhatten bei erkrankingen Giladi N, Kao R, Fahn S (1997). Freezing phenomenon in
des extrapyramidalen systems. Klin Wochenschr 24: patients with parkinsonian syndromes. Mov Disord
12361239. 12 (3): 302305.
Errea-Abad JM, Ara-Callizo JR, Aibar-Remon C (1998). Glazer WM, Naftolin F, Moore DC et al. (1983). The rela-
Drug-induced parkinsonism. Clinical aspects compared tionship of circulating estradiol to tardive dyskinesia in
with Parkinson disease. Rev Neurol 27 (155): 3539. men and postmenopausal women. Psychoneuroendocrinol-
Faber R, Trimble MR (1991). Electroconvulsive therapy in ogy 8: 429434.
Parkinsons disease and other movement disorders. Mov Goswami U, Dutta S, Kuruvilla K et al. (1989). Electrocon-
Disord 6 (4): 293303. vulsive therapy in neuroleptic-induced parkinsonism. Biol
Fahn S, Mayeux R (1980). Unilateral Parkinsons disease Psychiatry 26: 234238.
and contralateral tardive dyskinesia: a unique case with Hall RA, Jackson RB, Swaim JM (1956). Neurotoxic reac-
successful therapy that may explain the pathophysiology tions resulting from chlorpromazine administration.
of these two disorders. J Neural Transm Suppl 16: JAMA 161: 214218.
179185. Hanin B, Lerner Y, Srour N (1995). An unusual effect of
ECT on drug-induced parkinsonism and tardive dystonia.
Fann WE, Lake CR (1976). Amantadine versus trihexyphe-
Convuls Ther 11: 271274.
nidyl in the treatment of neuroleptic-induced parkinson-
Hansen TE, Brown WL, Weigel RM et al. (1992). Under-
ism. Am J Psychiatry 133 (8): 940943.
recognition of tardive dyskinesia and drug-induced parkin-
Farde L, NordstriSm AL, Wiesel FA et al. (1992). Posi-
sonism by psychiatric residents. Gen Hosp Psychiatry
tron emission tomographic analysis of central D1 and
14 (5): 340344.
D2 dopamine receptor occupancy inpatients treated Hardie RJ, Lees AJ (1988). Neuroleptic-induced Parkinsons
with classical neuroleptics and clozapine: relation to syndrome: clinical features and results of treatment with
extrapyramidal side effects. Arch Gen Psychiatry 49: levodopa. J Neurol Neurosurg Psychiatry 51: 850854.
538544. Hoffman WF, Ballard L, Turner EH et al. (1991). Three-year
Fernandez Pardal M, Fernandez Pardal J, Micheli F (1988). follow-up of older schizophrenics: extrapyramidal syn-
Aggravation of Parkinsons disease by cinnarizine. J Neu- dromes, psychiatric symptoms, and ventricular brain ratio.
rol Neurosurg Psychiatry 51: 158159. Biol Psychiatry 30 (9): 913926.
Fleischhacker W (2000). Negative symptoms in patients with Horiguchi J, Nishimatsu O (1992). Usefulness of antiparkin-
schizophrenia with special reference to the primary versus sonian drugs during neuroleptic treatment and the effects
secondary distinction. Encephale 26 Spec No 1: 1214. of clonazepam on akathisia and parkinsonism occurred
Freyhan FA (1959). Therapeutic implications of differential after antiparkinsonian drug withdrawal: a double-blind
effects of new phenothiazine compounds. Am J Psychiatry study. Jpn J Psychiatry 46: 733739.
115: 577585. Huang TL (2005). Lorazepam and diazepam rapidly relieve
Friedman JH (1989). Drug-induced movement disorders. In: catatonic signs in patients with schizophrenia. Psychiatry
WJ Weiner, AE Lang (Eds.), Movement Disorders: a Clin Neurosci 59 (1): 5255.
Comprehensive Survey. Futura Publishing Co. Inc, Mount Ichise M, Kim YJ, Erami SS et al. (1999). Functional mor-
Kisco, New York, pp. 4183. phometry of the striatum in Parkinsons disease on three-
Frost JJ, Rosier AJ, Reich SG et al. (1993). Positron emis- dimensional surface display of 123I-beta-CIT SPECT
sion tomographic imaging of the dopamine transporter data. J Nucl Med 40 (4): 530538.
with 11C-WIN 35,428 reveals marked declines in mild Inoue F, Janikowski AM (1981). Withdrawal akinesia.
Parkinsons disease. Ann Neurol 34: 423431. J Neurol Neurosurg Psychiatry 44: 958.
414 F. E. MICHELI AND M. G. CERSOSIMO

Jankovic J (1995). Tardive syndromes and other drug- dopamine transporters brain SPECT. Med Clin (Barc)
induced movement disorders. Clin Neuropharmacol 18: 122 (9): 325328.
197214. Lucetti C, Nuti A, Pavese N et al. (1998). Flunarizine in
Jellinger K (1986). Overview of morphologic changes in migraine prophylaxis: predictive factors for a positive
Parkinsons disease. In: MD Yahr, KJ Bergman (Eds.), response. Cephalagia 18: 349352.
Advances in Neurology, Vol. 45, Raven Press, New York, Luchins DJ, Jackman H, Meltzer HY (1983). Lateral ventri-
pp. 118. cular size and drug-induced Parkinsonism. Psychiatry Res
Jennings DL, Seibyl JP, Oakes D et al. (2004). (123I) beta-CIT 9 (1): 916.
and single-photon emission computed tomographic imaging Lyne J, Kelly BD, OConnor WT (2004). Schizophrenia:
vs clinical evaluation in Parkinsonian syndrome: unmasking a review of neuropharmacology. Ir J Med Sci 173:
an early diagnosis. Arch Neurol 61 (8): 12241229. 155159.
Jensen K, Amdisen A (1964). Treatment of drug-induced Magnus RV (1980). A comparison of biperiden hydrochlor-
parkinsonism: a comparison between UK-738, orphena- ide (Akineton) and benzhexol (Artane) in the treatment
drine, and a placebo in a double blind study. Psychophar- of drug-induced Parkinsonism. J Int Med Res 8 (5):
macologia 192: 301305. 343346.
Jeste DV, Lohr JB, Eastham JH et al. (1998). Adverse neuro- Mamo DC, Sweet RA, Keshavan MS (1999). Managing
biological effects of long-term use of neuroleptics: human antipsychotic-induced parkinsonism. Drug Saf 20: 269275.
and animal studies. J Psychiatr Res 32 (34): 201214. Marek KL, Seibyl JP, Zoghbi SS et al. (1996). 123-ICIT/
Kelly JT, Abuzzahab FS Sr (1971). The antiparkinson prop- SPECT imaging demonstrates bilateral loss of dopamine
erties of amantadine in drug-induced parkinsonism. J Clin transporters in hemi-Parkinsons disease. Neurology 46:
Pharmacol New Drugs 11 (3): 211214. 231237.
Kelly JT, Zimmermann RL, Abuzzahab FS et al. (1974). A Marinus J, Ramaker C, Van Hilten JJ et al. (2002). Health
double-blind study of amantadine hydrochloride versus related quality of life in Parkinsons disease: a systematic
benztropine mesylate in drug-induced parkinsonism. review of disease specific instruments. J Neurol Neurosurg
Pharmacology 12 (2): 6573. Psychiatry 72: 241248.
Kennedy PF, Hershon HI, McGuire RJ (1971). Extrapyr- Marshall V, Grosset D (2003). Role of dopamine transporter
amidal disorders after prolonged phenothiazine therapy. imaging in routine clinical practice. Mov Disord 18 (12):
Br J Psychiatry 118: 509518. 14151423.
Kim JH, Byun HJ (2003). Prevalence and characteristics of Marti Masso JF, Poza JJ (1996). Drug-induced or aggravated
subjective akathisia, objective akathisia, and mixed akathi- parkinsonism: clinical signs and the changing pattern of
sia in chronic schizophrenic subjects. Clin Neuropharma- implicated drugs. Neurologia 11: 1015.
col 26 (6): 312316. Marti-Masso JF, Poza JJ (1998). Cinnarizine-induced parkin-
Klawans HL Jr, Bergen D, Bruyn GW (1973). Prolon- sonism: ten years later. Mov Disord 13: 453456.
ged drug-induced Parkinsonism. Confin Neurol 35 (6): Marti Masso JF, Carrera N, Urtasun M (1993). Drug-induced
368377. parkinsonism: a growing list. Mov Disord 8 (1): 125.
Kuzuhara S (1997). Drug-induced parkinsonism. Nippon Melamed E, Achiron A, Shapira A et al. (1991). Persistent
Rinsho 55 (1): 112117. and progressive parkinsonism after discontinuation of
Kuzuhara S (2000). Essential points to differentiate various chronic neuroleptic therapy: an additional tardive syn-
diseases causing parkinsonism. Nippon Rinsho 58 (10): drome? Clin Neuropharmacol 14 (3): 273278.
20492053. Mena MA, Garcia de Yebenes MJ, Tabernero C et al.
Kurz M, Hummer M, Oberbauer H et al. (1995). Extrapyra- (1995). Effects of calcium antagonists on the dopamine
midal side effects of clozapine and haloperidol. Psycho- system. Clin Neuropharmacol 18: 410426.
pharmacology (Berl) 118: 5256. Metzer WS, Newton JE, Steele RW et al. (1989). HLA anti-
Leo RJ, Lichter DG, Hershey LA (1995). Parkinsonism asso- gens in drug-induced parkinsonism. Mov Disord 4 (2):
ciated with fluoxetine and cimetidine: a case report. J Ger- 121128.
iatr Psychiatr Neurol 84: 231233. Metzer WS, Paige SR, Newton JE (1993). Inefficacy of
Leopold NA (1996). Dysphagia in drug-induced parkinson- propranolol in attenuation of drug-induced parkinsonian
ism: a case report. Dysphagia 11 (2): 151153. tremor. Mov Disord 8 (1): 4346.
Llau ME, Nguyen L, Senard JM et al. (1994). Drug-induced Micheli F, Pardal MF, Gatto M et al. (1987). Flunarizine-
parkinsonian syndromes: a 10-year experience at a regio- and cinnarizine-induced extrapyramidal reactions. Neurol-
nal center of pharmaco-vigilance. Rev Neurol (Paris) ogy 37: 881884.
150 (11): 757762. Micheli F, Fernandez Pardal M, Gatto M et al. (1993). Brux-
Llau ME, Senard JM, Rascol O et al. (1995). Movement disorders ism secondary to chronic antidopaminergic drug exposure.
induced by drugs: experience at a pharmaco-vigilance center Clin Neuropharmacol 16 (4): 315323.
over five years. Therapie 50 (5): 425427. Micheli FE, Pardal MM, Giannaula R et al. (1989). Move-
Lorenzo Bosquet C, Miquel Rodriguez F, Roca Bielsa I et al. ment disorders and depression due to flunarizine and
(2004). Differential diagnosis of parkinsonism using cinnarizine. Mov Disord 4: 139146.
DRUG-INDUCED PARKINSONISM 415

Moghal S, Rajput AH, Meleth R et al. (1995). Prevalence of Rajput AH, Pahwa R, Pahwa P et al. (1993). A Prognostic
movement disorders in institutionalized elderly. Neuroepi- significance of the onset mode in parkinsonism. Neurol-
demiology 14 (6): 297300. ogy 43 (4): 829830.
Moleman P, Schmitz PJ, Ladee GA (1982). Extrapyramidal Reyes MG, Faraldi F, Senseng CS et al. (1991). Nigral
side effects and oral haloperidol: an analysis of explanatory degeneration in acquired immune deficiency syndrome
patient and treatment characteristics. J Clin Psychiatry 43: (AIDS). Acta Neuropathol (Berl) 82: 3944.
492496. Robertson GS, Fibiger HC (1992). Neuroleptics increase
Morris JC, Drazner M, Fulling K et al. (1989). Clinical and c-fos expression in the forebrain: contrasting effects of
pathological aspects of parkinsonism in Alzheimers dis- haloperidol and clozapine. Neuroscience 46: 315328.
ease. A role for extranigral factors? Arch Neurol 46 (6): Rollema H, Skolnik M, DEngelbronner J et al. (1994). P(C)-
651657. like neurotoxicity of a pyridinium metabolite derived from
Naito Y, Kuzuhara S (2004). Essential points to differentiate haloperidol: in vivo microanalysis and in vitro mitochon-
various diseases causing parkinsonism. Nippon Rinsho drial studies. J Pharmacol Exp Ther 268: 380387.
62 (9): 16081616. Rosen A (1963). Drug-induced parkinsonism and its treat-
Negrotti A, Calzetti S (1997). A long-term follow-up study ment with Akineton. Ther Umsch 20: 912.
of cinnarizine- and flunarizine-induced parkinsonism. Saltz BL, Woerner MG, Robinson DG et al. (2000). Side
Mov Disord 12: 107110. effects of antipsychotic drugs. Avoiding and minimizing
Negrotti A, Calzetti S, Sasso E (1992). Calcium-entry blockers- their impact in elderly patients. Postgrad Med 107 (2):
induced parkinsonism: possible role of inherited suscepti- 169178.
bility. Neurotoxicology 13 (1): 261264. Sandyk R (1993). Cigarette smoking: effects on cognitive
Nguyen N, Pradel V, Micallef J et al. (2004). Drug-induced functions and drug-induced parkinsonism in chronic schi-
parkinson syndromes. Therapie 59 (1): 105112. zophrenia. Int J Neurosci 70 (34): 193197.
Offen D, Ziv I, Gorodin S et al. (1995). Dopamine-induced Sandyk R, Kay SR (1992). Positive and negative move-
programmedcell death in mouse thymocytes. Biochem ment disorders in schizophrenia. Int J Neurosci 66 (34):
Biophys Acta 1268: 171177. 143151.
Ohashi K, Hamamura T, Lee Y et al. (1998). Propranolol Schmidt WJ, Mayerhofer A, Meyer A et al. (2002). Ecstasy
attenuates haloperidol-induced Fos expression in discrete counteracts catalepsy in rats, an anti-parkinsonian effect?
regions of rat brain: possible brain regions responsible Neurosci Lett 330: 251254.
for akathisia. Brain Res 802: 134140. Sethi KD, Zamrini EY (1990). Asymmetry in clinical fea-
OShea E, Colado MI (2003). Is frequent dosing with ecstasy tures of drug-induced parkinsonism. J Neuropsychiatry
a risky business for dopamine-containing neurons? Trends Clin Neurosci 2 (1): 6466.
Pharmacol Sci 24: 272274. Spigset O (1999). Adverse reactions of selective serotonin
OSuilleabhain P, Giller C (2003). Rapidly progressive reuptake inhibitors: reports from a spontaneous reporting
parkinsonism in a self-reported user of ecstasy and other system. Drug Saf 20: 277287.
drugs. Mov Disord 18 (11): 13781381. Stacy M, Jankovic J (1992). Tardive tremor. Mov Disord 7:
Pina Latorre MA, Modrego PJ, Rodilla F et al. (2001). 5357.
Parkinsonism and Parkinsons disease associated with Steck H (1954). Le syndrome extrapyramidal et diencephali-
long-term administration of sertraline. J Clin Pharm Ther que au cours de traitments au Largactril et au serpasil. Ann
26: 111112. Med Psychol 2: 737744.
Pitagoras de Mattos J, Zuma de Rosso AL, Branco Correa Strous RD, Alvir JM, Robinson D et al. (2004). Premorbid
R et al. (2002). Movement disorders in 28 HIV-infected functioning in schizophrenia: relation to baseline symp-
patients. Arq Neuropsiquiatr 60: 525530. toms, treatment response, and medication side effects.
Prosser ES, Csernansky JG, Kaplan J et al. (1987). Depres- Schizophr Bull 30 (2): 265278.
sion, parkinsonian symptoms, and negative symptoms in Sulzer D, Maidment NT, Rayport S (1993). Amphetamine
schizophrenics treated with neuroleptics. J Nerv Ment and other weak bases act to promote reverse transport of
Dis 175 (2): 100105. dopamine in ventral midbrain neurons. J Neurochem 60:
Prueter C, Habermeyer B, Norra C et al. (2003). Akathisia as 527535.
a side effect of antipsychotic treatment with quetiapine in Teive HA, Troiano AR, Germiniani FM et al. (2004). Flunar-
a patient with Parkinsons disease. Mov Disord 18: izine and cinnarizine-induced parkinsonism: a historical
712713. and clinical analysis. Parkinsonism Relat Disord 10 (4):
Quinn NP (1995). Parkinsonism: recognition and differential 243245.
diagnosis. BMJ 310; 44: 7452. Terland O, Flatmark T (1999). Drug-induced parkinsonism:
Rajput AH, Rozdilsky B, Hornykienicz O (1982). Reversible cinnarizine and flunarizine are potent uncouplers of the
drug-induced parkinsonism, clinicopathologic study of vacuolar H-ATPase in catecholamine storage vesicles.
two cases. Neurology 39: 644646. Neuropharmacology 38: 879882.
Rajput AH, Offord KP, Beard CM et al. (1984). Epidemiol- Tolosa E, Coelho M, Gallardo M (2003). DAT imaging in
ogy of parkinsonism: incidence, classification, and mortal- drug-induced and psychogenic parkinsonism. Mov Disord
ity. Ann Neurol 16: 278282. 18 (Suppl 7): S28S33.
416 F. E. MICHELI AND M. G. CERSOSIMO

Uhnbrand L, Faurbye A (1969). Reversible and irreversible disorders: a cautionary study. Am J Psychiatry 144:
dyskinesias after treatment with perphenazine, chlorpro- 11481153.
mazine, reserpine and electroconvulsive therapy. Psycho- Yamasue H, Fukui T, Fukuda R et al. (2003). Drug-induced
pharmacology (Berl) 1: 408418. parkinsonism in relation to choline-containing compounds
Vaccari A, Saba P, Gessa G (1993). Potent, extra-channel influ- measured by 1H-MR spectroscopy in putamen of chroni-
ence of several calcium-channel modulators on striatal bind- cally medicated patients with schizophrenia. Int J Neurop-
ing of [3H]tyramine. Neurochem Res 18: 11251130. sychopharmacol 6 (4): 353360.
Van der Heijden FM, Tuinier S, Arts NJ et al. (2005). Catatonia: Ziv I, Melamed E, Nardi N et al. (1995). Dopamine induces
disappeared or under-diagnosed? Psychopathology 38: 38. apoptosis-like cell death in cultured chick sympathetic
Veitch K, Hue L (1994). Flunarizine and cinnarizine inhibit neuronsa possible novel pathogenic mechanism in
mitochondrial complexes I and II: possible implication Parkinsons disease. Neurosci Lett 170: 136140.
for parkinsonism. Mol Pharmacol 45: 158163.
Wada Y, Yamaguchi N (1992). The rabbit syndrome and
Further Reading
antiparkinsonian medication in schizophrenic patients.
Neuropsychobiology 25 (3): 149152. Bhatia K, Daniel SE, Marsden CD (1993). Orofacial dysto-
Weiden PJ, Mann JJ, Haas G et al. (1987). Clinical nia and rest tremor in a patient with normal brain pathol-
nonrecognition of neuroleptic-induced movement ogy. Mov Disord 8 (3): 361362.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 52

Vascular parkinsonism

YACOV BALASH1 AND AMOS D. KORCZYN2*

1
Movement Disorders Unit, Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
2
Sieratzki Chair of Neurology, Tel-Aviv University Medical School, Ramat-Aviv, Israel

Parkinsons disease (PD) is a degenerative brain This concept was criticized by Schwab and England
disease consisting of a progressive loss of dopaminer- (1968) and Parkes et al. (1974), who claimed that a coin-
gic neurons in the substantia nigra (SN) and other neu- cidence or superposition of vascular changes in some
ronal systems in certain brain areas. A loss of 6070% patients with PD could account for this overlap. Many
of the nearly 450 000 dopamine-producing neurons of years later Critchley himself corrected his basic idea
the SN pars compacta and related decrease of the of arteriosclerosis as a possible cause of PD and termed
dopamine level at the striatum must occur before PD the condition arteriosclerotic pseudoparkinsonism,
symptoms will take place. Several other diseases share considered as alien to PD both clinically and pathogen-
some clinical features of PD, like tremor, muscle rigid- etically (Critchley, 1981). Nevertheless, the neurologi-
ity, hypokinesia and bradykinesia, gait instability and cal entity of VP remains a matter of debate ever since.
freezing of gait. These disorders, collectively called Up to now, VP and PD are not clearly separated clini-
parkinsonian, can sometimes be clearly distinguished cally and for example are lumped together in the Ninth
from PD, although on other occasions the distinction Revision of the International Classification of Diseases
is difficult. Neuroimaging data (e.g. single-photon (ICD-9) as item 332.0 without any differentiation
emission computed tomography, SPECT), drug between arteriosclerotic and idiopathic, primary
response or the pathological demonstration of specific parkinsonism (ICD-9-CM International Classification
changes in the SN or elsewhere can help in such cases. of Diseases, 2005).
The present review will discuss one of these entities, Until recently, VP remained a poorly defined
called vascular parkinsonism (VP). clinicopathological entity, which overlaps with other
In 1929 Critchley, after analysis of the literature, diagnoses. There are few pathologically verified
described five types of parkinsonism induced, accord- cases with suitable clinical correlation. The clinical
ing to his opinion, by cerebrovascular diseases in diagnosis of VP may be difficult and sometimes its
elderly patients. He identified the first (commonest) confirmation is only possible by neuropathological
type as rigidity, fixed facies and short-stepping gait examination, which is still considered the gold
with absence of rest tremor; the second type is similar standard for making the definite diagnosis of VP
but with the addition of pseudobulbar manifestations; (Jellinger, 2002). However, pathological diagnosis of
the third type has the addition of dementia and incon- VP is non-specific and the changes are largely those
tinence; the fourth type shows signs of pyramidal dis- of small-vessel disease. Because of this fact there are
ease; and the fifth has superimposition of a cerebellar doubts whether VP should be regarded as a clinical
syndrome. entity or, more correctly, whether VP is a variant of
Critchley suggested that VP is a spectrum of hetero- small-vessel brain disease, manifested neurologically
geneous syndromes resulting from multiple vascular by parkinsonian-like features, usually accompanied
(arteriosclerotic or syphilitic) lesions in the basal by additional neurological signs (Zijlmans et al.,
ganglia (BG), rather than a certain idiopathic disease. 2004).

*Correspondence to: Professor Amos D. Korczyn, Tel-Aviv University Medical School, Sieratzki Chair of Neurology,
Ramat-Aviv 69978, Israel. Email: amosk@tasmc.health.gov.il, Tel: 972-3-697-4229, Fax: 972-3-640-9113.
418 Y. BALASH AND A. D. KORCZYN
52.1. Parkinsonism associated with course, more extensive lesions due to basilar occlusion
brainstem lesions will also affect the SN but the extrapyramidal signs
will be overshadowed by other manifestations of
Vascular lesions to the brain can cause almost any con- brainstem dysfunction.
stellation of neurological symptoms, depending on the Acute or subacute parkinsonism was also observed
site of the lesions and other factors, and parkinsonism as a result of tentorial herniations with brainstem
cannot be assumed to be as exception. Thus, focal hemor- compression in patients with subdural hematomas of
rhagic and ischemic brainstem strokes were shown to traumatic or hypertensive etiology or as a result of
produce contralateral parkinsonian signs. To illustrate rupture of arterial aneurysms (Cosi and Tonali, 1964;
the point we can discuss a case of a patient in whom right Pau et al., 1989; Trosch and Ransom, 1990; Turjanski
hemiparkinsonism was diagnosed following hemorrhage et al., 1997). In some of these cases parkinsonism
into the left SN. Technetium-99m hexamethylpropylene improved following removal of the hematoma (Sunada
amine oxime (99mTc-HMPAO) single photon emission et al., 1996), but in others it persisted and usually
computed tomography (SPECT) showed reduced uptake responded to levodopa (Trosch and Ransom, 1990).
in the left putamen, globus pallidus (GP) and thalamus Sudden onset of small-stepping gait with stooped pos-
(Abe and Yanagihara, 1996). The SPECT abnormalities ture, severe akinesia and freezing of gait and increased
were assumed to be the result of the stroke, rather than muscle tone with bilateral cogwheel phenomenon in
a coincidental abnormality. the arms was seen in a patient with a small high-
In another report, a patient developed right-sided density lesion in the medial side of the right cerebral
cogwheel rigidity and resting tremor. In this case the peduncle and a lens-shaped low-density lesion in
magnetic resonance imaging (MRI) scan showed a the left putamen. On T1- and T2-weighted MRI
contralateral midbrain hemorrhage affecting the SN images a low signal was seen in a right peduncular
and n-isopropyl-p-123I iodoamphetamine (123I-IMP) lesion extending to the SN, which was also partially
SPECT images showed reduced radioisotope uptake destroyed. The parkinsonism rapidly improved and
in the left striatum, thalamus and frontal lobe (Inoue completely disappeared within 2 weeks. The high-
et al., 1997). density lesion of the right peduncle also disappeared
These cases had in common an apoplectic appearance on CT (Miyagi et al., 1992). Interestingly, destruc-
of unilateral parkinsonian signs, which are the logical tion of the left subthalamic nucleus by hematoma
result of an ischemic or hemorrhagic lesion of the SN can improve contralateral parkinsonian rigidity after
or the nigrostriatal tract and therefore constitute the most extinction of the ballistic movement phase (Yamada
straightforward examples of VP. Such cases should have et al., 1992).
an acute onset, be non-progressive and even improve Kim (2001) reported patients with (hemi)parkinson-
spontaneously over time and respond to levodopa. How- ism after infarction in the territory of the anterior cer-
ever, such cases are extremely rare. Microscopically ebral artery. Parkinsonism was usually related to large
such cases should not contain Lewy bodies. lesions involving the supplementary motor area and
The etiology of vascular damage should not observed after the motor dysfunction improved in
necessarily be atherosclerotic. Unilateral rest tremor patients with initially severe limb weakness.
associated with bilateral extrapyramidal syndrome In patients with clinically suspected VP, the com-
responsive to levodopa was seen in a 25-year-old parison of levodopa response with the pathologically
man 7 months after subarachnoid hemorrhage due to confirmed presence of macroscopic vascular damage
rupture of a peduncular subthalamic arteriovenous in the nigrostriatal pathway and cell loss in the SN
malformation (Defer et al., 1994). showed that the majority of patients with good or
Parkinsonism has been described in patients with excellent response to levodopa had infarcts or lacunae
central nervous system vasculitis (Fabiani et al., in the SN in the absence of Lewy bodies (Zijlmans
2002), rheumatoid arthritis (Ertan et al., 1999; Hrycaj et al., 2004).
et al., 2003), systemic lupus erythematosus (Tan These anecdotal cases of vascular lesions of the SN
et al., 2001; Garcia-Moreno and Chacon, 2002), anti- or nigrostriatal pathway had features resembling PD,
phospholipid syndrome (Milanov and Bogdanova, including responsiveness to levodopa. However, the
2001; Adhiyaman et al., 2002; Huang et al., 2002), follow-up, long-term prognosis and special features
disseminated intravascular coagulation (Yoritaka of the dopaminergic treatment in relation to PD are
et al., 1997) and some neuroinfections like syphilis unknown. Motor fluctuations, so common in advanced
(Critchley, 1929; Sandyk, 1983; Pineda et al., 2000). PD (Korczyn, 1972), have not so far been described in
Not unexpectedly, most patients had additional cases such as these. Neither had a gradual increase in
manifestations, consistent with the damaged area. Of dose over the years been documented. These cases
VASCULAR PARKINSONISM 419
then typically consist of a non-progressive, levodopa- following short terms after exposure to CO (Silverman
responsive (hemi)parkinsonism of acute onset. et al., 1993). MRI abnormalities augmented with CT
findings that revealed bilateral lucencies of the GP
52.2. Parkinsonism associated with basal suggested that their damage is of vascular origin, prob-
ganglia lesions ably hemorrhagic infarction (Bianco and Floris, 1996).
Old necrotic lesions of the GP were earlier found on CT
Ischemic infarcts in the territory of the lenticulostriate scans (Klawans et al., 1982). No correlation between
arteries were said to be the most frequent cause of the neuroimaging findings and the development of
unilateral parkinsonism (Friedman et al., 1986; parkinsonism has, however, been demonstrated (Choi,
Kulisevsky et al., 1996; Fenelon and Houeto, 1997; 2002). The damage in CO poisoning, however, is not
Sibon et al., 2004a). In these cases, there was an acute restricted to the BG and the direct role of the BG
onset of the disorder with supportive imaging data to damage is unknown in these cases.
suggest the vascular etiology.
A lateralized subacute parkinsonism with CT evi- 52.3. Parkinsonism with white-matter lesions
dence of a single lacunar infarct in the contralateral
BG was described by Lazzarino et al. (1990). A clini- A more common picture is that of short-stepped gait
cal syndrome indistinguishable from PD, in which with or without freezing (Giladi et al., 1997), lead-pipe
postmortem examination revealed extensive lacunar rigidity, no or mild upper-limb involvement, absence
infarctions of the BG without evidence of coexistent of rest tremor, which develops insidiously, and nega-
PD, was also reported (Murrow et al., 1990). However, tive response to levodopa (FitzGerald and Jankovic,
subdivision of parkinsonian patients with lacunar 1989; Chang et al., 1992; Yamanouchi and Nagura,
infarcts in the BG into those with lacunes in the caudate 1995; van Zagten et al., 1998; Winikates and Jankovic,
nucleus, lentiform nucleus or both did not correlate with 1999). Pseudobulbar palsy is observed in nearly half
the clinical presentation (Reider-Groswasser et al., of the patients (Yamanouchi and Nagura, 1995). The
1995). Lacunar infarctions in the BG are frequently patients frequently have cognitive decline and detrusor
observed in patients without parkinsonism and even in overactivity (Cummings, 1994; Holstein et al., 1994).
completely asymptomatic cases. The extrapyramidal Long tract signs in the limbs, indicating damage to
signs, if present, can be asymmetrical without consis- the corticospinal tracts, are found in more then half
tent relationship to the side of the lacuna (Inzelberg of patients (Yamanouchi and Nagura, 1997). Many of
et al., 1994). these patients have vascular risk factors and particu-
Specific regions of the brain, such as central white larly hypertension. The evolution of this disease is
matter, GP and hippocampus, are selectively vulnerable typically slow, occurring over several month or even
to carbon monoxide (CO) (Koehler and Traystman, years (Inzelberg et al., 1994), an evolution similar to
2002). Parkinsonism after CO poisoning could be seen that seen in PD. Brain imaging shows diffuse subcortical
in 10% of sufferers from CO encephalopathy (Choi, white-matter damage, not unlike that seen in patients
2002). The most frequent signs are symmetric rigidity, with Binswangers disease, sometimes accompanied by
hypokinesia, masked face, glabellar sign, grasping, unilateral or bilateral BG infarcts (Demirkiran et al.,
short-step gait and retropulsion. Rest tremor has not 2001). Loss of postural reflexes and gait abnormalities
been observed but intentional tremor may occasionally seen in these cases may indicate extrapyramidal features,
be found. The latency before the appearance of parkin- but are difficult to disassociate from coexisting spasticity
sonism varies from 2 weeks to 6 months, but most and the slowness may also reflect pyramidal, rather than
frequently occurs within 1 month of CO exposure. extrapyramidal, damage.
Together with the parkinsonism, all patients have mild Comparing the brain pathology in these patients
to severely impaired cognitive functions. Other com- with that in matched patients with Binswangers dis-
mon symptoms are mutism, gait disturbances and ease who had no parkinsonism according to clinical
urinary incontinence. Extreme cases manifest a general records failed to reveal any significant differences in
akinetic-mute state up to bed-bound (Lee and Marsden, the extent of BG damage (Yamanouchi and Nagura,
1994). Neither levodopa nor anticholinergic drugs were 1997), thus pointing to the central role of white-matter
shown to be effective. However, nearly 80% of patients damage. The number of oligodendrocytes in the fron-
improved or recovered spontaneously within 6 months tal white matter of VP patients was significantly less
(Choi, 2002). than in age-matched normal control subjects, but
Abnormally high signal bilaterally in the GP on T1- significantly more than in those with Binswangers
weighted MRI images was observed in patients disease. Widespread lesions in the frontal white matter
420 Y. BALASH AND A. D. KORCZYN
with only meager lesions in the BG were the main BG lacunae are quite common in the elderly, fre-
pathological features of VP. The extent of frontal quently without parkinsonian signs. If parkinsonian
white-matter pallor tended to be less broad in VP signs do exist they do not bear a clear relationship to
than in Binswangers disease without parkinsonism the side of the lesion. The etiology of most BG lesions
(Yamanouchi and Nagura, 1997). Marked atrophy in is microvascular and in fact many, perhaps most, cases
the frontal cortex as well as lacunar lesions around with BG lacunae also have white-matter lesions and it
the lateral ventricles in VP were previously shown by is best to look at these cases as reflecting an incidental
Grundl et al. (1991). The exact pathogenesis of these manifestation of BG lesions, whereas the principal
white-matter changes is debated and likely heteroge- changes responsible for the motor dysfunction occur
neous, but generally thought to represent areas of in the white matter.
chronic or recurrent partial ischemia. Although
patients with BG lacunar infarct could recover sponta-
neously, those with frontal lobe infarcts could remain 52.3.1. Brain imaging
static and those with periventricular and deep subcorti-
cal white-matter lesions usually had progressive The integrity of the nigrostriatal pathway can be
deterioration (Chang et al., 1992). inferred from radioligand studies, in which the chemi-
From the clinical and pathological data presented so cals used have affinity to the dopamine transporter
far, it seems that VP can occur as an acute syndrome, molecules at the terminals of the dopaminergic
resulting from lesions of the SN or the nigrostriatal synapses in the BG.
tract. These cases would be unilateral, non-progressive Small studies failed to find specific changes in VP
and respond well to levodopa. patients and indeed were not significantly different from
Diffuse white-matter damage, resulting from micro- those in healthy individuals. The characteristic reduc-
vascular lesions, typically manifests as a combination tion of the uptake is pronounced in the contralateral
of extrapyramidal, pyramidal and sometimes vascular putamen of PD patients with unilateral symptoms, but
changes, frequently with cognitive decline not in VP (Tzen et al., 2001). Normal striatal 123I-FP-
(Fig. 52.1). These cases would not typically respond CIT binding with no significant differences in striatal
to levodopa, because the lesions involve the ascending or subregional binding ratios compared with those of
loop of the BGthalamiccortical loop (Bergman and the controls was seen in patients with VP. PD patients
Deuschl, 2002), distal to the striatal site of action of had significantly diminished striatal binding compared
dopamine. Such cases are best described as pseudopar- with that of controls (Lorberboym et al., 2004). As
kinsonian because the extrapyramidal features are only compared to normal controls, patients with VP had sig-
part of a more extensive clinical picture and because nificant reduction of the ratio between N-acetylaspartate
of a lack of response to dopaminergic drugs. and creatine plus phosphocreatine in the frontal cortex

Fig. 52.1. T1 and T2 magnetic resonance images of a 73-year-old patient with severe parkinsonism resistant to levodopa, gait
disorders, freezing of gait, recurrent falls and urinary urgency. Multiple infarcts are seen in the basal ganglia and brain white
matter as well as periventricular white-matter changes (own observation).
VASCULAR PARKINSONISM 421
(Abe et al., 2000), suggesting a lesion beyond the clas- (Berding et al., 2003; Goldstein, 2004). In patients
sical extrapyramidal system. with PD, even without clinical evidence of autonomic
failure, reduction in iodine-123-metaiodobenzylgua-
52.3.2. Transcranial ultrasonography nidine (123I-MIBG) uptake on scintigraphy occurs in
the heart. This is considered to be a specific finding
The structurally normal SN could be visualized using for PD and useful for the differential diagnosis from
ultrasound waves delivered through a cranial other parkinsonian syndromes, occurring even in the
window, similar to that when flow in the middle earliest stage of the disease (Taki et al., 2000). Parkin-
cerebral arteries is examined (Fig 52.2). sonian syndromes other than PD did not demonstrate
This method can distinguish between lesions affect- reduction in MIBG uptake.
ing the SN directly, such as PD and other disorders 2. Testing olfactory function could be helpful in
causing extrapyramidal features (Berg et al., 1999). This differentiating VP from PD (Katzenschlager
effect is based on the high iron level in the SN, thought to et al., 2004). For example, olfactory function,
promote generation of reactive oxygen species (Berg diagnosed according to the Pennsylvania smell
et al., 2002) and has been shown to be specific to PD identification test, was significantly better in VP
(Berg et al., 2005). Thus, if all PD patients show a patients than in PD and did not differ from normal
bilateral SN hyperechogenicity, a low echogenic SN, controls.
particularly combined with hyperechogenic lentiform 3. Colonic transit time measured in VP patients was
nuclei, could suggest other kinds of parkinsonism normal (about 59 hours), as opposed to PD
(Walter et al., 2002; Behnke et al., 2005). (Hardoff et al., 2001), where markedly slow transit
is observed (Jost et al., 1994).
52.3.3. Additional methods of diagnosis of vascular
parkinsonism
52.3.4. Parkinsons disease versus vascular
parkinsonism: comparison of pathophysiology
Non-motor differences between VP and PD could be
used in the differential diagnosis:
PD results from the slowly progressive death of
1. Measurement of myocardial innervation may contri- selected populations of neurons, including the neuro-
bute to the differential diagnosis of parkinsonism melanin-containing dopaminergic neurons of the pars

Fig. 52.2. Sonographic image of the midbrain axial section in a patient with PD. The butterfly-shaped midbrain section of
low echogenicity is surrounded by the hyperchogenic basal cisterns (encircled area, centre of fig.). Note the marked hyperecho-
genicity of the left SN. This area is encircled for computerized measurement. Courtesy of Prof. Heinz Reichmann from the
Department of Neurology, Technical University of Dresden, Germany.
422 Y. BALASH AND A. D. KORCZYN
compacta of the SN, some other catecholaminergic Thus, PD is a neurodegenerative disease, which
and serotoninergic brainstem nuclei, the cholinergic selectively affects certain categories of neurons,
nucleus basalis of Meynert, hypothalamic neurons chiefly manifests as a progressive diminution of
and small cortical neurons (particularly in the cingu- dopamine level in the nigrostriatal system, induces an
late gyrus and entorhinal cortex), as well as the olfac- overactivation of the output nuclei of the BG and
tory bulb, sympathetic ganglia and parasympathetic STN and inhibits the thalamocortical motor system.
neurons in the gut (Korczyn, 1993; Braak and Braak, VP is a clinical syndrome that may have similarities
2000). to the cardinal motor manifestations of PD. Approxi-
According to the accepted model, dopamine loss mately 10% of cases of parkinsonism clinically recog-
increases the activity of the indirect pathway and nized as PD could pathologically be the result of
reduces the activity of the direct pathway (Wich- cerebrovascular disease and, in contrast, the emer-
mann and DeLong, 2003). The final result of these gence of one or more clinical signs of parkinsonism
changes is an overexcitation of the main output was revealed in 30% patients after stroke (Sibon
nuclei of the BG, GP interna and SN pars reticulata, et al., 2004b). As distinct from PD, however, VP is
leading to inhibition of the thalamocortical motor the result of endothelial dysfunction and arteriolo-
system. If the latter is essentially important in the sclerosis causing lacunar strokes or punctate hemor-
pathogenesis of rigidity, bradykinesia and loss of rhages, disseminated mainly in the centrum
postural reflexes, the white-matter lesions affecting semiovale and/or BG. Risk factors of cerebrovascular
the latter pathway should result in a similar pheno- disease, like hypertension, diabetes mellitus type 2,
type. However, these should not respond to levodopa hyper-/dyslipidemia, hyperhomocysteinemia, previous
since there is no dopamine loss and in any case the stroke history and genetic predisposition are usually
site of action of dopamine, the BG, is disconnected present (Keskin and Yurdakul, 1996; Ekinci et al.,
from the motor cortex. Thompson and Marsden 2004).
(1987) suggested that the gait disorder of Binswan- In the pathogenesis of VP without a direct lesion
gers disease is probably due to diffuse vascular affecting the SN, the dopamine level and its
lesions disrupting the interconnecting fiber tracts metabolites in the cerebrospinal fluid should be normal
between the BG and the motor cortex. Multiple lacu- (Loeffler et al., 1995). Thus, the accepted explanation
nar subcortical infarcts interrupting thalamocortical is that the clinical symptoms in VP are related to post-
drive are critical for the development of VP (Bhatia synaptic lesions of the nigrostriatal system mainly as a
and Marsden, 1994; van Zagten et al., 1998; Peralta result of a patchy destruction of neurons or axons at
et al., 2004). the putamen, GP and probably the cerebral white
Three characteristic features of presynaptic process matter.
affecting the dopaminergic system in PD are:
1. A major reduction of striatal levodopa uptake 52.4. Conclusions
of the nigrostriatal pathways (Leenders, 1997;
Broussolle et al., 1999; Brooks, 2004; Thobois VP is not a homogenous syndrome. Its etiology may
et al., 2004) which is correlated with degeneration be atherosclerotic in many cases, but it could result
of the SN neurons found by pathological studies from embolic, arteritic or hemorrhagic processes.
(Jellinger, 2002; Snow, 2003). This process may be Pathophysiologically, two forms of VP should be con-
accompanied by increased raclopride binding of sidered: the first affects the nigrostriatal system unilat-
postsynaptic dopamine receptors in the putamen as erally. A discrete lesion of the SN or the nigrostriatal
a sign of postsynaptic hypersensitivity in de novo, pathway should be seen in imaging. These patients
drug-naive PD patients (Antonini et al., 1994; Kaasi- should have an acute onset, should not progress and
nen et al., 2000). should respond to levodopa. The other, much more
2. An obvious effect of levodopa, which can be common group, consists of patients presenting at more
seen particularly at the early stages of PD advanced age and with predominantly lower-body
(Hornykiewicz, 2002). involvement and gait abnormalities rather than tremor.
3. Presence of pathologic hallmarks degenerating A history of stroke, heart disease, arterial hypertension
ubiquitin-positive neuronal processes or neurites and other risk factors for stroke support a vascular ori-
(Lewy neurites), which have been found in all gin of these symptoms. Usually, these patients do not
affected brainstem regions and especially at the have the typical parkinsonian rest tremor. In these
dorsal motor nucleus of the vagus and later in cases imaging demonstrates an extensive white-matter
the SN (Braak et al., 1999; Gai et al., 2000). disease.
VASCULAR PARKINSONISM 423
BG lesions are frequently seen in patients with Bhatia KP, Marsden CD (1994). The behavioural and motor
parkinsonian symptoms, but their relationship to the consequences of focal lesions of the basal ganglia in man.
pathology is unclear since the onset of parkinsonism Brain 117: 859876.
is rarely acute and not strictly contralateral to the Bianco F, Floris R (1996). MRI appearances consistent with
damage (typically, a small lacuna). Moreover, some haemorrhagic infarction as an early manifestation of car-
people with similar lacunae do not demonstrate any bon monoxide poisoning. Neuroradiology 38 (Suppl 1):
clinical manifestations. Probably, this VP syndrome S70S72.
is the result of coexistent white-matter changes. Braak H, Braak E (2000). Pathoanatomy of Parkinsons dis-
ease. J Neurol 247 (Suppl 2): II3II10.
These cases are most likely to represent the entity of
pseudoparkinsonism, as defined by Critchley. The Braak H, Sandmann-Keil D, Gai W et al. (1999). Extensive
axonal Lewy neurites in Parkinsons disease: a novel
extensive damage ensures that a patient suffers not
pathological feature revealed by alpha-synuclein immuno-
only from increased muscle tone and bradykinesia, cytochemistry. Neurosci Lett 265: 6769.
but typically also from cognitive decline, pseudobulbar
Brooks DJ (2004). Neuroimaging in Parkinsons Disease.
syndrome, posture and gait instability and urinary Neurorx 1: 243254.
incontinence. Obviously these patients will respond Broussolle E, Dentresangle C, Landais P et al. (1999). The
poorly to levodopa and there is an urgent need to relation of putamen and caudate nucleus 18F-Dopa uptake
develop drugs that could help these patients. to motor and cognitive performances in Parkinsons dis-
ease. J Neurol Sci 166: 141151.
Chang CM, Yu YL, Ng HK et al. (1992). Vascular pseudo-
References parkinsonism. Acta Neurol Scand 86: 588592.
Choi IS (2002). Parkinsonism after carbon monoxide poison-
Abe K, Yanagihara T (1996). Hemiparkinsonism following ing. Eur Neurol 48: 3033.
haemorrhage in the contralateral substantia nigra. Cosi V, Tonali P (1964). Acute Parkinson syndrome follow-
Neuroradiology 38 (Suppl 1): S67S69. ing subarachnoid hemorrhage in a schizophrenic subject.
Abe K, Terakawa H, Takanashi M et al. (2000). Proton Riv Patol Nerv Ment 85: 287301.
magnetic resonance spectroscopy of patients with Critchley M (1929). Arteriosclerotic parkinsonism. Brain 52:
parkinsonism. Brain Res Bull 52: 589595. 2383.
Adhiyaman V, Meara RJ, Bhowmick BK (2002). Anti-
Critchley M (1981). Arteriosclerotic pseudoparkinsonism.
phospholipid syndrome and dystonia-parkinsonism: need
In: FC Rose, R Capildeo (Eds.), 74: Pitman, London, pp.
for anticoagulation. Parkinsonism Relat Disord 8: 215.
745752.
Antonini A, Schwarz J, Oertel WH et al. (1994). [11C]raclo-
Cummings JL (1994). Vascular subcortical dementias: clini-
pride and positron emission tomography in previously
cal aspects. Dementia 5: 177180.
untreated patients with Parkinsons disease: influence of
Defer GL, Remy P, Malapert D et al. (1994). Rest tremor
L-dopa and lisuride therapy on striatal dopamine
and extrapyramidal symptoms after midbrain haemor-
D2-receptors. Neurology 44: 13251329.
rhage: clinical and 18F-dopa PET evaluation. J Neurol
Behnke S, Berg D, Naumann M et al. (2005). Differentiation
Neurosurg Psychiatry 57: 987989.
of Parkinsons disease and atypical parkinsonian syn-
dromes by transcranial ultrasound. J Neurol Neurosurg Demirkiran M, Bozdemir H, Sarica Y (2001). Vascular par-
Psychiatry 76: 423425. kinsonism: a distinct, heterogeneous clinical entity. Acta
Berg D, Becker G, Zeiler B et al. (1999). Vulnerability of the Neurol Scand 104 (2): 6367.
nigrostriatal system as detected by transcranial ultrasound. Ekinci B, Apaydin H, Vural M et al. (2004). Two siblings
Neurology 53: 10261031. with homocystinuria presenting with dystonia and parkin-
Berg D, Roggendorf W, Schroder U et al. (2002). Echogeni- sonism. Mov Disord 19: 962964.
city of the substantia nigra: association with increased iron Ertan S, Fresko I, Apaydin H et al. (1999). Extrapyramidal
content and marker for susceptibility to nigrostriatal type rigidity in rheumatoid arthritis. Rheumatology
injury. Arch Neurol 59: 9991005. (Oxford) 38: 627630.
Berg D, Merz B, Reiners K et al. (2005). Five-year follow-up Fabiani G, Teive HA, Germiniani FM et al. (2002). Reversi-
study of hyperechogenicity of the substantia nigra in ble parkinsonian syndrome in systemic and brain vasculi-
Parkinsons disease. Mov Disord 20: 383385. tis. Mov Disord 17: 601604.
Berding G, Schrader CH, Peschel T et al. (2003). [N-methyl Fenelon G, Houeto JL (1997). Unilateral parkinsonism fol-
11C]meta-Hydroxyephedrine positron emission tomogra- lowing a large infarct in the territory of the lenticulostriate
phy in Parkinsons disease and multiple system atrophy. arteries. Mov Disord 12: 10861090.
Eur J Nucl Med Mol Imaging 30: 127131. FitzGerald PM, Jankovic J (1989). Lower body parkinsonism:
Bergman H, Deuschl G (2002). Pathophysiology of evidence for vascular etiology. Mov Disord 4: 249260.
Parkinsons disease: from clinical neurology to basic Friedman A, Kang UJ, Tatemichi TK et al. (1986). A case
neuroscience and back. Mov Disord 17 (Suppl 3): of parkinsonism following striatal lacunar infarction.
S28S40. J Neurol Neurosurg Psychiatry 49: 10871088.
424 Y. BALASH AND A. D. KORCZYN

Gai WP, Yuan HX, Li XQ et al. (2000). In situ and in vitro Parkinsons disease. J Neurol Neurosurg Psychiatry 75:
study of colocalization and segregation of alpha-synuclein, 17491752.
ubiquitin, and lipids in Lewy bodies. Exp Neurol 166: Keskin S, Yurdakul F (1996). Parkinsonian manifestations in
324333. a patient with homocystinuria. Parkinsonian manifesta-
Garcia-Moreno JM, Chacon J (2002). Juvenile parkinsonism tions in a patient with homocystinuria. Child Neurol 11:
as a manifestation of systemic lupus erythematosus: case 235236.
report and review of the literature. Mov Disord 17: Kim JS (2001). Involuntary movements after anterior cere-
13291335. bral artery territory infarction. Stroke 32: 258261.
Giladi N, Kao R, Fahn S (1997). Freezing phenomenon in Klawans HL, Stein RW, Tanner CM et al. (1982). A pure
patients with parkinsonian syndromes. Mov Disord 12: parkinsonian syndrome following acute carbon monoxide
302305. intoxication. Arch Neurol 39: 302304.
Goldstein DS (2004). Functional neuroimaging of sympa- Koehler RC, Traystman RJ (2002). Cerebrovascular effects
thetic innervation of the heart. Ann NY Acad Sci 1018: of carbon monoxide. Antioxid Redox Signal 4: 279290.
231243. Korczyn AD (1972). Pathophysiology of drug-induced dys-
Grundl W, Ziegler R, Westphal KP et al. (1991). Parkinsons kinesias. Neuropharmacology 11: 601607.
syndrome: cranial computed-tomography findings. Korczyn AD (1993). The gut in PD. Neurology 43: 629630.
Their dependence on sex and age. Acta Med Hung 48: Kulisevsky J, Berthier ML, Avila A et al. (1996). Unilateral
127136. parkinsonism and stereotyped movements following a
Hardoff R, Sula M, Tamir A et al. (2001). Gastric emptying right lenticular infarction. Mov Disord 11: 752754.
time and gastric motility in patients with Parkinsons Lazzarino LG, Nicolai A, Toppani D (1990). Subacute par-
disease. Mov Disord 16: 10411047. kinsonism from a single lacunar infarct in the basal gang-
Holstein J, Chatellier G, Piette F et al. (1994). Prevalence of lia. Acta Neurol (Napoli) 12: 292295.
associated diseases in different types of dementia among Loeffler DA, LeWitt PA, DeMaggio AJ et al. (1995). Mar-
elderly institutionalized patients: analysis of 3447 records. kers of dopamine depletion and compensatory response
J Am Geriatr Soc 42: 972977. in striatum and cerebrospinal fluid. J Neural Transm Park
Hornykiewicz O (2002). L-DOPA: from a biologically inac- Dis Dement Sect 9: 4553.
tive amino acid to a successful therapeutic agent. Amino Lorberboym M, Djaldetti R, Melamed E et al. (2004).
Acids 23: 6570. 123I-FP-CIT SPECT imaging of dopamine transporters
Hrycaj P, Korczowska I, Lacki JK (2003). Severe Parkin- in patients with cerebrovascular disease and clinical diag-
sons disease in rheumatoid arthritis patient treated with nosis of vascular parkinsonism. J Nucl Med 45:
infliximab. Rheumatology (Oxford) 42: 702703. 16881693.
Huang Z, Jacewicz M, Pfeiffer RF (2002). Anticardiolipin Lee MS, Marsden CD (1994). Neurological sequelae follow-
antibody in vascular parkinsonism. Mov Disord 17: ing carbon monoxide poisoning clinical course and out-
992997. come according to the clinical types and brain computed
ICD-9-CM International Classification of Diseases (2005), tomography scan findings. Mov Disord 9: 550558.
9th Rev: Clinical Modification Vol. 1. Practice Manage- Leenders KL (1997). Pathophysiology of movement disor-
ment Information Corporation. ders studied using PET. J Neural Transm Suppl 50: 3946.
Inoue H, Udaka F, Takahashi M et al. (1997). Secondary Milanov I, Bogdanova D (2001). Antiphospholipid syndrome
parkinsonism following midbrain hemorrhage. Rinsho and dystonia-parkinsonism. A case report. Parkinsonism
Shinkeigaku 37: 266269. Relat Disord 7: 139141.
Inzelberg R, Bornstein NM, Reider I et al. (1994). Basal Miyagi K, Imaizumi T, Nagura H et al. (1992). A case of
ganglia lacunes and parkinsonism. Neuroepidemiology transient parkinsonism due to mesencephalic hemorrhage.
13: 108112. Rinsho Shinkeigaku 32: 743746.
Jellinger KA (2002). Vascular parkinsonismneuropathological Murrow RW, Schweiger GD, Kepes JJ et al. (1990). Parkin-
findings. Acta Neurol Scand 105: 414415. sonism due to a basal ganglia lacunar state: clinicopatho-
Jost WH, Jung G, Schimrigk K (1994). Colonic transit time logic correlation. Neurology 40: 897900.
in non-idiopathic Parkinsons syndrome. Eur Neurol 34: Parkes JD, Marsden CD, Rees JE et al. (1974). Parkinsons
329331. disease, cerebroarteriosclerosis, and senile dementia. Q J
Kaasinen V, Ruottinen HM, Nagren K et al. (2000). Upregu- Med 43: 4961.
lation of putaminal dopamine D2 receptors in early Pau A, Brambilla M, Cossu M et al. (1989). Parkinsonism in
Parkinsons disease: a comparative PET study with the presence of intracranial extracerebral haematomas.
[11C] raclopride and [11C] N-methylspiperone. J Nucl Acta Neurochir (Wien) 96: 159160.
Med 41: 6570. Peralta C, Werner P, Holl B et al. (2004). Parkinsonism fol-
Katzenschlager R, Zijlmans J, Evans A et al. (2004). Olfac- lowing striatal infarcts: incidence in a prospective stroke
tory function distinguishes vascular parkinsonism from unit cohort. J Neural Transm 111: 14731483.
VASCULAR PARKINSONISM 425

Pineda DA, Buritica O, Sanchez JL et al. (2000). Parkinso- Trosch RM, Ransom BR (1990). Levodopa-responsive par-
nian syndromes in Medellin (Colombia). Rev Neurol 31: kinsonism following central herniation due to bilateral
936943. subdural hematomas. Neurology 40: 376377.
Reider-Groswasser I, Bornstein NM, Korczyn AD (1995). Turjanski N, Pentland B, Lees AJ et al. (1997). Parkinsonism
Parkinsonism in patients with lucanar infarcts of the basal associated with acute intracranial hematomas: an [18F]
ganglia. Eur Neurol 35: 4649. dopa positron-emission tomography study. Mov Disord
Sandyk R (1983). Parkinsonism secondary to neurosyphilis. 12: 10351038.
A case report. S Afr Med J 63: 665666. Tzen KY, Lu CS, Yen TC et al. (2001). Differential diagno-
Schwab RS, England AC (1968). Parkinson syndromes due sis of Parkinsons disease and vascular parkinsonism by
to various specific causes. In: PJ Vinken, GW Bruyn (99m)Tc-TRODAT-1. J Nucl Med 42: 408413.
(Eds.), Vol. 6, North-Holland Publishing Co, Amsterdam, Van Zagten M, Lodder J, Kessels F (1998). Gait disorder and
pp. 227247. parkinsonian signs in patients with stroke related to small
Sibon I, Guyot M, Allard M et al. (2004a). Parkinsonism deep infarcts and white matter lesions. Mov Disord 13:
following anterior choroidal artery stroke. Eur J Neurol 8995.
11: 283284. Walter U, Wittstock M, Benecke R et al. (2002). Substantia
Sibon I, Fenelon G, Quinn NP et al. (2004b). Vascular par- nigra echogenicity is normal in non-extrapyramidal cere-
kinsonism. J Neurol 251: 513524. bral disorders but increased in Parkinsons disease.
Silverman CS, Brenner J, Murtagh FR (1993). Hemorrhagic J Neural Transm 109: 191196.
necrosis and vascular injury in carbon monoxide poison- Wichmann T, DeLong MR (2003). Pathophysiology of Par-
ing: MR demonstration. AJNR Am J Neuroradiol 14: kinsons disease: the MPTP primate model of the human
168170. disorder. Ann NY Acad Sci 991: 199213.
Snow B (2003). Objective measures for the progression of Winikates J, Jankovic J (1999). Clinical correlates of vascu-
Parkinsons disease. J Neurol Neurosurg Psychiatry 74: lar parkinsonism. Arch Neurol 56: 98102.
287. Yamada A, Takeuchi H, Miki H (1992). Unilateral abolition
Sunada I, Inoue T, Tamura K et al. (1996). Parkinsonism due of parkinsonian rigidity after subthalamic nucleus hemor-
to chronic subdural hematoma. Neurol Med Chir (Tokyo) rhage. Rinsho Shinkeigaku 32: 887889.
36: 99101. Yamanouchi H, Nagura H (1995). Cerebrovascular parkin-
Taki J, Nakajima K, Hwang EH et al. (2000). Peripheral sonismclinicopathologic study. Rinsho Shinkeigaku
sympathetic dysfunction in patients with Parkinsons 35: 14571458.
disease without autonomic failure is heart selective and Yamanouchi H, Nagura H (1997). Neurological signs and
disease specific. Eur J Nucl Med 27: 566573. frontal white matter lesions in vascular parkinsonism.
Tan EK, Chan LL, Auchus AP (2001). Reversible parkinsonism A clinicopathologic study. Stroke 28: 965969.
in systemic lupus erythematosus. J Neurol Sci 193: 5357. Yoritaka A, Hattori T, Hattori Y, Mori H, Matsuoka S, Shirai
Thobois S, Jahanshahi M, Pinto S et al. (2004). PET and T, Kondo T, Mizuno Y (1997). A 85-year-old woman with
SPECT functional imaging studies in Parkinsonian syn- the onset of progressive gait disturbance at 80 years of the
dromes: from the lesion to its consequences. Neuroimage age. No To Shinkei 49: 379389.
23: 116. Zijlmans JC, Daniel SE, Hughes AJ, Revesz T, Lees AJ (2004).
Thompson PD, Marsden CD (1987). Gait disorder of subcor- Clinicopathological investigation of vascular parkinsonism,
tical arteriosclerotic encephalopathy: Binswangers including clinical criteria for diagnosis. Mov Disord 19:
disease. Mov Disord 2: 18. 630640.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 53

Old age and Parkinsons disease

ALEX RAJPUT AND ALI H. RAJPUT*

University of Saskatchewan, Saskatoon, SK, Canada

53.1. Introduction of vertical gaze (Jenkyn et al., 1985) with aging. These
age-related anatomical and physiological changes lead
There is no uniformly agreed definition of old age. For to a slowed motor function (bradykinesia) and altera-
the purpose of this chapter, we will use the commonly uti- tions in station, posture, gait and postural reflexes, all
lized employment-related retirement age of 65 as the start of which are well-known features of PD. Therefore,
of old age (Desai et al., 1990; Canadian Study of Health normal changes of aging need to be distinguished from
and Aging Working Group, 1994; Moghal et al., 1994, PD in elderly persons. Most PS, including PD, are con-
1995). The proportion of the elderly has been steadily centrated in later age. Thus, the incidence of PS and
rising for several decades in western countries (Rowe, PD rises sharply with advancing age.
1988). Therefore, the frequency of those disorders that The pathological hallmark of PD is a marked loss
are concentrated in old age is progressively increasing. of substantia nigra pigmented neurons and neuronal
The motor manifestations of Parkinsons disease Lewy body inclusions (Jellinger, 1987; Duvoisin and
(PD) include bradykinesia, rigidity, tremor, postural Golbe, 1989; Rajput et al., 1991c). Autopsy studies
abnormality, gait abnormality, impaired postural ref- show that a significant proportion of the elderly with
lexes, extraocular movement abnormality, presence no clinical features of PS have incidental Lewy body
of primitive reflexes and reduced facial expression. inclusions in the brain. The frequency of incidental
Some similar features also seen in the normal elderly. Lewy body inclusions in autopsy brains rises with
The most common variant of Parkinson syndrome age, reaching 13% in the ninth decade (Graybiel
(PS: parkinsonism) is the Lewy body disease also et al., 1990; Fearnley and Lees, 1991; Gibb, 1991;
known as PD or idiopathic PD (Jellinger, 1987; Duvoi- Ross et al., 2004). The incidental Lewy body cases
sin and Golbe, 1989; Rajput et al., 1991c; Bower et al., are believed to represent a preclinical stage of PD
1999; Robinson and Rajput, 2005). The primary focus (Fearnley and Lees, 1991; Gibb, 1991; Ross et al.,
of this chapter will be on aging and PD, with brief 2004) who would develop clinical features of PD with
consideration of the less common variants of PS and time or with an added stress of neuroleptic use (Rajput
of other disorders that may mimic PD. et al., 1982). The elderly have a disproportionately
Normal posture and gait depend on vestibular, large number of other diseases (Rowe, 1988). They
visual, proprioceptive and motor functions and an abil- receive more prescription and non-prescription drugs
ity to adjust to the postural sway. There is a decline in compared to the younger population and have a higher
vestibular (Parker, 1994), visual (Kline et al., 1982; incidence of drug adverse effects, including parkinson-
Matjucha and Katz, 1994), proprioceptive (Drachman ism (Blaschke et al., 1985; Rowe, 1988; Desai et al.,
et al., 1994) and motor functions (Drachman et al., 1990). Other disorders which are common in old age,
1994), as well as reduced muscle mass (Lexell et al., notably stroke and Alzheimers disease, may be asso-
1988) in old age. In addition, there is increased pos- ciated with clinical features of parkinsonism. When
tural sway (Maki et al., 1990; Baloh et al., 1994), pos- all these factors are considered together, the elderly
tural instability (Weiner et al., 1984; Ross et al., 2004), represent a large segment of the population with an
stooped posture (Ross et al., 2004) and reduced range increased risk of developing parkinsonism or PD.

*Correspondence to: Professor Ali H. Rajput, University of Saskatchewan, Royal University Hospital, 103 Hospital Drive,
Saskatoon, SK S7N OW8, Canada. E-mail: ali.rajput@saskatoonhealthregion.ca, Tel: (306) 966-8009, Fax: (306) 966-8030.
428 A. RAJPUT AND A. H. RAJPUT
The onset of PS is very rare before age 30 years Impaired postural reflexes evolve relatively late in
(Rajput et al., 1984a; Marttila, 1987; Bower et al., younger cases and are used to classify the clinical
1999) and the incidence and prevalence rates rise with severity of the disease (Hoehn and Yahr, 1967; Fahn
age. Whereas there were 0.8/105 new cases of PS et al., 1987).
before age 30 years and 26.5/105 new cases per year
between ages 50 and 59 years, the annual incidence
53.2.1. Bradykinesia of old age and parkinsonism
rose to 211.6 between ages 70 and 79 and was 304.8/
105 for those aged 8099 years in one community
The akinesiahypokinesiabradykinesia complex
(Bower et al., 1999). Assuming no competing cause
implies slowed motor function. Akinesia is the inabil-
of death, the risk of developing PS was estimated at
ity to initiate movement, hypokinesia indicates
0.7% by age 60 and 7.5% by age 90 years (Bower
reduced amplitude of movement and bradykinesia im-
et al., 1999).
plies slowed speed of movement (Marsden, 1989). In
The other major consideration is the survival after
clinical practice, these three features are collectively
onset of disease, as that would impact the number of
referred to as bradykinesia. Bradykinesia is the cardi-
PS cases in old age. Levodopa is the most widely used
nal feature of PD and other variants of PS (Webster,
drug for PD in industrialized countries (Global Parkin-
1968; Fahn et al., 1987; Marsden, 1989; Rajput et al.,
sons Disease Survey (GPDS) Steering Committee,
1991c; Rajput, 1994a). The clinical manifestations of
2002). Patients treated with levodopa have a
bradykinesia are similar in all variants of parkinson-
significantly longer survival after onset of illness
ism, though there may be different anatomical sites
(Rajput, 2001) compared to untreated cases and that
of involvement. Slowed motor function is also a part
observed in the past (Hoehn and Yahr, 1967). Higher
of normal aging (Duncan and Wilson, 1989; Drachman
incidence and prolonged survival result in higher
et al., 1994; Bennett et al., 1996; Ross et al., 2004).
prevalence rates in the elderly population.
Distinction between the normal age-related slowing
Bennett et al. (1996) studied the prevalence of par-
and bradykinesia of parkinsonism is therefore impor-
kinsonism in community residents 65 years and older.
tant. Table 53.1 summarizes features that are helpful
The overall prevalence of PS between age 65 and 74
in distinguishing between age-related slowing, PS
years was 14.9%, between 75 and 85 years it was
and some other neurological and focal abnormalities
29.5% and in those over age 85 years the prevalence
which may cause motor slowing.
rate was 52.4%. In another community-based study,
Bradykinesia as a symptom reported by PD patients
Schoenberg et al. (1985) reported that the prevalence
depends on the lifestyle. In general, the motor activ-
rate of PS in those over 75 years was more than seven
ities that require repeated change in direction are
times higher compared to those aged 4060 years.
impaired early. Someone who jogs or walks for exer-
Because of the similarities between normal aging
cise may note a foot dragging when tired, a piano
and PD motor features, the first question the neuro-
player may report being unable to play certain notes
logist must answer is whether the clinical features are
and a writer would find slowed and smaller handwrit-
an indication of old age or of PD.
ing. Slowing of other activities, e.g. walking, easing
gracefully into a chair and difficulty turning in bed
53.2. Normal aging and Parkinsons disease are other common complaints. In clinical practice, bra-
dykinesia is tested by observing the patient as he/she
As noted above, some of the motor manifestations of sits, talks, walks and when asked to perform certain
aging resemble PS. Because there are no readily avail- motor tasks.
able biological markers to distinguish between normal The normal automatic arm-swing with walking,
aging and parkinsonism, the best tool is careful clinical hand gestures during conversation and facial expres-
assessment (Weiner et al., 1984; Jenkyn et al., 1985; sion are all reduced in parkinsonism. Simons et al.
Sudarsky, 1990, 1994; Rajput, 1993c). The presence (2004) studied 19 PD patients and 26 healthy controls
of two of the three cardinal features of bradykinesia, for emotional expression during conversation and
rigidity and resting tremor are needed for a clinical while watching videos and for the ability to use an
diagnosis of PS (Rajput et al., 1991a, 2002; Rajput, emotional facial gesture or imitate facial movements.
1994a; de Rijk et al., 1997). Impaired postural reflexes All these factors were reduced in PD cases compared
are not used as additional evidence for PS diagnosis in to controls (Simons et al., 2004). In a microcomputer-
the elderly (Rajput et al., 1991c; Rajput, 1994a), as based observation, Katsikitis (1988) noted that, when
they are also seen in a large proportion of the normal watching funny cartoons, parkinsonian patients smiled
elderly (Weiner et al., 1984; Ross et al., 2004). less frequently than the controls.
OLD AGE AND PARKINSONS DISEASE 429
Table 53.1
Motor slowing in aging, parkinsonism and some other disorders

Other nervous system


Normal age-related Parkinsonism Focal pathology pathology (central or
slowing bradykinesia emulating slowing peripheral) causing slowing

Symmetry of Symmetrical Often Asymmetrical Frequently asymmetrical


findings asymmetrical
Focal Normal Normal Abnormal Normal
examination
Tone Normal Increased Normal (if pain and May have paratonia in
rigidity joint movement demented cases Spasticity
restriction if corticospinal
discounted). Test at involvement Decreased if
unaffected joint lower motor neuron
helpful to identify pathology
normal tone
Sensory Normal (except vibration As in normal As in normal aging May be impaired depending
function reduction in feet) aging on the nature of lesion
Reflexes Normal symmetrical As in normal As in normal aging Hyperactive with extensor
(ankle jerks may be aging plantar or hypoactive
hypoactive) Plantars depending on site of lesion.
flexor Frequently asymmetrical
Muscle Normal Normal Normal, but patient Reduced or inconsistent
strength unable to exert fully
due to pain or joint
restriction
Rest tremor Absent Usually present Absent Absent
Facial Normal Reduced Normal Normal
expression
Handwriting Normal Micrographia Normal Normal
Speech Normal Low volume Normal Normal

The smooth face of young age becomes wrinkled in In most PD patients, the bradykinesia and rigidity
the elderly. Facial immobility in PD results in a mask- are of comparable severity when the standardized
like face and prolonged facial immobility leads to a assessments are done (Webster, 1968; Fahn et al.,
smoother and younger-looking face. Since the facial 1987). However, the pathophysiology of bradykinesia
wrinkles develop over many years, the reduced facial and rigidity may not be the same in elderly cases.
expression must exist for a long time before parkinson- Levodopa which relieves rigidity may not improve
ism is clinically evident. Depression, which is com- bradykinesia in advanced PD (Marsden, 1989).
mon in PD, adds to the reduced facial expression. Asymmetry of bradykinesia without any other focal
Motor slowing is most pronounced in those activ- or neurological cause is a strong indication of PD.
ities that require repetitive, simultaneous or sequential That should, however, be assessed with more than
movements. Repetitive movements of finger/thumb- one activity. We have observed asymmetrically
tapping, forearm pronation/supination, fist-opening/ reduced arm-swing without any other features of PS
closing and foot-tapping are common clinical maneu- in several otherwise neurologically normal members
vers utilized to assess bradykinesia (Fahn et al., of one family. It is not uncommon for the elderly to
1987). With repeated movements, the amplitude of have a shoulder problem which restricts movements.
the movement declines and the movement may arrest. When there is evidence of localized motor function
The total slowing of an activity is more pronounced slowing, ask the patient if there is pain associated with
than the sum total of the slowing of each component the movement and evaluate the area for any mechani-
of the complex movement (Marsden, 1989). cal or pain-related passive movement restriction at the
430 A. RAJPUT AND A. H. RAJPUT
joint. Detailed assessment of sensory function, motor Cognition and language impairment are more com-
strength, tone and reflexes, as noted in Table 53.1, is mon in the elderly and impact their ability to cooperate
a valuable adjunct to distinguish parkinsonism from for adequate assessment. They may resist passive
age-related slowing and other disorders. The age-related movement proportional to the force applied by the
slowing is typically symmetrical and generalized. examiner. Alternatively, the patient may oppose the
Although bradykinesia due to PD is often asymmetrical, passive movement intermittently, giving it a feeling of
it is more often symmetrical in multiple system atrophy cogwheel rigidity. This is known as paratonia or gegen-
and in progressive supranuclear palsy. halten (Klawans, 1981; Jenkyn et al., 1985; Rajput,
The severity of bradykinesia in PD is reported to 1993c). Such tone abnormality is not reproducible in a
correlate with the degree of striatal dopamine loss predictable fashion (Jenkyn et al., 1985; de la Monte
(Ehringer and Hornykiewicz, 1960b, 1998; Hornykie- et al., 1989; Rajput, 1993c).
wicz and Kish, 1986; Agid et al., 1987, 1989; Agid, Dystonia is characterized by co-contraction of ago-
1991). We investigated the relationship between Uni- nist and antagonist muscles. The force of muscle con-
fied Parkinsons Disease Rating Scale (UPDRS)-based traction is, however, unequal in the opposing muscle
bradykinesia (Fahn et al., 1987) and the striatal dopa- groups. On passive movement, there is greater resis-
mine levels in PD. The striatal dopamine loss and the tance when the part is moved away from the dystonic
severity of bradykinesia revealed a correlation in posture compared to when the movement is attempted
the akinetic-rigid PD cases but not in the tremor-domi- in the same direction as the dystonic posture. Dystonia
nant or the mixed clinical picture cases (unpublished may be associated with intermittent tremor mainly
observations). when the body part is moved away from the dystonic
position. This is known as dystonic tremor.
53.2.2. Tone change in old age, parkinsonism and The increased tone with corticospinal tract lesions
some other disorders is known as spasticity. It is characterized by a velocity
-dependent catch which is followed by a release
The major tone abnormality in PS is rigidity. It is clini- without further resistance, as seen in opening or clos-
cally similar in different variants of PS. Some PS ing a pocket knife. It is, therefore, called clasp-knife
patients may also manifest dystonia. hypertonicity. Although spasticity and rigidity are easy
Tone is defined as involuntary resistance to passive to distinguish in most cases, in cases with mild spasti-
movement. In testing tone, one depends on patient city the distinction from rigidity may be difficult. The
cooperation. The patient is asked to relax: neither to following maneuver may be helpful: Have the patient
assist nor resist the passive movement attempted by lie on his/her back, lift one leg with one hand under
the examiner. In the normal elderly, there is no change the heel and put your other arm under the extended
in tone. Rigidity may manifest as continued smooth knee of the patient. Now suddenly release the heel grip
resistance to passive movement through the entire and allow the leg to flex at the knee, falling towards
range of movement. That is known as lead-pipe rigid- the examining table. In case of spasticity, there will
ity. More often in rigidity, the increased tone is asso- be an initial catch and then the leg will drop to the
ciated with recurring interruptionsa ratchety quality table rapidly. In the case of rigidity, the leg falls down
known as cogwheel rigidity. at a steady speed throughout the downward movement.
When asked to relax, most patients do so and allow An additional distinguishing feature is the change in
satisfactory assessment of tone. The instructions may reflexes. Muscle stretch reflexes are hyperactive in cor-
need to be repeated to ensure patient cooperation dur- ticospinal lesions but there is little, if any, change in PD.
ing the passive movement. Rigidity in PD patients is The plantar response is extensor in corticospinal tract
predictably reproducible and is approximately equal lesions whereas in PD the plantar reflex is flexor or
in the muscles with opposite mode of action (Findley equivocal. In some PS patients, there is spontaneous
and Koller, 1995). Cogwheel rigidity is usually or ambulation-related dorsiflexion of the big toe. This
observed in patients who have hypertonicity as well dystonic posture is called striatal toe (Duvoisin,
as tremor. However, some patients with no visible tre- 1990). When the plantar reflex is performed, the big
mor may also manifest cogwheel rigidity. The patient toe does not extend further but may flex.
may try to cooperate and actively move the body part
in the same direction as the examiner moves it. In such 53.2.3. Tone testing and tremor disorders
case, the passive movement should be performed irre-
gularly so that the patient cannot predict and oppose The presence of medium- or large-amplitude tremor
or facilitate the movement. Distracting the patient by may produce rhythmic interruption of the passive move-
asking a question may be helpful in the evaluation. ment known as cogwheeling phenomenon (Findley
OLD AGE AND PARKINSONS DISEASE 431
et al., 1981; Findley and Koller, 1995). In such cases who are likely to have degenerative cervical spine
increased tone on reinforcement is known as Froments changes. If there is still a doubt about the nature of
phenomenon. During examination, when the ratchety the cogwheeling at a joint, e.g. at wrist, the tone
feeling first emerges, stop further movement and gently should be tested at another joint where there is no tre-
hold the body part still. In Froments phenomenon, the mor, such as the shoulder.
rhythmic movement continues. If, on the other hand, Table 53.2 summarizes the tone change in the normal
there is cogwheel rigidity related to PD, there would elderly, parkinsonism and selected disorders.
be no continuation of the rhythmic movement. In
patients with head tremor, when the passive move- 53.2.4. Station, posture and postural reflexes in old
ment is performed in the direction of the tremor, there age and Parkinsons disease
may be cogwheeling and on reinforcement Froments
phenomenon. Passive movement in the direction The manner or attitude of standing is known as station
where there is no tremor will help determine the tone and the position of the whole body and its different
more accurately. In PD-related neck rigidity, there parts indicates the posture. Visual, vestibular and
will be resistance in all directions of neck movements proprioceptive functions, which are critical for main-
lateral flexion and turning the head from side to taining normal posture, all decline with normal aging
side. Special attention should be paid to the elderly (Kline et al., 1982; Drachman et al., 1994; Matjucha

Table 53.2
Tone changes in elderly Parkinsons disease and selected conditions

Cogwheeling phe-
Parkinsons Spasticity Paratonia nomenon (Froments
Normal elderly disease rigidity (mild) (gegenhalten) phenomenon)

Characteristic Equal and normal Resistance Increased tone Irregular, Rhythmic


resistance in sustained and Velocity- intermittently Interruption of
all directions equal in opposite dependent increased tone or passive movement
of movement directions of catch progressively coinciding with
movement. clasp-knife greater tremor. Rhythmic
Reproducible resistance on resistance to
attempted passive movement
movement only seen on
Unpredictable reinforcement in
tremor-producing
conditions
Froments
phenomenon
Symmetry Symmetrical Usually Frequently Usually May be symmetrical
asymmetrical asymmetrical symmetrical or asymmetrical
depending on
tremor location
Reflexes Normal Reduced Normal. May have Exaggerated Usually normal Normal
at ankles striatal toe and extensor
Plantars flexor plantar
response
Tremor Absent Frequently part of Absent Absent Prominent feature
Parkinsons
disease
Reinforcement- No significant Usually May increase Unpredictable Rhythmic
related tone change. asymmetrical slightly change symmetrical
increase Minimal, if increase increase
any, and is (Froments
symmetrical phenomenon)
432 A. RAJPUT AND A. H. RAJPUT
and Katz, 1994; Parker, 1994). There is normally repeated in the backward direction (Weiner et al.,
swaying of the body while in the standing position; 1984; Fahn et al., 1987; Sudarsky, 1990). This is
this increases with advancing age (Maki et al., 1990; known as the pull test. If the patient takes no or one
Baloh et al., 1994; Bennett et al., 1996; Du Pasquier step, that is considered normal. When there are two
et al., 2003; Fransson et al., 2004). Dizziness is the to three steps it is considered as slight impairment
subjective sensation of instability. By the age of 65 of postural reflexes. Four or more steps on pull or
years, 30%, and by age 80 years, 66% of the general when the patient would fall if not prevented is defi-
population would have experienced dizziness at some nitely abnormal (Fahn et al., 1987). The global clinical
time (Luxon, 1984). This subjective feeling adds to severity of PD is measured using the pull test as a
postural instability in older individuals. major parameter (Hoehn and Yahr, 1967; Fahn et al.,
Normal elderly people have a slightly wide base 1987).
when standing and walking (Drachman et al., 1994). Impaired postural reflexes are part of normal aging
The body becomes flexed at the neck and trunk (Ross (Tinetti et al., 1988; Rajput, 1993c; Ross et al., 2004).
et al., 2004) but the knees and elbows remain straight Postural reflexes were impaired in 43% between age
in old age. By contrast, in PD patients, there is flex- 60 and 69 years and in 70% of individuals between
ion at the neck, trunk, hips, knees and elbows. Those ages 80 and 89 in persons who had no neurological
changes are more pronounced in multiple system or other identifiable cause (Weiner et al., 1984). Pro-
atrophy. In progressive supranuclear palsy, the posture gressive decline of postural reflexes was reported in
is erect, with the neck in a normal or even extended a longitudinal study of normal elderly people (Wilson
position. When asked to stand erect, advanced PD et al., 2002). Thus, the postural instability as tested
patients have difficulty maintaining the erect posture in the clinical setting is also a part of the normal aging
for several seconds, whereas the normal elderly can process. It can not be utilized therefore as a require-
do that easily. ment for the diagnosis of PD in the elderly. However
The ability to regain balance after spontaneous it can be used as an adjunct when the other cardinal
sway or when the body is actively perturbed from features of PD bradykinesia, rigidity and tremor
standing position depends on the integrity of the pos- are evident, especially if those abnormalities are asy-
tural reflexes. Maki et al. (1990) noted that both spon- mmetrical (Tinetti et al., 1988; Rajput et al., 1991c;
taneous and induced sway of balance are exaggerated Rajput, 1993a, b, c). Although the PD-related loss of
in old age, regardless of whether the eyes are open or postural reflexes may improve with levodopa, the
closed. Increased anteroposterior displacement velo- age-related abnormality does not benefit from medical
city in the elderly has been reported in other studies therapy.
(Du Pasquier et al., 2003; Fransson et al., 2004). The
most pronounced velocity change is seen in those 53.2.5. Gait abnormality in old age and Parkinsons
who have a fear of falling. With vibratory perturbation disease
applied to the gastrocnemius muscle, the elderly used
higher-frequency motion and complex dynamics to In normal elderly people, the gait is slightly wide-
adjust compared to younger persons (Fransson et al., based, slow and the strides are shorter compared to
2004). The elderly also have a reduced capability to younger individuals but the heel strike and the arm-
use visual information to adjust to perturbation. In gen- swing are normal (Rajput, 1993c; Drachman et al.,
eral, the elderly have more difficulty in compensating 1994). In normal elderly people, the stride velocity
for balance perturbation (Fransson et al., 2004). declines by 1020% by age 80 years (Sudarsky,
In the clinical setting, postural stability is tested as 1990; Winter et al., 1990) and they adopt a more con-
follows: The patient is asked to stand with eyes open servative gait to reduce the risk of falling (Menz et al.,
and feet approximately shoulderwidth apart. He/she 2003). Individuals who have an exceptionally slow or
is asked to resist a sudden pull by the examiner. The abnormal pattern of ambulation are classified as hav-
patient is advised to take one step if necessary to ing a gait disorder. It is estimated that 15% of persons
regain balance and is assured that the examiner will 60 years and older have a gait abnormality (Sudarsky,
not allow him/her to fall. Depending on the size and 1990). Sudarsky and Ronthal (1983) assessed 50 indi-
agility of the patient, there will be a different degree viduals who had a mean 1-year duration of gait diffi-
of force required to displace. The force of the pull culty. They found that 10% of those cases had other
should be such as would reasonably displace the per- features of parkinsonism and improved on levodopa.
son so the capability to regain balance can be assessed. In approximately 15% of elderly gait disorder cases,
Initially, the examiner applies a pull in the forward no cause is discovered despite extensive investiga-
direction. Following that, the same maneuver is tions (Sudarsky and Ronthal, 1983; Sudarsky, 1990).
OLD AGE AND PARKINSONS DISEASE 433
Such cases are classified as having idiopathic patients. However, freezing of gait as an initial mani-
(Sudarsky and Ronthal, 1983) or senile gait disorder festation of PD is not different in young- and old-
(Koller et al., 1983). Senile gait is characterized by age-onset cases (Bloem et al., 2004). On the other
stooped posture, broad base, reduced arm-swing, stiff hand, postural instability and gait disturbance with
turns and a tendency to fall (Koller et al., 1983). PD onset are more common in old age (Bloem et al.,
Gait and balance abnormalities with no identifiable 2004).
neurological disease were evaluated prospectively in In the parkinsonian variant of multiple system atro-
70 healthy ambulatory elderly subjects (mean 79 phy, the gait abnormality is similar to PD, but it mani-
years, range 7488) (Whitman et al., 2001). Brain fests early and the decline is more rapid than in PD
magnetic resonance imaging (MRI) studies were per- (Rajput et al., 1972, 1993a). In progressive supranuclear
formed at baseline and 4 years later. Those who devel- palsy cases, the posture is erect and the gait is character-
oped gait and balance problems had a significantly ized by a high-stepping and robotic quality. Progres-
greater mean increase in volume of T2-weighted sive supranuclear palsy cases have predominantly
white-matter hyperintensities on brain MRI (Whitman axial akinetic-rigid features and supranuclear ophthal-
et al., 2001). moplegia. Nearly one-half of the autopsy-confirmed
PD is the most common movement disorder result- cases were reported without ophthalmoplegia in one
ing in gait abnormality in the elderly. The gait is char- study (Birdi et al., 2002). Most progressive supra-
acterized by flexed posture, narrow base, slow and nuclear palsy cases show no significant improvement
short shuffling steps, slow multistep turning, loss of on levodopa (Birdi et al., 2002).
heel strike, reduced toe elevation, reversal of ankle
flexion/extension movement, reduced movement at 53.2.6. Gait apraxia
the knee joints, loss of dynamic vertical force and loss
of backward-directed sheer force resulting in a ten- In gait apraxia cases, the functional abnormality is
dency to propulsion (Forssberg et al., 1984). A Parkin- restricted to walking and there is no motor weakness,
son patient may break into an uncontrollable forward sensory loss or cerebellar dysfunction in the lower
run, known as festination. Because of the impaired limbs to account for the gait difficulty. Typically,
postural reflexes, any perturbation may result in invo- these patients have an erect posture, slightly broad
luntarily walking forward (propulsion) or walking base, difficulty initiating gait, reduced cadence (steps
backward (retropulsion). per minute) and short shuffling and hesitating steps,
Freezing of gait may be evident as start hesitation as if glued to the floor (Estanol, 1981; Fisher,
on approaching an obstacle or on attempts to change 1982; Sudarsky and Ronthal, 1983; Forssberg et al.,
direction ambulatory efforts needed to overcome a 1984; Sudarsky, 1990). Unlike the PD cases, the gait
block (Bloem et al., 2004). Abnormal gait, freezing apraxia patients do not improve with visual or auditory
of gait and impaired postural reflexes are all major cues and are unable to mimic a normal gait (Forssberg
problems at advanced stages of PD (Hoehn and Yahr, et al., 1984; Suteerawattananon et al., 2004). There is a
1967; Fahn et al., 1987; Bloem et al., 2004) and lead marked dissociation between the leg functions in the
to significant functional handicap and falls. One study supine and sitting positions compared to that during
noted that, compared to healthy controls, the relative walking. Gait apraxia patients are able to use the lower
risk of recurrent falls in PD cases over a period of 6 limbs for activities such as writing on the floor with a
months was nine times greater (Bloem et al., 2004). foot while in the sitting position, kicking an imaginary
In moderately advanced PD patients, the gait may ball and emulating bicycle riding in the supine posi-
improve with visual guided patterns (Forssberg et al., tion. However, in the weight-bearing position, the
1984; Suteerawattananon et al., 2004) and with audi- execution of the lower-limb motor activity required
tory cues (Suteerawattananon et al., 2004). Prior to for walking is markedly impaired. By contrast, in mild
the advent of levodopa therapy, no drug reversed the to moderate PD cases, the impairment of lower-limb
impaired postural reflexes and gait abnormality in PD function during non-weight-bearing and the weight-
significantly. On modern drugs, some patients may bearing activities is similar. Della Sala et al. (2002)
regain those functions. Freezing of gait is difficult to reported a case of gait apraxia due to a stroke affecting
treat. Response to treatment is less than satisfactory bilateral supplementary motor areas. In normal-
during advanced stages of PD, but some improvement pressure hydrocephalus, the exact site pathology is
may be seen on dopaminergic drugs and on selegiline not known (Fisher, 1982; Sudarsky and Simon,
(Bloem et al., 2004). Freezing of gait during the off 1987). The normal-pressure hydrocephalus patients
period may improve on levodopa (Schaafsma et al., often manifest dementia, frontal-release signs and
2003). Freezing of gait is more common in older PD bladder incontinence (Estanol, 1981). In some of those
434 A. RAJPUT AND A. H. RAJPUT
cases, the gait abnormality may be the only manifesta- gaze increased with age. The frequencies for upgaze
tion (Fisher, 1982; Sudarsky and Simon, 1987). Rare and downgaze impairments, respectively, were 6%
patients with gait apraxia may have extensor plantar and 8% for those aged 6569 years, increasing to
responses. Resting tremor, which is a common feature 29% and 34% for those 80 years and older (Jenkyn
of PD, is not seen with gait apraxia. Limb bradykinesia et al., 1985). Gaze impairment is characteristic of
and rigidity, the other major features of PD, are not progressive supranuclear palsy (Steele et al., 1964;
seen in either the upper or the lower limbs when Jackson et al., 1983; Birdi et al., 2002). It is also seen
assessed in the sitting or supine position. The gait in rare multiple system atrophy patients (Jankovic
apraxia patients do not improve on levodopa. et al., 1993) and we have observed it in an autopsy
Table 53.3 summarizes gait in the normal elderly, PD confirmed PD cases.
and gait apraxia patients. Oculogyric crisis was a feature of postencephalitic
parkinsonism but is also seen in some cases of drug-
53.2.7. Eye movements in the elderly and induced parkinsonism (DIP) and in dopa-responsive
in Parkinsons disease dystonia (Rajput, 1973).

Voluntary and pursuit upward gaze movement of 5 53.2.8. Primitive reflexes in the elderly and
mm or less and downward gaze deviation of 7 mm in Parkinsons disease
or less are considered abnormal (Jenkyn et al., 1985).
In a study of more than 2000 normal volunteers 50 Grasp and other primitive reflexes seen in childhood
93 years old, impairment of upward and downward are suppressed during development but may re-emerge

Table 53.3
Posture and gait in normal elderly, Parkinsons disease and gait apraxia

Clinical features Normal elderly Parkinsons disease Gait apraxia

Base (distance between feet) Slightly wider than at Narrow Broad


younger age
Symmetry of abnormality Symmetrical Usually asymmetrical Symmetrical
Functions in the involved Normal and symmetrical Impaired regardless of Impaired only when
lower limb weight-bearing or not weight-bearing but
(no dissociation) normal when not weight-
bearing (dissociation
pronounced and early)
Upper-limb motor function Normal Impaired on the involved side Normal
Arm-swing Normal Reduced on involved side Normal
Posture Erect or neck and trunk Generalized flexion neck, As in normal elderly
flexion trunk, hip, knee, elbow
Postural reflexes May be normal or impaired Impaired in moderately Impaired early
advanced disease
Foot-tapping in sitting or lying Normal Affected side slow and As in normal elderly
position progressively slower
Gait abnormality Uncommon increases Late manifestation usually Major problem early
with age manifestation
Visual and auditory cue No change in gait Improve gait No improvement in gait
Tremor Absent Frequently seen typically As in normal elderly
resting
Dementia Absent In about one-third of cases Common but not invariable
Reflexes Normal (may be As in normal (may have May be brisk may have
reduced at ankles) striatal toe) Babinski sign
Grasp reflex Normal Usually negative Usually positive
Bladder function Normal Normal or hypertonic or Incontinence common
hypotonic bladder
Levodopa response None Improvement None
OLD AGE AND PARKINSONS DISEASE 435
in old age (Bennett et al., 1996; Schott and Rossor, of normal aging and is a major distinguishing feature
2003). Typically, PD patients have a reduced rate of between PD and normal elderly.
eye-blinking, resulting in a reptilian stare. Normally, Striatal dopaminergic denervation has been reported
the glabellar reflex habituates after 24 taps. Sustained in normal aging. Positron emission tomography (PET)
glabellar reflex was observed in 10% of normal volun- studies revealed reduced dopaminergic functions in
teers ages 6569 years and in 37% of those aged 80 grandparents compared to their grandchildren (Cordes
years and older (Jenkyn et al., 1985). In PD, this reflex et al., 1994). Reduced striatal dopamine levels are
may persist and in rare cases it may produce blephar- reported in normal elderly autopsy brains (Kish et al.,
ospasm. Spontaneous blepharospasm is rare in PD 1992). However, the pattern of the brain regional dopa-
but is more common in progressive supranuclear palsy mine loss is different in normal aging (Kish et al., 1992)
cases. Repeated blinking after a tap at the bridge of the and PD (Kish et al., 1988). The rate of dopaminergic
nose (Myersons sign) is common in PD. A positive denervation was estimated to be twice as high in PD
snout reflex correlates with advancing age (Koller compared to normal aging (Scherman et al., 1989). It
et al., 1982). As the primitive reflexes are common was concluded that dopamine deficiency does not play
in the normal elderly, their presence cannot be used a role in normal aging (Scherman et al., 1989). Unlike
as evidence of PD. PD, age-related slowing does not improve on levodopa.
It is therefore concluded that the substantia nigra loss
53.3. Pathophysiological consideration and the associated dopamine loss characteristic of PD
of motor abnormalities in the elderly do not account for the Parkinson-like features of normal
aging.
The presence of some parkinson-like motor features in
the elderly suggests that PD and old age may share 53.4. Tremor disorders in old age
pathophysiological mechanisms. The most consistent
pathology in PD is a marked loss of substantia nigra pig- Tremor is the most common movement disorder in
mented neurons and Lewy body inclusions (Jellinger, human adults (Jankovic and Fahn, 1980; Findley and
1987; Gibb and Lees, 1988b; Duvoisin and Golbe, Gresty, 1981; Rautakorpi et al., 1982a, b; Louis
1989; Gibb, 1991; Rajput et al., 1991c; Robinson and et al., 1995). The two most common tremor disorders
Rajput, 2005). The characteristic biochemical finding are PD and essential tremor (ET) (Haerer et al.,
in PD is a marked striatal dopamine loss (Ehringer and 1982; Rautakorpi et al., 1982a; Moghal et al., 1994;
Hornykiewicz, 1960a; Rajput, 2002) and an uneven Salemi et al., 1994; Mayeux et al., 1995; Louis et al.,
subregional pattern of dopamine decline (Kish et al., 1996). Both PD and ET are concentrated in old age
1988). Improvement of PD motor symptoms is almost (Haerer et al., 1982; Rautakorpi et al., 1982b; Rajput
universal in those patients who can tolerate an adequate et al., 1984a, b; Schoenberg et al., 1985; Bharucha
dose of levodopa (Birkmayer and Hornykiewicz, 1961, et al., 1988; Bergareche et al., 2001). There is no lit-
1998; Cotzias et al., 1967; Rajput et al., 1990c). erature evidence that postural/kinetic tremor is part
Several studies have compared the pathological and of the normal aging process and rest tremor is never
biochemical findings of PD with the normal elderly a part of normal aging. There is clinical overlap
population who may manifest some parkinsonian-like between PD and ET-associated tremors. The character-
features. One study (Ross et al., 2004) observed that istic tremor of PD is at rest, but these cases may also
the elderly who had a stooped posture, postural have postural/kinetic tremor. Rest tremor is not a fea-
instability or undue motor slowing had significantly ture of normal aging (Rajput, 1994a; Bennett et al.,
reduced neuronal cell counts in the dorsolateral quad- 1996; Ross et al., 2004), although it is seen in nearly
rant of the substantia nigra pars compacta. The counts every PD case (Rajput et al., 1990a, 1991a), and in
were lowest when all three signs were present (Ross some ET patients (Rajput et al., 2004b).
et al., 2004). A detailed study (Fearnley and Lees, The elderly more often use medications that enhance
1991) revealed that the regional pattern of substantia the physiological tremor, making it clinically evident.
nigra loss in normal aging (dorsal tier) is different from This makes it difficult to distinguish drug-induced tre-
PD (ventral tier). They concluded that age-related loss mor from pathological tremor, including ET. Medica-
of dorsal tier substantia nigra pars compacta neurons is tions that may cause or worsen underlying tremor
not important in the pathogenesis of PD (Fearnley and include tricyclics, monoamine oxidase inhibitors, val-
Lees, 1991). Resting tremor has not been observed in proic acid, corticosteroids, calcium channel blockers,
elderly individuals without clinical PD, nor in cases amiodarone, sympathomimetics and rugs for asthma,
with incidental Lewy body inclusions only (Ross et al., including theophylline (Desai et al., 1990; Koller
2004). This indicates that resting tremor is not a feature et al., 1994; Kelly et al., 1995; LeWitt, 1995).
436 A. RAJPUT AND A. H. RAJPUT
53.5.1. Parkinsonian tremor et al., 1994; Salemi et al., 1994). In a population study
using door-to-door interviews and neurological exams
The characteristic parkinsonian tremor is resting tre- in Turkey, 4% of those aged 40 years and older had
mor, i.e. when the body part is fully supported against ET (Dogu et al., 2003). In a Finnish study, 5.6% of
gravity to prevent muscle activity. The postural and the population over age 40 years had ET (Rautakorpi
kinetic tremor that is characteristic of ET is also seen et al., 1982b) whereas 13% of those between the ages
in a significant proportion of PD patients (Jankovic of 70 and 79 years had ET. We found ET prevalence
et al., 1995, 1999). Tremor is the most common initial rates of 14% in the community (Moghal et al., 1994)
symptom reported by PD patients (Hoehn and Yahr, and 10% in the chronic care institution population
1967; Martin et al., 1973; Gibb and Lees, 1988a; (Moghal et al., 1995) aged 65 years and older. Life
Rajput et al., 1991a). The upper-limb tremor onset expectancy in ET is normal (Rajput et al., 1984b).
was noted 13 times more frequently compared to the The tremor frequency in ET changes with time.
lower-limb rest tremor onset in an autopsy study of The tremor in young ET cases has a higher frequency
PD (Rajput et al., 1993a). James Parkinson was so (810 Hz) than PD tremor (46 Hz). ET tremor fre-
impressed with the tremor in PD that he called the quency deceases by 0.060.08 Hz/year (Elble, 2000).
disorder shaking palsy. The PD tremor is usually Therefore, with time, ET tremor frequency may
intermittent at the start and appears under stress. Pro- become indistinguishable from PD. The tremor ampli-
gressively lower-level stress triggers tremor and even- tude progressively increases in ET (Stiles, 1976; Elble,
tually the tremor may become persistent (Hallett, 1986; Calzetti et al., 1987) and becomes more wide-
1986). We observed 1 patient who had a 33-year his- spread throughout the body (Rajput et al., 2004b).
tory of intermittent upper-limb tremor which only Classical ET is kinetic and/or postural (Louis, 2001),
appeared under stress before the other classical fea- but nearly one-third of the autopsy-verified ET
tures of PD appeared. Putting the patient under stress patients developed resting tremor in the upper or lower
is helpful in establishing the presence of rest tremor. limbs, without bradykinesia or rigidity (Rajput et al.,
It can be done by having the patient perform one or 2004b). In the past, such cases were classified as
multiple tasks simultaneously. While the patient is senile tremor (Koller et al., 1994; Kelly et al.,
lying on the examining table, ask him/her to squeeze 1995). Autopsy studies revealed similar brain histol-
your finger with one hand, close the eyes tightly while ogy in classical ET and those with additional resting
you try to open them and have the patient count back- tremor (Rajput et al., 2004b). Therefore, the term
wards from 100 by 7s. This maneuver would bring out senile tremor is no longer justified.
resting tremor in the opposite upper limb and both Coarse tremor can produce cogwheeling and
lower limbs. Upper-limb resting tremor on both sides Froments phenomenon, which may be misinterpreted
can be simultaneously evaluated as follows: in the as a sign of parkinsonism. Clinical distinction between
supine position, the patient is asked to close his/her that and the rigidity of PS has been discussed above.
eyes tightly, count backwards from 100 by 7s and In a large series of pathologically verified ET
forcefully dorsiflex both ankles against resistance patients, 15% subsequently developed dopa-responsive
(with the examiner opposing the dorsiflexion). parkinsonism (Rajput et al., 2004b): 5% had Lewy
In most patients, the parkinsonian resting tremor pro- body disease and 10% progressive supranuclear palsy
duces limited or no functional disability as it improves that was undiagnosed during life. Making a distinction
with activity. After reaching the peak severity (amp- between ET cases that develop resting tremor alone
litude and persistence), the tremor may become less pro- and those with additional PS can be challenging. If a
nounced or cease altogether (Stern, 1987; Birdi et al., known ET patient develops unilateral resting tremor,
2002). When tremor is only present in the resting rigidity and bradykinesia, a second diagnosis of PS is
position but in no other situation, it is a sign of PS justified (Rajput et al., 1991a, b, 1993b, 2004b).
(Rajput, 1994b; Bennett et al., 1996; Rajput et al., Most ET patients developing resting tremor in old
2004b; Rajput and Rajput, 2004; Ross et al., 2004). age. However, when the ET first manifests in child-
hood, such evolution may occur at a younger age, indi-
53.5.2. Essential tremor cating that rest tremor evolution is related to the
duration of the ET. We have observed an accelerated
ET can manifest at any age, from childhood to old age emergence of rest tremor in ET patients when both
(Haerer et al., 1982; Rajput et al., 1984b; Koller et al., parents had ET, suggesting that a higher degree of
1994). The incidence and the prevalence rates of ET genetic abnormality increases the risk of rest tremor.
increase markedly in old age (Haerer et al., 1982; Rau- The beneficial effect of alcohol on tremor is not
takorpi et al., 1982a, b; Rajput et al., 1984b; Bain specific to distinguish ET from parkinsonian tremor
OLD AGE AND PARKINSONS DISEASE 437
or other causes of action tremor, as each may or may The diagnosis of PD in the elderly needs careful
not improve with alcohol (Rajput et al., 1975; Koller consideration of the age-related motor changes which
and Biary, 1984). may be mistaken for PD and vice versa, as discussed
The available literature indicates that ET and PD above. Community-based surveys indicate that
are two different disorders and the risk of developing between 35 and 41% of parkinsonian cases (mostly
PD in ET cases is comparable to that in the general the elderly) remain undiagnosed (Schoenberg et al.,
population (Rajput et al., 2004b). 1985; Morgante et al., 1992). In a survey of commu-
Since there are no biological markers specific for ET nity residents 65 years and older, we observed that
or PD, in the event of clinical uncertainty, a therapeutic 50% of parkinsonian cases were undiagnosed (Moghal
trial on levodopa for up to 8 weeks is justified. Whereas et al., 1994). The undiagnosed patients are usually
PD patients improve, there is no benefit to ET. those who do not have prominent tremor or those in
whom the tremor is erroneously interpreted as a man-
53.6. Parkinsons disease in old age ifestation of aging. A patient who is not diagnosed
remains untreated and, therefore, will suffer unneces-
Epidemiology, major clinical features, non-motor sarily. In one case, a 60-year-old mother of seven chil-
manifestations of PD, other parkinsonian syndromes dren (including one nurse) noted that she could not
and details of treatment are covered in the appropriate look after her household. The children concluded that
chapters of this volume. We will focus on those fea- she was getting old and was, therefore, placed in a
tures that have special significance for PD in old age. private care-home where she was expected to go to
PD in elderly patients can be divided into two main the dining area to eat. However, she could not do that
categories: (1) those with an early age at onset and sur- and her condition deteriorated. At that point, she was
viving to old age; and (2) patients in whom the PD hospitalized. Neurological exam revealed stage 4
first manifests in old age. (Hoehn and Yahr, 1967; Fahn et al., 1987) akinetic-
When the disease begins at a younger age, nearly all rigid PD which improved on levodopa. Another exam-
patients in the industrialized countries would have ple is that of an elderly man who had been in a nursing
received levodopa (Global Parkinsons Disease Survey home for several years. With an impending influenza
(GPDS) Steering Committee, 2002) before reaching old season, he was prescribed amantadine. The attending
age. Most of those patients are likely to have motor physician was surprised to see that the patient improved
response fluctuations and/or dyskinesia (Rajput et al., remarkably to the point that he could be discharged
2002), freezing of gait (Bloem et al., 2004) and a reduced from the nursing home. Since the diagnosis of PD is
therapeutic benefit (Rajput et al., 1984c, 2002). Such based on clinical observations alone, a trial of levodopa
patients are most likely to have been previously treated is justified when in doubt.
with dopamine agonists and the other currently available Tremor is the most common first manifestation of
agents. The treatment options in such cases are limited. PD, ranging from 41% (Gibb and Lees, 1988a) to
Levodopa and adjunct catecholamine-O-methyltransfer- 70.5% (Hoehn and Yahr, 1967) in different studies. In
ase inhibitors, entacapone or tolcapone (where available) autopsy-verified cases, we noted that tremor alone was
are probably the safest options for this group of patients. the first manifestation in 49% of PS cases (Rajput
If a patient is well controlled and has stage 2 (Hoehn and et al., 1993a). The first symptom was upper-limb tremor
Yahr, 1967; Fahn et al., 1987) disability, there is no need alone in 41% and lower-limb tremor alone in 3% of PS
to change the drugs. cases. Tremor in conjunction with other parkinsonian
motor features was the first manifestation in 53% of
53.6.1. Parkinsons disease beginning in old age PS cases (Rajput et al., 1993a). Postural instability and
gait difficulty (PIGD) at onset are reported to be more
In one community study (Bower et al., 1999), the inci- common among the elderly (Zetusky et al., 1985; Jan-
dence of PS rose sharply with age. In the same commu- kovic et al., 1990). Whereas 56% of the tremor-onset
nity (Rocca et al., 2001), the age-specific incidence was parkinsonian cases had only Lewy body PD pathology,
unchanged over several decades but there was an 27% of the PIGD cases had similar pathology (Rajput
increased incidence of DIP in those aged 70 years and et al., 1993a). Clinical studies (Zetusky et al., 1985;
older (Rocca et al., 2001). In a study of 6584-year-old Jankovic et al., 1990) and clinicopathological studies
population, the annual incidence of PS was 529.7/105 (Rajput et al., 1993a) indicate that the PIGD onset cases
whereas it was 326.3/105 for clinically diagnosed PD have a less favorable prognosis. Such cases are more
(Baldereschi et al., 2000). Thus the incidence of PD con- likely to suffer from other variants of PS than PD
tinues to rise with advancing age (Mayeux et al., 1995; (Rajput et al., 1993a), which accounts for some unfa-
Bower et al., 1999; Rocca et al., 2001). vorable outcomes. Since tremor is not a feature of
438 A. RAJPUT AND A. H. RAJPUT
normal aging, new-onset tremor should arouse the suspi- Jankovic et al., 1990). Late-onset age by itself does
cion of PD. Rest tremor is the most reliable distinguish- not unduly shorten survival when the age and sex are
ing feature between normal aging and PS (Rajput et al., taken into account (Rajput et al., 1997). The accelerated
1991a, 1993a; Brooks et al., 1992; Rajput, 1994a; Kelly disability in the old-age-onset PD cases may be due in
et al., 1995). Isolated lower-limb rest tremor onset has part to aging or concurrent senile gait (Koller et al.,
even higher diagnostic value in PD (Rajput et al., 1983) or gait apraxia (Sudarsky and Simon, 1987).
1990b, 1993a). At any given time, approximately one-third of PD
The most common variant of PS is the Lewy body cases have dementia (Rajput and Rozdilsky, 1975;
disease (Rajput et al., 1984a, 1991c; Jellinger, 1987; Marttila and Rinne, 1976; Rajput et al., 1987, 1997;
Duvoisin and Golbe, 1989; Gibb, 1991; Hughes Mayeux et al., 1988; Rajput, 1992; Uitti et al., 1993,
et al., 1992), also known as idiopathic PD. There are 1996) and over 5 years new cases of dementia
other uncommon variants, such as neurofibrillary tan- emerged 35 times more commonly in PD compared
gle pathology, which is focused on the substantia nigra to age-matched controls (Mindham et al., 1982; Rajput
(Rajput et al., 1989). Clinical distinction between PD et al., 1987; Mayeux et al., 1988; Marder et al., 1991;
and other variants of PS is not always possible (Rajput Rajput, 1992; Aarsland et al., 2003). When compared
et al., 1991c; Hughes et al., 2002). However, parkin- with the early-onset cases, dementia was reported to
sonism, when the pathology is focused on the substan- be nearly 10 times more common in the old-age-onset
tia nigra, can be clinically recognized accurately in PD cases (Hietanen and Teravainen, 1988; Mayeux
85% of cases (Rajput et al., 1991c). The multiple sys- et al., 1988, 1990; Hobson and Meara, 2004). Com-
tem atrophy Parkinson subtype (Rajput et al., 1972, pared to the non-demented cases, demented PD cases
1991c; Quinn, 1989; Rajput, 1994a, b) has a younger have a shortened survival (Uitti et al., 1996; Rajput
age at onset, but in rare autopsy cases we have et al., 1997) compared to non-demented cases. (See
observed it to begin in old age. The most prominent Ch. 18 for further discussion of dementia in PD.)
clinical features are symmetrical bradykinesia and Dyskinesia is a common adverse effect of levodopa
rigidity but very little tremor. The presence of early therapy. It has been suggested that the incidence of
autonomic dysfunction and corticospinal tract or cere- dyskinesia is different among young- and old-age-
bellar dysfunction is helpful in distinguishing multiple onset cases. Kostic et al. (1991) compared 25 PD cases
system atrophy from PD. The progression of disability with onset before age 40 years (mean 33.5 years) with
in the multiple system atrophy cases is more rapid and 25 PD patients who had onset at a later age (mean 55.8
the response to levodopa is less favorable than in PD years). After 3 years of levodopa therapy there was a
(Quinn, 1989; Rajput et al., 1990c, 1991c, 1993a). Pro- significantly higher (72% versus 28%) incidence of dys-
gressive supranuclear palsy, on the other hand, has kinesia in the early-onset cases. Gibb and Lees (1988a)
similar age of onset as PD. The motor manifestations compared PD patients with mean onset at 38 years
are symmetrical with predominant axial involvement. with those at mean onset at 73 years. They detected no
The posture is erect, tremor is not a prominent feature difference in the duration of levodopa exposure before
(Birdi et al., 2002) and supranuclear ophthalmoplegia dyskinesia onset. We have observed levodopa-induced
is a characteristic manifestation. dyskinesia emerging early and on a small dose in some
Levodopa is the drug of first choice in PD cases old-age-onset cases. The age-related difference with
with onset at 75 years or older. In the 6575-year respect to levodopa-induced dyskinesia was not evident
age group, depending on health status, one may use in our PD autopsy series (Rajput et al., 2002).
amantadine or a dopamine agonist initially. The dyskinesia and wearing-off phenomena repre-
Though selegiline is generally well tolerated, it has sent opposite ends of the spectrum for motor response
only a mild symptomatic benefit and we do not use it complications. Dyskinesia is most common at the peak
often. Anticholinergics frequently cause adverse plasma level of levodopa, whereas the wearing off
effects and we avoid using them in the elderly. coincides with the lowest plasma level. As expected,
The response to levodopa therapy is most favorable dopamine metabolism in the autopsy brains was found
in PD. Response to levodopa treatment is not diagnostic to be accelerated in the wearing-off compared to dyski-
of the underlying pathology, as other PS variants of par- nesia cases (Rajput et al., 2004a). Similar observations
kinsonism may also improve (though the response is were reported on PET scanning in PD wearing-off cases
less robust and of shorter duration) (Rajput et al., (Fuente-Fernandez et al., 2001).
1990c; Birdi et al., 2002). The functional decline in DIP is a well-known complication of dopamine-
patients with late-age PD onset is more rapid compared depleting and receptor-blocking agents that was first
to younger-age-onset cases (Gibb and Lees, 1988a; recognized in the 1960s (Ayd, 1961; Delay and Deniker,
Goetz et al., 1988; Hietanen and Teravainen, 1988; 1968; Friedman, 1992). The DIP incidence is second
OLD AGE AND PARKINSONS DISEASE 439
only to PD in community studies of PS (Rajput et al., stroke patients, 6 (0.4%) developed parkinsonism
1984a; Bower et al., 1999). The elderly take a large (Alarcon et al., 2004). The onset of PS was rapid after
number of drugs, have age-related striatal dopamine approximately 4 months of stroke. Five of the 6 cases
loss (Kish et al., 1992) and up to 13% have preclinical had persistent parkinsonian features with initial improve-
pathology of PD, known as incidental Lewy body dis- ment on levodopa. The lesions were located in the mid-
ease (Ross et al., 2004). DIP is, therefore, more common brain/pons, caudate/internal capsule, lentiform nucleus,
in older individuals (Rajput, 1984, 1986; Friedman, the frontal cortex or frontal subcortex.
1992; Bower et al., 1999) and that incidence increases
with age (Bower et al., 1999). In more than 90% of these
53.7.2. Alzheimers disease and parkinsonism
cases, the parkinsonism emerges within 90 days of
neuroleptic initiation (Friedman, 1992). Some such
Alzheimers disease, a common disorder in the
patients have no known pathology, whereas others with
elderly, may be associated with parkinsonian features
a preclinical PD pathology would develop parkinsonian
in some patients (Molsa et al., 1984; Bennett et al.,
features on a modest neuroleptic dose (Rajput et al.,
1989; Morris et al., 1989; Tyrrell and Rossor, 1989;
1982). Although akinesia appears early in the DIP cases,
Soininen et al., 1992). Longer reaction time (bradyki-
that is due in part to the tranquilizing effect of the
nesia) and increased muscle tone (rigidity) are signif-
neuroleptics. The drug-induced patients do not always
icantly more common in Alzheimers disease patients
present an akinetic-rigid picture and may have clinical
compared to normal controls (Kischka et al., 1993).
features that are indistinguishable from idiopathic PD
In a longitudinal study, the bradykinesia rate nearly
(Rajput et al., 1982, 1991c; Hardie and Lees, 1988; Burn
doubled and rigidity frequency multiplied by nearly
and Brooks, 1993). We have observed reversible drug-
six times after 3 years in Alzheimers disease cases
induced tremor-dominant parkinsonism in an individual
(Soininen et al., 1992). When the Alzheimers disease
who had a strong family history of PD. Theoretically,
patients were treated with neuroleptics, nearly every-
the DIP should be symmetrical, but that is not always
one developed bradykinesia and rigidity (Soininen
the case.
et al., 1992), but tremor was not a prominent feature
Parkinsonism due to dopamine-depleting or recep-
in these cases. Morris et al. (1989) followed 44 clini-
tor-blocking drugs improves when the offending agent
cally diagnosed Alzheimers disease patients and 58
is discontinued (Friedman, 1992); however, it may
controls. At baseline, neither group had parkinsonian
take several months to resolve. The best treatment
features. At the end of 66 months, at least one of
option for such cases is discontinuing the offending
the three clinical features of parkinsonism resting
agent. A careful history of drug intake is, therefore,
tremor, bradykinesia and rigidity was detected in
important in elderly parkinsonian cases.
36% of Alzheimers cases compared to 5% in the
Anticholinergics are theoretically the best agents
control subjects. An additional 27% of Alzheimers
for the symptomatic treatment of DIP. However, the
cases developed at least one cardinal feature of parkin-
elderly do not tolerate these drugs well. Amantadine
sonism within 6 months of exposure to neuroleptics
and levodopa can be tried for symptomatic control.
(Morris et al., 1989).
The use of prophylactic anticholinergics in subjects
In a pathological study, histological findings were
treated with neuroleptics is not recommended, as only
such that a diagnosis of Alzheimers disease and of
a small proportion would develop DIP. Additionally,
Lewy body PD each could be made (Rajput et al.,
cholinergic-blocking agents produce significant
1993c). Two clinical patterns were detected. One
adverse effects in the elderly. If the neuroleptics can-
group presented with PS and dementia developed sev-
not be discontinued, atypical antipsychotic agents
eral years later. In the other group, the dementia and
such as olanzapine, clozapine or quetiapine should
parkinsonism onset were simultaneous (Rajput et al.,
be substituted to reduce the possibility of DIP
1993c). Those with PD at onset improved on levodopa
(Friedman, 1992).
until the onset of dementia, when they developed
small-amplitude irregular jerky postural and kinetic
53.7. Parkinsonism in other diseases of old age tremor and the levodopa response declined. In those
53.7.1. Stroke and parkinsonism who manifested PD and Alzheimers disease simulta-
neously, the typical resting tremor of PD was not a pro-
Stroke and parkinsonism are covered in detail in minent feature and the response to levodopa was poor,
Chapter 52. Although both stroke and PS are concen- producing major psychiatric side-effects (Rajput et al.,
trated in old age, there is no evidence that stroke 1993c). Alzheimers disease, dementia and PD overlap
produces Lewy body PD. In a recent report of 1500 will be discussed in detail in Chapters 18 and 60.
440 A. RAJPUT AND A. H. RAJPUT
References Bower JH, Maraganore DM, McDonnell SK et al. (1999).
Incidence and distribution of parkinsonism in Olmsted
Aarsland D, Andersen K, Larsen JP et al. (2003). Prevalence County, Minnesota, 19761990. Neurology 52: 12141220.
and characteristics of dementia in Parkinson disease: an Brooks DJ, Playford ED, Ibanez V et al. (1992). Isolated
8-year prospective study. Arch Neurol 60: 387392. tremor and disruption of the nigrostriatal dopaminergic sys-
Agid Y (1991). Parkinsons disease: pathophysiology. tem: an 18F-dopa PET study. Neurology 42: 15541560.
Lancet 337: 13211324. Burn DJ, Brooks DJ (1993). Nigral dysfunction in drug-
Agid Y, Javoy-Agid F, Ruberg M (1987). Biochemistry of induced parkinsonism: an 18F-dopa PET study. Neurology
neurotransmitters in Parkinsons disease. In: CD Marsden, 43: 552556.
S Fahn (Eds.), Movement Disorders 2. Butterworths and Calzetti S, Baratti M, Findley LJ (1987). Frequency/ampli-
Co, London, pp. 166230. tude characteristic of postural tremor of the hands in a
Agid Y, Cervera P, Hirsch E et al. (1989). Biochemistry of population of patients with bilateral essential tremor:
Parkinsons disease 28 years later: a critical review. Mov implications for the classification and mechanism of essen-
Disord 4: S126S144. tial tremor. J Neurol Neurosurg Psychiatry 50: 561567.
Alarcon F, Zijlmans JC, Duenas G et al. (2004). Post-stroke Canadian Study of Health and Aging Working Group
movement disorders: report of 56 patients. J Neurol Neu- (1994). Canadian study of health and aging: study meth-
rosurg Psychiatry 75: 15681574. ods and prevalence of dementia. CMAJ 150 (6): 899913.
Ayd FJ (1961). A survey of drug-induced extrapyramidal Cordes M, Snow BJ, Cooper S et al. (1994). Age-dependent
reaction. JAMA 175: 10541060. decline of nigrostriatal dopaminergic function: a positron
emission tomographic study of grandparents and their
Bain PG, Findley LJ, Thompson PD et al. (1994). A study of
grandchildren. Ann Neurol 36: 667670.
hereditary essential tremor. Brain 117: 805824.
Cotzias GC, Van Woert MH, Schiffer LM (1967). Aromatic
Baldereschi M, De Carlo A, Rocca WA et al. (2000). ILSA
amino acids and modification of parkinsonism. N Engl
Working Group. Parkinsons disease and parkinsonism in
J Med 276: 374379.
a longitudinal study. Two-fold higher incidence in men.
Neurology 55: 13581363. de la Monte SM, Wells SE, Hedley-Whyte ET et al. (1989).
Baloh RW, Fife TD, Zerling L et al. (1994). Comparison of Neuropathological distinction between Parkinsons
static and dynamic posturography in young and older normal dementia and Parkinsons plus Alzheimers disease. Ann
people. J Am Geriatr Soc 42 (4): 405412. Neurol 26: 309320.
Bennett DA, Beckett LA, Murray AM et al. (1996). Prevalence de Rijk MC, Rocca WA, Anderson DW et al. (1997).
of parkinsonian signs and associated mortality in a commu- A population perspective on diagnostic criteria for Parkin-
nity population of older people. N Engl J Med 334: 7176. sons disease. Neurology 48: 12771281.
Bennett RG, Greenough WB, Gloth FMI et al. (1989). Extra- Delay J, Deniker P (1968). Drug-induced extrapyramidal
pyramidal signs in dementia of Alzheimer type. Lancet syndromes. In: PJ Vinken, GW Bruyn (Eds.), Handbook
2 (8676): 1392. of Clinical Neurology.6th edn., Elsevier, North Holland,
Bergareche A, De La Puento E, Lopez de Munain A et al. Inc, New York, pp. 248266.
(2001). Prevalence of essential tremor: a door-to-door sur- Della Sala S, Francescani A, Spinnler H (2002). Gait apraxia
vey in Bidasoa, Spain. Neuroepidemiology 20: 125128. after bilateral supplementary motor area lesion. J Neurol
Bharucha NE, Bharucha AE, Bharucha EP et al. (1988). Pre- Neurosurg Psychiatry 72: 7785.
valence of Parkinsons disease in the Parsi Community of Desai T, Rajput AH, Desai HB (1990). Use and abuse of
Bombay, India. Arch Neurol 45: 13211323. drugs in the elderly. Prog Neuropsychopharmacol Biol
Birdi S, Rajput AH, Fenton M et al. (2002). Progressive Psychiatry 14: 779784.
supranuclear palsy diagnosis and confounding features Dogu O, Sevim S, Camdeviren H et al. (2003). Prevalence of
report on 16 autopsied cases. Mov Disord 17 (6): essential tremor: door-to-door neurologic exams in Mersin
12551264. Province, Turkey. Neurology 61: 18041806.
Birkmayer W, Hornykiewicz O (1961). The effect of L- Drachman DA, Long RR, Swearer JM (1994). Neurological
3,4-dihydroxyphenylalanine (L-DOPA) on akinesia in evaluation of the elderly patient. In: ML Albert, JE Knoefel
parkinsonism. Wien Klin Wochenschr 73 (45): 787788. (Eds.), Clinical Neurology of Aging, 2nd edn. Oxford
Birkmayer W, Hornykiewicz O (1998). The effect of L-3, University Press, New York, pp. 159180.
4-dihydroxyphenylalanine (DOPA) on akinesia in Du Pasquier RA, Blanc Y, Sinnreich M et al. (2003). The
parkinsonism. Parkinsonism Relat Disord 4: 5960. effect of aging on postural stability: a cross sectional and
Blaschke TF, Nies AS, Mamelok RD (1985). Principles of longitudinal study. Neurophysiol Clin 33 (5): 213218.
therapeutics. In: AG Gilman et al. (Eds.), The Pharmaco- Duncan G, Wilson JA (1989). Extrapyramidal signs in
logical Basis of Therapeutics, 7th edn, McMillan Publish- normal elderly. (Letter to the Editor) Lancet 2 (8676): 1392.
ing Company, New York, pp. 4965. Duvoisin RC (1990). The differential diagnosis of parkinson-
Bloem BR, Hausdorff JM, Visser JE et al. (2004). Falls and ism. In: GM Stern, (Ed.), Parkinsons Disease. The John
freezing of gait in Parkinsons disease: a review of two Hopkins University Press, Baltimore, pp. 431466.
interconnected, episodic phenomena. Mov Disord 19 (8): Duvoisin RC, Golbe LI (1989). Toward a definition of
871884. Parkinsons disease. Neurology 39: 746.
OLD AGE AND PARKINSONS DISEASE 441

Ehringer H, Hornykiewicz O (1960a). Distribution of nora- Global Parkinsons Disease Survey (GPDS) Steering Com-
drenaline and dopamine (3-hydroxytyramine) in human mittee (2002). Factors impacting on quality of life in Par-
brain: their behaviour in extrapyramidal system diseases. kinsons disease: results from an international survey.
Klin Wochenschr 38: 12361239. Mov Disord 17 (1): 6067.
Ehringer H, Hornykiewicz O (1960b). Verteilung von nora- Goetz CG, Tanner CM, Stebbins GT et al. (1988). Risk
drenalin und dopamin (3-hydroxytyramin) im gehirn des factors for progression in Parkinsons disease. Neurology
menschen und ihr verhalten bei erkrankungen des extra- 38: 18411844.
pyramidalen systems. Klin Wochenschr 38: 12361239. Graybiel AM, Hirsch EC, Agid Y (1990). The nigrostriatal
Ehringer H, Hornykiewicz O (1998). Distribution of noradre- system in Parkinsons disease. Adv Neurol 53: 1729.
naline and dopamine (3-hydroxytyramine) in the human Haerer AF, Anderson DW, Schoenberg BS (1982). Preva-
brain and their behavior in diseases of the extrapyramidal lence of essential tremor: results from the Copiah County
system. Parkinsonism Relat Disord 4: 5357. Study. Arch Neurol 39: 750751.
Elble RJ (1986). Physiologic and essential tremor. Neurol- Hallett M (1986). Differential diagnosis of tremor. In: PJ
ogy 36: 225231. Vinken, GW Bruyn, HL Klawans (Eds.), Handbook of
Elble RJ (2000). Essential tremor frequency decreases with Clinical Neurology; Extrapyramidal Disorders, 49th edn.
time. Neurology 55: 15471551. Elsevier Science Publishing Co., Inc, New York, pp.
Estanol BV (1981). Gait apraxia in communicating hydroce- 583595.
phalus. J Neurol Neurosurg Psychiatry 44: 305308. Hardie RJ, Lees AJ (1988). Neuroleptic-induced Parkinsons
Fahn S, Elton RL (1987), UPDRS Development Committee. syndrome: clinical features and results of treatment with
Unified Parkinsons disease rating scale. In: S Fahn et al. levodopa. J Neurol Neurosurg Psychiatry 51: 850854.
(Eds.), Recent Developments in Parkinsons disease, 2nd Hietanen M, Teravainen H (1988). The effect of age of disease
edn. Macmillan Healthcare Information. Florham Park, onset on neuropsychological performance in Parkinsons
NJ, pp. 153305. disease. J Neurol Neurosurg Psychiatry 51: 244249.
Fearnley JM, Lees AJ (1991). Ageing and Parkinsons disease: Hobson P, Meara J (2004). Risk and incidence of dementia
substantia nigra regional selectivity. Brain 114: 22832301. in a cohort of older subjects with Parkinsons disease in
Findley LJ, Gresty MA (1981). Tremor. Br J Hosp Med 26: the United Kingdom. Mov Disord 19 (9): 10431049.
1632. Hoehn MM, Yahr MD (1967). Parkinsonism: onset, progres-
Findley LJ, Koller WC (1995). Definitions and behavioral clas- sion, and mortality. Neurology 17: 427442.
sifications. In: WC Koller, LJ Findley (Eds.), Handbook of Hornykiewicz O, Kish SJ (1986). Biochemical pathophysiol-
Tremor Disorders. Marcel Dekker Inc., New York, pp. 15. ogy of Parkinsons disease. In: MD Yahr, KJ Bergmann
Findley LJ, Gresty MA, Halmagyi GM (1981). Tremor and (Eds.), Advances in Neurology, Vol. 45, Raven Press,
cogwheel phenomena and clonus in Parkinsons disease. New York, pp. 1934.
J Neurol Neurosurg Psychiatry 44: 534546. Hughes AJ, Daniel SE, Kilford L et al. (1992). Accuracy of
Fisher CM (1982). Hydrocephalus as a cause of disturbances clinical diagnosis of idiopathic Parkinsons disease: a clin-
of gait in the elderly. Neurology 32: 13581363. ico-pathological study of 100 cases. J Neurol Neurosurg
Forssberg H, Johnels B, Steg G (1984). Is parkinsonian gait Psychiatry 55: 181184.
caused by a regression to an immature walking pattern? Hughes AJ, Daniel SE, Ben-Schlomo Y et al. (2002). The
In: RG Hasler, JF Christ (Eds.), Advances in Neurology, accuracy of diagnosis of parkinsonian syndromes in a spe-
40th edn. Raven Press, New York, pp. 375379. cialist movement disorder service. Brain 125: 861870.
Fransson PA, Kristinsdottir EK, Hafstrom A et al. (2004). Jackson JA, Jankovic J, Ford J (1983). Progressive supranuc-
Balance control and adaptation during vibratory perturba- lear palsy: clinical features and response to treatment in 16
tions in middle-aged and elderly humans. Eur J Appl patients. Ann Neurol 13: 273278.
Physiol 91: 595603. Jankovic J, Fahn S (1980). Physiologic and pathologic tre-
Friedman JH (1992). Drug-induced parkinsonism. In: AE Lang, mors Diagnosis, mechanism, and management. Ann Intern
WJ Weiner (Eds.), Drug-Induced Movement Disorders. Futura Med 93: 460465.
Publishing Co., Inc, Mount Kisco, New York, pp. 4183. Jankovic J, McDermott M, Carter J et al. (1990). Parkinson
Fuente-Fernandez R, Lu JQ, Sossi V et al. (2001). Biochem- Study Group. Variable expression of Parkinsons disease:
ical variations in the synaptic level of dopamine precede a base-line analysis of the DATATOP cohort. Neurology
motor fluctuations in Parkinsons disease: PET evidence 40: 15291534.
of increased dopamine turnover. Ann Neurol 49: 298303. Jankovic J, Rajput AH, Golbe LI et al. (1993). What is it?
Gibb WRG (1991). Neuropathology of the substantia nigra. Case 1, 1993: parkinsonism, dysautonomia, and ophthal-
Eur Neurol 31 (Suppl 1): 4859. moparesis. Mov Disord 8 (4): 525532.
Gibb WR, Lees AJ (1988a). A comparison of clinical and Jankovic J, Beach J, Schwartz K et al. (1995). Tremor and
pathological features of young- and old-onset Parkinsons longevity in relatives of patients with Parkinsons disease,
disease. Neurology 38: 14021406. essential tremor, and control subjects. Neurology 45: 645648.
Gibb WRG, Lees AJ (1988b). The relevance of the Lewy Jankovic J, Schwartz KS, Ondo W (1999). Re-emergent tre-
body to the pathogenesis of idiopathic Parkinsons disease. mor of Parkinsons disease. J Neurol Neurosurg Psychiatry
J Neurol Neurosurg Psychiatry 51: 745752. 67: 646650.
442 A. RAJPUT AND A. H. RAJPUT

Jellinger K (1987). The pathology of parkinsonism. In: CD Louis ED, Marder K, Cote L et al. (1996). Prevalence of a
Marsden, S Fahn (Eds.), Movement Disorders 2. Butter- history of shaking in persons 65 years of age and older:
worths and Co., London, pp. 124165. diagnostic and functional correlates. Mov Disord 11:
Jenkyn LR, Reeves AG, Warren T et al. (1985). Neurological 6369.
signs in senescence. Arch Neurol 42: 11541157. Luxon LM (1984). A bit dizzy. Br J Hosp Med 32: 315.
Katsikitis MPI (1988). A study of facial expression in Par- Maki BE, Holliday PJ, Fernie GR (1990). Aging and postural
kinsons disease using a novel microcomputer-based control A comparison of spontaneous-and induced-sway
method. J Neurol Neurosurg Psychiatry 51: 362366. balance tests. J Am Geriatr Soc 38 (1): 19.
Kelly J, Taggart HM, McCullagh P (1995). Normal and Marder K, Leung D, Tang M et al. (1991). Are demented
abnormal tremor in the elderly. In: LJ Findley, WC Koller patients with Parkinsons disease accurately reflected in
(Eds.), Handbook of Tremor Disorders. Marcel Dekker, prevalence surveys? A survival analysis. Neurology 41:
Inc, New York, pp. 351370. 12401243.
Kischka U, Mandir AS, Ghika J et al. (1993). Electrophysio- Marsden CD (1989). Slowness of movement in Parkinsons
logic detection of extrapyramidal motor signs in Alzhei- disease. Mov Disord 4 (Suppl 1): S26S37.
mers disease. Neurology 43: 500505. Martin WE, Loewenson RB, Resch JA et al. (1973). Parkin-
Kish SJ, Shannak K, Hornykiewicz O (1988). Uneven sons disease: clinical analysis of 100 patients. Neurology
pattern of dopamine loss in the striatum of patients with 23: 783790.
idiopathic Parkinsons disease. N Engl J Med 318: Marttila RJ (1987). Epidemiology. In: WC Koller, (Ed.),
876880. Handbook of Parkinsons Disease. Marcel Dekker, Inc,
Kish SJ, Shannak K, Rajput A et al. (1992). Aging produces New York, pp. 3550.
a specific pattern of striatal dopamine loss: implications Marttila RJ, Rinne UK (1976). Dementia in Parkinson dis-
for the etiology of idiopathic Parkinsons disease. J Neuro- ease. Acta Neurol Scand 54: 431441.
chem 58: 642648. Matjucha ICA, Katz B (1994). Neuro-ophthalmology of aging.
Klawans HL (1981). Abnormal movements in the elderly. In: ML Albert, JE Knoefel (Eds.), Clinical Neurology of
Sandorama (Special Issue): 1518. Aging.2nd edn., Oxford University Press, New York p. 447.
Kline D, Sekuler R, Dismukes K (1982). Social issues, Mayeux R, Stern Y, Rosenstein R et al. (1988). An estimate
human needs, and opportunities for research on the effects of the prevalence of dementia in idiopathic Parkinsons
of age on vision: an over-view. In: R Sekuler, D Kline, disease. Arch Neurol 45: 260262.
K Dismukes (Eds.), Aging and Human Visual Function. Mayeux R, Chen J, Mirabello E et al. (1990). An estimate of
Alan R Liss, New York, pp. 36. the incidence of dementia in idiopathic Parkinsons dis-
Koller WC, Biary N (1984). Effect of alcohol on tremors: ease. Neurology 40: 15131517.
comparison with propranolol. Neurology 34: 221222. Mayeux R, Marder K, Cote LJ et al. (1995). The frequency
Koller WC, Glatt S, Wilson RS et al. (1982). Primitive of idiopathic Parkinsons disease by age, ethnic group,
reflexes and cognitive function in the elderly. Ann Neurol and sex in northern Manhattan, 19881993. Am J Epide-
12: 302304. miol 142: 820827.
Koller WC, Wilson R, Glatt S et al. (1983). Senile gait; correlation Menz HB, Lord SR, Fitzpatrick RC (2003). Age-related dif-
with computed tomographic scan. Ann Neurol 13: 343344. ferences in walking stability. Age Ageing 32 (2): 137142.
Koller WC, Hubble JP, Busenbark KL (1994). Essential tre- Mindham RHS, Ahmed SWA, Clough CG (1982). A con-
mor. In: DB Calne, (Ed.), Neurodegenerative Diseases. trolled study of dementia in Parkinsons disease. J Neurol
WB Saunders Co., Philadelphia, pp. 717742. Neurosurg Psychiatry 45: 969974.
Kostic V, Przedborski S, Flaster E et al. (1991). Early devel- Moghal S, Rajput AH, DArcy C et al. (1994). Prevalence of
opment of levodopa-induced dyskinesias and response movement disorders in elderly community residents. Neu-
fluctuations in young-onset Parkinsons disease. Neu- roepidemiology 13: 175178.
rology 41: 202205. Moghal S, Rajput AH, Meleth R et al. (1995). Prevalence of
LeWitt PA (1995). Tremor induced or enhanced by pharma- movement disorders in institutionalized elderly. Neuroepi-
cological means. In: LJ Findley, WC Koller (Eds.), Hand- demiology 14: 297300.
book of Tremor Disorders. Marcel Dekker, Inc, New Molsa PK, Marttila RJ, Rinne UK (1984). Extrapyramidal
York, pp. 473481. signs in Alzheimers disease. Neurology 34: 11141116.
Lexell J, Taylor CC, Sjostrom M (1988). What is the cause of Morgante L, Rocca WA, Di Rosa AE et al. (1992). Preva-
the ageing atrophy? Total number, size and proportion of lence of Parkinsons disease and other types of parkinson-
different fiber types studied in whole vastus lateralis muscle ism: a door-to-door survey in three Sicilian municipalities.
for 15- to 83-year-old men. J Neurol Sci 84: 275294. Neurology 42: 19011907.
Louis ED (2001). Essential tremor. N Engl J Med 345: Morris JC, Drazner M, Fulling K et al. (1989). Clinical and
887891. pathological aspects of parkinsonism in Alzheimers dis-
Louis ED, Marder K, Cote L et al. (1995). Differences in the ease. Arch Neurol 46: 651657.
prevalence of essential tremor among elderly African Parker SW (1994). Dizziness in the elderly. In: ML Albert,
Americans, whites, and Hispanics in northern Manhattan, JE Knoefel (Eds.), Clinical Neurology of Aging. 2nd edn.,
NY. Arch Neurol 52: 12011205. Oxford University Press, New York, pp. 569579.
OLD AGE AND PARKINSONS DISEASE 443

Quinn N (1989). Multiple system atrophythe nature of Rajput AH, Offord KP, Beard CM et al. (1987). A case control
the beast. J Neurol Neurosurg Psychiatry 1 (Special Suppl) study of smoking habits, dementia and other illnesses in
(7800): 7889. idiopathic Parkinsons disease. Neurology 37: 226232.
Rajput AH (1973). Levodopa in dystonia musculorum Rajput AH, Uitti RJ, Sudhakar S et al. (1989). Parkinsonism
deformans. Lancet 1: 432. and neurofibrillary tangle pathology in pigmented Nuclei.
Rajput AH (1984). Drug induced parkinsonism in the Ann Neurol 25: 602606.
elderly. Geriatr Med Today 3: 99107. Rajput AH, Rozdilsky B, Ang L (1990a). Site(s) of lesion
Rajput AH (1986). Parkinsons disease in the elderly. Med and resting tremor. Ann Neurol 28 (No.2): 296297.
North Am 1 (38): 101106. Rajput AH, Rozdilsky B, Rajput AH (1990b). Essential leg
Rajput AH (1992). Prevalence of dementia in Parkinsons tremor. Neurology 40: 1909.
disease. In: SJ Huber, JL Cummings (Eds.), Parkinsons Rajput AH, Rozdilsky B, Rajput A et al. (1990c). Levodopa
Disease. Neurobehavioral Aspects. Oxford University efficacy and pathological basis of Parkinson syndrome.
Press, New York, pp. 119131. Clin Neuropharmacol 13 (6): 553558.
Rajput AH (1993a). Accuracy of clinical diagnosis of idio- Rajput AH, Rozdilsky B, Ang L (1991a). Occurrence of resting
pathic Parkinsons disease. J Neurol Neurosurg Psychiatry tremor in Parkinsons disease. Neurology 41: 12981299.
56: 938939. Rajput AH, Rozdilsky B, Ang L et al. (1991b). Clinico-
Rajput AH (1993b). Diagnosis of PD. Letter. Neurology 43: pathological observations in essential tremor. Report of 6
16291630. cases. Neurology 41: 14221424.
Rajput AH (1993c). Parkinsonism, aging and gait apraxia. Rajput AH, Rozdilsky B, Rajput A (1991c). Accuracy of
In: MB Stern, WC Koller (Eds.), Parkinsonian Syndromes. clinical diagnosis in parkinsonisma prospective study.
Marcel Dekker, Inc, New York, pp. 511532. Can J Neurol Sci 18: 275278.
Rajput AH (1994a). Clinical features and natural history of
Rajput AH, Pahwa R, Pahwa P et al. (1993a). Prognostic sig-
Parkinsons disease (special consideration of aging). In:
nificance of the onset mode in parkinsonism. Neurology
DB Calne, (Ed.), Neurodegenerative Diseases. 1st edn,
43: 829830.
WB Saunders Company, Philadelphia, pp. 555571.
Rajput AH, Rozdilsky B, Ang L et al. (1993b). Significance
Rajput AH (1994b). Clinical features of tremor in extrapyra-
of parkinsonian manifestations in essential tremor. Can
midal syndromes. In: LJ Findley, WC Koller (Eds.),
J Neurol Sci 20: 114117.
Handbook of Tremor Disorders. Marcel Dekker, Inc,
Rajput AH, Rozdilsky B, Rajput A (1993c). Alzheimers dis-
New York, pp. 275291.
ease and idiopathic Parkinsons disease coexistence.
Rajput AH (2001). Levodopa prolongs life expectancy and is
J Geriatr Psychiatry Neurol 6: 170176.
non-toxic to substantia nigra. Parkinsonism Relat Disord
Rajput AH, Uitti RJ, Rajput AH et al. (1997). Timely levo-
8: 95100.
dopa (LD) administration prolongs survival in Parkinsons
Rajput AH (2002). Contributions of human brain biochem-
disease. Parkinsonism Relat Disord 3 (3): 159165.
ical studies to movement disorders. Parkinsonism Relat
Rajput AH, Fenton ME, Birdi S et al. (2002). Clinical-patho-
Disord 8: 425431.
Rajput AH, Rajput A (2004). Movement disorders and aging. logical study of levodopa complications. Mov Disord
In: RL Watts, WC Koller (Eds.), Movement Disorders. Neuro- 17 (2): 289296.
logic Principles & Practice, 2nd edn. McGraw-Hill, New Rajput AH, Fenton ME, Di Paolo T et al. (2004a). Human
York, pp. 837853. brain dopamine metabolism in levodopa-induced dys-
Rajput AH, Rozdilsky B (1975). Parkinsonism and dementia: kinesia and wearing-off. Parkinsonism Relat Disord 10:
effects of L-dopa. Lancet 1: 1084. 221226.
Rajput AH, Kazi KH, Rozdilsky B (1972). Striatonigral Rajput A, Robinson C, Rajput AH (2004b). Essential tremor
degeneration response to levodopa therapy. J Neurol Sci course and disability: a clinicopathological study of 20
16: 331341. cases. Neurology 62: 932936.
Rajput AH, Jamieson H, Hirsch S et al. (1975). Relative effi- Rautakorpi I, Marttila RJ, Takala J et al. (1982a). Occur-
cacy of alcohol and propranolol in action tremor. Can rences and causes of tremors. Neuroepidemiology 1:
J Neurol Sci 2: 3135. 209215.
Rajput AH, Rozdilsky B, Hornykiewicz O et al. (1982). Rautakorpi I, Takala J, Marttila RJ et al. (1982b). Essential
Reversible drug-induced parkinsonism Clinicopathologic tremor in a Finnish population. Acta Neurol Scand 66:
study of two cases. Arch Neurol 39: 644646. 5867.
Rajput AH, Offord KP, Beard CM et al. (1984a). Epidemiol- Robinson CA, Rajput AH (2005). The Neuropathology of
ogy of parkinsonism: incidence, classification, and mortal- Parkinsons Disease and Other Parkinsonian Disorders-
ity. Ann Neurol 16: 278282. Vol. 8493, CRC Press, Boca Raton, pp. 15901595.
Rajput AH, Offord KP, Beard CM et al. (1984b). Essential Rocca WA, Bower JH, McDonnell SK et al. (2001). Time
tremor in Rochester, Minnesota: a 45-year study. J Neurol trends in the incidence of parkinsonism in Olmsted
Neurosurg Psychiatry 47: 466470. County, Minnesota. Neurology 57: 462467.
Rajput AH, Stern W, Laverty WH (1984c). Chronic low dose Ross GW, Petrovitch H, Abbott RD et al. (2004). Parkinso-
therapy in Parkinsons disease: an argument for delaying nian signs and substantia nigra neuron density in decen-
levodopa therapy. Neurology 34 (8): 991996. dents elders without PD. Ann Neurol 56: 532539.
444 A. RAJPUT AND A. H. RAJPUT

Rowe JW (1988). Aging and geriatric medicine. In: JB Sudarsky L (1994). Gait disturbances in the elderly. In: ML
Wyngaarden, LH Smith (Eds.), Cecil Textbook of Medi- Albert, JE Knoefel (Eds.), Clinical Neurology of Aging.
cine. WB Saunders, Philadelphia, pp. 2127. 2nd edn., Oxford University Press, New York, pp. 483492.
Salemi G, Savettieri G, Rocca WA et al. (1994). Prevalence Sudarsky L, Ronthal M (1983). Gait disorders among elderly
of essential tremor: a door-to-door survey in Terrasini, patients A survey study of 50 patients. Arch Neurol 40:
Sicily. Neurology 44 (1): 6164. 740743.
Schaafsma JD, Balash Y, Gurevich T et al. (2003). Character- Sudarsky L, Simon S (1987). Gait disorder in late-life hydro-
ization of freezing of gait subtypes and the response of cephalus. Arch Neurol 44: 263267.
each to levodopa in Parkinsons disease. Eur J Neurol Suteerawattananon M, Morris GS, Etnyre BR et al. (2004).
10 (4): 391398. Effects of visual and auditory cues on gait in individuals
Scherman D, Desnos C, Darchen F et al. (1989). Striatal with Parkinsons disease. J Neurol Sci 219 (12): 6369.
dopamine deficiency in Parkinsons disease: role of aging. Tinetti ME, Speechley M, Ginter SF (1988). Risk factors for
Ann Neurol 26: 551557. falls among elderly persons living in the community.
Schoenberg BS, Anderson DW, Haerer AF (1985). Preva- N Engl J Med 319: 17011707.
lence of Parkinsons disease in the biracial population Tyrrell PJ, Rossor MN (1989). Extrapyramidal signs in
of Copiah County, Mississippi. Neurology 35 (6): dementia of Alzheimer type. Lancet 2 (8668): 920.
841845. Uitti RJ, Ahlskog JE, Maraganore DM et al. (1993). Levodo-
Schott JM, Rossor MN (2003). The grasp and other primitive pa therapy and survival in idiopathic Parkinsons disease:
reflexes. J Neurol Neurosurg Psychiatry 74: 558560. Olmsted County project. Neurology 43: 19181926.
Simons G, Pasqualini MC, Reddy V et al. (2004). Emotional Uitti RJ, Rajput AH, Ahlskog JE et al. (1996). Amantadine
and non-emotional facial expressions in people with Par- treatment is an independent predictor of improved survival
kinsons disease. J Int Neuropsychol Soc 10: 521535. in Parkinsons disease. Neurology 46: 15511556.
Soininen H, Laulumaa V, Helkala EL et al. (1992). Extrapyra- Webster DD (1968). Critical analysis of disability in Parkin-
midal signs in Alzheimers disease: a 3-year follow-up study. sons disease. Mod Treat 5 (2): 257282.
J Neural Transm Park Dis Dement Sect 4 (2): 107119. Weiner WJ, Nora LM, Glantz RH (1984). Elderly inpatients:
Steele JC, Richardson JC, Olszewski J (1964). Progressive postural reflex impairment. Neurology 34: 945947.
supranuclear palsy: a heterogeneous degeneration invol- Whitman GT, Tang T, Lin A et al. (2001). A prospective
ving the brain stem, ganglia and cerebellum with vertical study of cerebral white matter abnormalities in older
gaze and pseudobulbar palsy, nuchal dystonia and demen- people with gait dysfunction. Neurology 57: 990994.
tia. Arch Neurol 10: 333359. Wilson RS, Schneider JA, Beckett LA et al. (2002). Progres-
Stern G (1987). Prognosis in Parkinsons disease. In: CD sion of gait disorder and rigidity and risk of death in older
Marsden, S Fahn (Eds.), Movement Disorders 2. Butter- persons. Neurology 58: 18151819.
worth and Co. Ltd, London, pp. 9198. Winter DA, Patla AE, Frank JS et al. (1990). Biomechanical
Stiles RN (1976). Frequency and displacement amplitude walking pattern changes in the fit and healthy elderly.
relations for normal hand tremor. J Appl Physiol 40 (1): Phys Ther 70 (6): 340347.
4454. Zetusky WJ, Jankovic J, Pirozzolo FJ (1985). The Hetero-
Sudarsky L (1990). Geriatrics: gait disorders in the elderly. geneity of Parkinsons disease: clinical and prognostic
N Engl J Med 322: 14411446. implications. Neurology 35: 522526.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 54

Other degenerative processes

J. CARSTEN MOLLER* AND WOLFGANG H. OERTEL

Department of Neurology, Philipps-University Marburg, Marburg, Germany

54.1. Frontotemporal dementia and of affected patients from 62 known families was 470
parkinsonism linked to chromosome 17 in 2003 (Wszolek et al., 2003).
(FTDP-17) and related disorders The mean age at disease onset is 49 years and
the average disease duration is 8.5 years (Wszolek
Previously, several familial parkinsonismdementia syn- et al., 2003). The principal central nervous system
dromes, such as disinhibitiondementiaparkinsonism (CNS) manifestations consist of motor, cognitive and
amyotrophy complex and autosomal-dominant behavioral disturbances (Foster et al., 1997). Motor
parkinsonism and dementia with pallidopontonigral symptoms include:
degeneration, were distinguished (Wszolek et al.,
 akinetic-rigid parkinsonian features without
1992; Lynch et al., 1994). Subsequently, according
resting tremor
to a consensus conference, these syndromes were
 dystonia
summarized and termed FTDP-17 (Foster et al.,
 spasticity
1997). In 1998 this disease was found to be caused by
 supranuclear gaze palsy
mutations in the tau gene in several affected families
 occasionally amyotrophy
(Hutton et al., 1998). At least 26 different mutations
are known, with the exon-10 P301L missense mutation The cognitive disturbances comprise impaired
being most prevalent (Wszolek et al., 2003). Subse- executive functions: visuospatial orientation and com-
quently, several transgenic mouse models of FTDP-17 mon memory functions are relatively preserved until
have been developed based on these mutations (Lewis later stages of the disease. The behavioral symptoms
et al., 2000). Tau is an intracellular protein that pro- include:
motes the assembly and stabilization of microtubules.
 impaired social conduct
Mutations in the tau gene can be distinguished into
 hyperorality
two types (Rosso and van Swieten, 2002):
 hyperphagia
1. The first type of (missense) mutations reduces the  obsessive stereotyped behavior
ability of the tau protein to bind to microtubules.  psychosis
2. The second type of mutation results in a change in
Most patients harboring exon-10 missense and
the ratio (and subsequent accumulation) of tau
intronic mutations in the tau gene develop a parkinson-
isoforms with three amino acid repeats to those
ism-predominant phenotype (Wszolek et al., 2003).
with four amino acid repeats.
However, FTDP-17 features a wide range of phenoty-
Neuropathologically, frontotemporal and basal pic variations between kindreds with different tau
ganglia atrophy and substantia nigra depigmentation mutations but also among family members from the
are usually found. Microscopic examination reveals same kindred. This issue has recently been confirmed
neuronal loss and gliosis of variable intensity, tau- by a prospective study in two siblings carrying the
positive intraneuronal (and glial) inclusions without S305N tau mutation, in whom different rates of change
the characteristics of Pick bodies and ballooned neu- in whole-brain and ventricular volume as measured by
rons (Foster et al., 1997). The estimated total number magnetic resonance imaging (MRI) were found

*Correspondence to: PD Dr. med. Jens Carsten Moller, Klinik fur Neurologie, Philipps-Universitat Marburg, Rudolf-Bultmann-Str. 8,
35039 Marburg, Germany. E-mail: carsten.moeller@med.uni-marburg.de, Tel: 49-6421-286-5200, Fax: 49-6421-286-7055.
446 J. C. MOLLER AND W. H. OERTEL
(Boeve et al., 2005). Useful diagnostic criteria are in those with so-called atypical disease (Hayflick et al.,
general: (1) age at disease onset between the third and 2003). Classic NBIA was assumed in patients with
the fifth decade; (2) rapid disease progression; (3) par- disease onset during the first two decades, dystonia
kinsonism-plus symptoms (i.e. akinetic-rigid features and high globus pallidus iron with characteristic radio-
associated with early falls, supranuclear gaze palsy, graphic appearance. The term atypical disease was
apraxia, dystonia and lateralization); (4) frontotemporal applied to individuals not fitting the above criteria
dementia; and (5) personality and behavioral abnor- but with radiographic evidence of increased basal
malities (Wszolek et al., 2003). The diagnosis can be ganglia iron (Zhou et al., 2001). Patients with the clas-
confirmed by genetic testing, if available. Structural sic variant of the disease were usually shown to have
and functional imaging often reveals frontotemporal mutations resulting in predicted protein truncation
atrophy and/or hypometabolism. (Hayflick et al., 2003). The precise pathophysiology
There is a wide range of possible differential of PKAN is still unknown. PANK2 catalyzes the
diagnoses, including sporadic tauopathies such as pro- initial step in coenzyme A synthesis. It has thus been
gressive supranuclear palsy, corticobasal ganglionic suggested that the PANK2 mutations lead to coenzyme
degeneration and Picks disease. Apart from FTDP-17, A depletion associated with defective membrane bio-
there are also other types of familial frontotemporal synthesis (Zhou et al., 2001). Accordingly, it has been
dementia (FTD), i.e. ubiquitin-positive FTD and demen- shown that some point mutations indeed result in a
tia lacking distinctive histology (Morris et al., 2001). reduced catalytic activity during coenzyme A synth-
Ubiquitin-positive FTD can be accompanied by histolo- esis and that the most common mutation (G521R) is
gical features of (clinically often inapparent) motor neu- associated with a decreased production of mature
rone disease (Paviour et al., 2004). Ubiquitin-positive PANK2 (Kotzbauer et al., 2005).
FTD is possibly caused by mutations in the valosin- Besides accumulated cysteine, which would nor-
containing protein and may overlap with dementia lack- mally condense with phosphopantothenate, may form
ing distinctive histology (Josephs et al., 2004; Watts complexes with iron and cause oxidative damage in
et al., 2004; Schroder et al., 2005). Parkinsons disease the brain (Hayflick et al., 2003). Neuropathologically,
(PD) with dementia, dementia with Lewy bodies the most striking feature of NBIA is the rust-brown
and Alzheimers disease represent further differential pigmentation of the globus pallidus and pars reticulata
diagnoses of FTDP-17. of substantia nigra. On the microscopic level, iron
A specific therapy is not known. Paients usually granules were found in neurons, microglial cells
show poor response to levodopa. Psychiatric treatment and astrocytes. Some iron was also localized extra-
is usually necessary and often proves challenging cellularly. Furthermore, mulberry concretions were
despite pharmacological and behavioral therapies. observed in tissue and regarded as so-called pseudo-
calcium. A further prominent finding of the disease
54.2. Neurodegeneration with brain iron is a widely distributed distal axonal swelling. These
acculumation (NBIA) swellings may be surrounded by glial cells and contain
pigment granules, particularly when located in globus
This disorder was first reported in 1922 in a sibship pallidus or pars reticulata of substantia nigra. These
characterized by gait impairment resulting from structures are known as spheroid bodies (Hallervorden
rigidity of legs and feet deformity and mental and Spatz, 1922; Swaiman, 1991) and resemble those
deterioration with juvenile onset. The condition was found in neuroaxonal dystrophy (Seitelberger, 1957),
subsequently named HallervordenSpatz syndrome but in the latter the spheroids in the pallidum store
(HSS) (Hallervorden and Spatz, 1922). In response to fatty material and not pigments. These neuropathologi-
the unethical activities of Hallervorden and Spatz dur- cal alterations are accompanied by some loss of
ing World War II, it has been suggested that the name neurons and myelinated fibers as well as by gliosis.
HSS is replaced by the term NBIA. In analogy to PD, Lewy body-like intraneuronal inclu-
Recently, it has been shown that this disorder can sions containing a-synuclein were also observed in
be due to mutations in the gene for pantothenate kinase NBIA (Galvin et al., 2000). No sufficient epidemiolo-
2 (PANK2) (Taylor et al., 1996; Zhou et al., 2001). gical data of this group of rare disorders are available.
This type of NBIA is known as pantothenate kinase- Dooling et al. (1974) examined 42 patients who
associated neurodegeneration (PKAN). It is usually fulfilled both clinical and neuropathological criteria
an autosomal-recessive disorder, although in some for NBIA. Onset of disease occurred in 24 patients
cases the PANK2 mutation was suggested to be semi- before the age of 10 and 39 patients were ill before
dominant. In one study, PANK2 mutations were found age 22. The mean duration of disease was 11 years.
in all patients with classic NBIA and in one-third of Gait difficulty and postural impairment were noted as
OTHER DEGENERATIVE PROCESSES 447
initial symptoms in 37 patients. This may have been more often suffered from retinopathy. Conversely,
the result of spasticity, whereas symptoms of extrapyr- atypical patients more frequently presented with
amidal dysfunction could be delayed by 1 year to speech problems as part of the early disease (Hayflick
several years. However, the usual course included pro- et al., 2003). Besides, patients with a younger age of
gression of rigidity and presence of posture abnormal- onset (< 20 years) may have a higher frequency of dys-
ities. Other basal ganglia signs were observed, tonia, gait disturbance and tremor, whereas parkinson-
including dystonia in 23 patients, choreoathethosis in ism seems more common in late-onset disease ( 20
19 patients and tremor without any distinctive charac- years) (Thomas et al., 2004).
ter in 15 patients. Additionally, cognitive impairment Possible differential diagnoses of PKAN include
was common and, in 9 out of 42 patients, seizures Wilsons disease, Huntingtons disease, chorea
occurred (Dooling et al., 1974). Besides personality acanthocytosis (CHAC), neurometabolic disorders
changes such as impulsivity and violent outbursts, and other types of NBIA such as NBIA without
depression and emotional lability were observed in PANK2 mutations, neuroferritinopathy and acerulo-
patients with atypical disease and PANK2 mutations plasmenia. Neuroferritinopathy is a recently recog-
(Hayflick et al., 2003). In general, there was a wide nized, dominantly inherited movement disorder
spectrum of variation in the clinical manifestations of caused by a mutation of the ferritin light-chain gene
NBIA. It is noteworthy that 1 case of late-onset PKAN (Curtis et al., 2001). The clinical phenotype of this
presenting as familial parkinsonism was reported disorder is highly variable, with symptoms beginning
(Jankovic et al., 1985). Apart from the CNS mani- in the third to sixth decade. Chorea, dystonia or an
festations, foot deformities were frequently observed akinetic-rigid syndrome can predominate (Crompton
and skin pigmentation was noted in some cases et al., 2005). Aceruloplasmenia is an autosomal-
(Wigboldus and Bruyn, 1968). recessive disorder of iron metabolism characterized
A summary of the clinical presentation of NBIA by diabetes, retinal degeneration and progressive
based on a genotypephenotype analysis is provided neurodegeneration with extrapyramidal disorders,
below. Several obligatory and corroborative symptoms ataxia and dementia (Gitlin, 1998). In addition, HARP
for the diagnosis of NBIA were defined in 1991 syndrome (hypoprebetalipoproteinemia, acanthocyto-
(Swaiman, 1991). The obligatory symptoms were: sis, retinitis pigmentosa, pallidal degeneration) was
shown to be allelic with PKAN (Ching et al., 2002).
1. onset during the first two decades of life
Clinical diagnosis of NBIA can be confirmed by
2. a progressive course
MRI and genetic testing, if available. Laboratory
3. evidence of extrapyramidal dysfunction
investigations do not normally reveal any distinctive
The corroborative symptoms included: abnormalities, except that some patients feature vacuo-
lated circulating lymphocytes containing abnormal
1. pyramidal tract signs
cytosomes and sea-blue histiocytes in bone marrow
2. progressive mental deterioration,
(Swaiman et al., 1983). Computed tomography (CT)
3. hypodensities in basal ganglia on MRI
in patients with NBIA has been reported to show atro-
4. occurrence of seizures
phy and low-density lesions in basal ganglia (Dooling
5. ophthalmological symptoms, such as retinitis
et al., 1980). MRI using a high-field-strength unit (1.5
pigmentosa or optic atrophy
Tesla) showed decreased signal intensity in the glo-
6. positive family history
bus pallidus and pars reticulata of substantia nigra in
7. abnormal cytosomes in circulating lymphocytes
T2-weighted images, which is compatible with heavy-
and sea-blue histiocytes in bone marrow
metal (iron) deposits and a small area of hyperintensity
in the internal segment of pallidum, constituting the so-
A recent genotypephenotype analysis provided a called eye of the tiger sign (Rutledge et al., 1987; Sethi
novel concept of the clinical presentation of NBIA: et al., 1988). In all (homozygous and heterozygous)
patients with classic NBIA and PANK2 mutations PKAN patients, whether classic or atypical, MRI showed
had a mean age of onset of 3.4 years; usually presented the eye of the tiger sign, whereas this pattern was not
with gait or postural symptoms; featured dystonia, seen in atypical patients without PANK2 mutations
dysarthria, rigidity and choreoathetosis; and frequently (Hayflick et al., 2003).
developed spasticity and cognitive decline, whereas There is no specific therapy for NBIA. Sympto-
atypical NBIA patients with PANK2 mutations had a matic management, such as using levodopa and dopa-
mean age of onset of 13.7 years and featured less mine agonists, may be beneficial to patients (Swaiman,
severe and more slowly progressive extrapyramidal 1991). Hypothetically, supplementation of panthothe-
symptoms. Furthermore, patients with classic disease nate could ameliorate the symptoms in patients with
448 J. C. MOLLER AND W. H. OERTEL
PANK2 mutations (Hayflick et al., 2003). Simple case human body. The term normal asymmetry should
reports in the media described a dramatic improve- only be applied to differences in opposite limb length
ment with pantothenate, but no studies have been that are not visually apparent in the absence of mea-
performed yet. surements (Halperin, 1931). Interestingly, 1 probable
patient showed no hemiatrophy, but an enlarged late-
54.3. Hemiparkinsonhemiatrophy (HPHA) ral ventricle as a sign of brain hemiatrophy (Giladi
syndrome et al., 1990). With respect to the CNS manifestations,
Buchman et al. (1988) reported that the mean age of
HPHA is defined by the occurrence of a body onset of parkinsonism was 43.7 years. The course of
hemiatrophy with features of a highly asymmetric, the disease was slowly progressive, i.e. the mean dura-
often levodopa-responsive parkinsonism more promi- tion of disease until initiation of levodopa therapy was
nent on the side of the hemiatrophy (Klawans, 1981; 14.2 years. In contrast, in patients suffering from
Buchman et al., 1988). An association with contralat- idiopathic PD, levodopa treatment was started after
eral brain atrophy was also shown (Giladi et al., 4.1 years (Buchman et al., 1988). Furthermore, no pro-
1990). Although no systematic genetic studies have gression to a Hoehn and Yahr stage greater than III
yet been performed, parkin mutations were found in was observed (Buchman et al., 1988). Eight out of 15
1 patient with HPHA (Pramstaller et al., 2002). It has patients remained asymmetric after the development
been reported that lesions of the postcentral gyrus of bilateral disease. Giladi et al. (1990) found a
before the age of 3 are associated with a relative small- more variable clinical course; for instance, one patient
ness of the contralateral parts of the body (Penfield progressed from Hoehn and Yahr stage I to V during
and Robertson, 1943). Accordingly, it was suggested off periods within 2.5 years. Ipsilateral dystonic
that the occurrence of parkinsonism in HPHA patients movements, sometimes presenting as the first symp-
is related to an additional subcortical lesion (Klawans, tom, often occur early in the course of the disease
1981). Supporting this point of view, Giladi et al. before exposure to levodopa. A total of 10 out of 15
(1990) presented neuroradiological evidence of a con- patients showed tremor as their initial symptom. Apart
tralateral brain hemiatrophy in 64% of their patients. from the parkinsonian features, pyramidal tract
Since an association between perinatal asphyxia, brain dysfunction ipsilateral to the side of HPHA was pre-
hemiatrophy and delayed-onset hemidystonia has sent in 8 out of 15 patients (Buchman et al., 1988).
been shown, HPHA could represent an example of a Psychiatric symptoms are not usually a major feature
movement disorder with delayed onset as a conse- of this syndrome. In the differential diagnosis early-
quence of neonatal brain injury (Giladi et al., 1990). onset PD has to be considered. Common features are
Other authors reported 4 patients with dopa-responsive early age of onset and unilateral symptoms with mild
dystonia and hemiatrophy (Greene et al., 2000). Since progression. The main differences are the evidence of
dopa-responsive dystonia results from a purely ipsilateral hemiatrophy and the prominence of early
biochemical deficit in the brain, these authors sug- dystonia in HPHA patients. Therefore, the diagnosis
gested that a deficiency of the nigrostriatal dopamine depends on a thorough physical examination. More-
system may by itself be sufficient to cause body hemi- over, in the beginning the differential diagnosis may
atrophy. However, neonatal ablation of the nigrostria- include corticobasal ganglionic degeneration, since
tal pathway did not influence limb development in HPHA may mimic its early stage (Giladi and Fahn,
rats (Hebb et al., 2002). A variable response to levo- 1992). Due to the involvement of the nigrostriatal
dopa and striatal hypometabolism, as shown by 18F- dopaminergic system, the level of homovanillic acid
fluorodeoxyglucose and positron emission tomography in the cerebrospinal fluid can be reduced. Central
(PET), suggests that both pre- and postsynaptic mec- motor conduction time as measured by transcranial
hanisms contribute to HPHA. No postmortem analysis magnetic stimulation was normal in 2 HPHA patients
has been performed. So far reports on a total of 42 (Nardone et al., 2003). CT and MRI showed contralat-
patients have been published. eral brain asymmetry in 64% of investigated patients
Patients suffer from a body hemiatrophy with small (Giladi et al., 1990). However, Buchman et al. (1988)
and narrow extremities on one side. There is a wide found no evidence of cerebral hemiatrophy (cortical
variation in the degree of hemiatrophy: in some or ventricular asymmetry) in 11 out of 12 patients.
patients the face, arm and a leg were affected, whereas Studies using 18F-fluorodeoxyglucose and PET show-
in other patients only the hand seemed to be affected. ed a focal hypometabolism in the basal ganglia and
The most likely part of the hand to demonstrate hemi- the medial frontal cortex of the contralateral side,
atrophy was the thumb. It has to be kept in mind that whereas 18F-fluorodopa and PET revealed a reduction
there is normal asymmetry of the two halves of the in the striatal 18F-fluorodopa uptake. Hence the former
OTHER DEGENERATIVE PROCESSES 449
examination might be useful to distinguish HPHA gene product could be involved in intracellular protein
from typical unilateral PD (Przedborski et al., 1993, cycling. Absence of chorein may therefore lead to
1994). This observation was not confirmed by other destabilization of the plasma membrane structure and
authors (Pramstaller et al., 2002). hence acanthocytosis (Rampoldi et al., 2001).
Seven out of 9 investigated patients showed a good The suggested occurrence of CHAC without
response to levodopa therapy and 7 out of 8 patients acanthocytes may indicate that the gene can be vari-
responded well to a combination of amantadine and ably expressed in different tissues and may cause
anticholinergics (Buchman et al., 1988). Other investi- neurological abnormalities alone. In a family with
gators found a good response to levodopa treatment in CHAC featuring an autosomal-dominant transmission
7 out of 11 patients (Giladi et al., 1990). One patient and polyglutamine-containing neuronal inclusions
showed a dramatic improvement after subthalamic abnormalities of the membrane protein band 3 were
nucleus stimulation (Giladi et al., 1990). demonstrated by gel electrophoresis of red blood cell
membranes (Walker et al., 2002). Neuropathologically,
54.4. Chorea acanthocytosis the brains of CHAC patients showed enlargement
of the ventricles, particularly of the frontal horns.
Acanthocytes are erythrocytes with changed morphol- Caudate and putamen were the most severely affected
ogy bearing spicules of variable length and breadth brain areas showing atrophy, neuronal loss and
(Brecher and Bessis, 1972). If they occur in associa- gliosis. Depletion of small- and medium-sized striatal
tion with neurological symptoms, the term neuroa- neurons was most apparent. Involvement of globus
canthocytosis is used. Four different types of pallidus was also present and in some cases thalamus
neuroacanthocytosis are known: acanthocytes are and the anterior horns of spinal cord showed neuronal
observed in abetalipoproteinemia, the so-called Bas- loss and mild gliosis (Rinne et al., 1994b). The sub-
senKornzweig syndrome and in familial hypobetali- stantia nigra of 3 CHAC patients was investigated
poproteinemia (Kornzweig and Bassen, 1957; Young in more detail (Rinne et al., 1994a): in CHAC with
et al., 1987). Furthermore, they are detectable in parkinsonism a reduced neuronal density (particularly
CHAC (Critchley et al., 1968; Levine et al., 1968). in the ventrolateral region) of the substantia nigra
Finally, the McLeod syndrome represents a clinical was determined, whereas in 1 CHAC patient without
entity that is characterized by the occurrence of parkinsonian features the number of neurons was at
acanthocytes and neurological symptoms (Allen the lower limit of the control range. Epidemiological
et al., 1961; Swash et al., 1983). data for this rare disease are not available.
In addition there are some other conditions in which The mean age of onset of CHAC was 32 years, ran-
a sporadic association with acanthocytes has been ging from 8 to 62 years. The most striking symptoms
described, i.e. PKAN and mitochondrial encephalo- were involuntary movements affecting the orofacial
myopathies (Hardie, 1989). In adults CHAC is prob- region and the limbs, presenting as orofacial dyskine-
ably the most important disease. CHAC patients sias and limb chorea (Sakai et al., 1981). The former
usually feature chorea, oromandibular dyskinesias, can cause tongue- and lip-biting and very often inter-
dementia, seizures and peripheral neuropathy. Addi- feres with speech and swallowing. Accordingly,
tionally, parkinsonism may occur either with chorea involuntary vocalizations were frequently observed.
or as a subsequent feature, when the hyperkinetic In some cases, the predominant manifestation was
movement disorder subsides (Yamamoto et al., 1982; dystonia rather than chorea. Furthermore, Yamamoto
Spitz et al., 1985). CHAC is an autosomal-recessive et al. (1982) and Hardie (1989) described the occur-
disorder. The disease gene on chromosome 9q21 has rence of akinetic-rigid features simultaneously with
been identified and named chorein (Rampoldi et al., the appearance of hyperkinetic disorders in 2 out of 2
2001; Ueno et al., 2001). In 43 CHAC patients, 57 dif- and 5 out of 19 cases, respectively. Spitz et al.
ferent chorein mutations were found, indicating a (1985) reported 2 CHAC cases presenting as tics, that
strong allelic heterogeneity with no single mutation were progressively replaced or masked by progressive
causing the majority of cases (Dobson-Stone et al., parkinsonism. In 2 out of 19 patients, no movement
2002). Most mutations, however, lead to premature disorder could be established, indicating that this
termination codons and therefore predict the absence might not be a necessary condition (Hardie et al.,
or marked reduction of chorein. A transgenic mouse 1991). Additionally, hypo- or areflexia was repeatedly
model featuring a mild phenotype and a late-adult found. In many cases muscle wasting occurred and
onset has recently been established (Tomemori et al., axonal neuropathy was demonstrated (Serra et al.,
2005). The function of chorein is not known. Due to 1987). Moreover, one-third of patients suffered from
homology studies it has been hypothesized that the seizures and in more than half of the cases cognitive
450 J. C. MOLLER AND W. H. OERTEL
impairment, psychiatric features and personality movements to drug treatment was generally poor
change were seen (Hardie, 1989; Hardie et al., 1991). (Hardie, 1989). Severe trunk spasms were improved
The most frequent psychiatric symptoms were impul- by bilateral thalamic stimulation in 1 case (Burbaud
sive and distractable behavior, apathy and loss of et al., 2002).
insight. Additionally, depression, anxiety, paranoid
delusions and obsessive-compulsive features were 54.5. Pallidonigroluysian degeneration
observed.
The correct diagnosis depends on the combination The pallidal, pallidonigral, pallidoluysian and pallido-
of significant acanthocytosis with normal plasma lipo- nigroluysian degenerations (PNLD) include a number
proteins and neurological abnormalities. In this context of familial or sporadically occurring movement disor-
the clinical picture of CHAC with a predominantly ders, clinically defined by a slowly progressive course
hyperkinetic movement disorder, personality change and a wide variety of extrapyramidal symptoms. This
and cognitive impairment may resemble that of category of disorders has previously been classified
Huntingtons disease. Therefore, in any suspected case into four distinct groups:
of Huntingtons disease acanthocytosis should be
1. pure pallidal atrophy,
excluded. Usually diagnosis of Huntingtons disease
2. pure pallidoluysian atrophy
will be confirmed by neurogenetic methods. This
3. extended forms of pallidal degeneration, i.e. palli-
may be the case for CHAC in the future as well, but
donigral and pallidoluysionigral atrophy
at present its genetic diagnosis is costly and time-con-
4. variable forms of these subtypes with other
suming due to its genetic heterogeneity.
cerebrospinal degenerations (Jellinger, 1986).
Several methods are known to prove significant
acanthocytosis. The most efficient method is probably For instance, the separately described pallidopyra-
the examination of a wet preparation of isotonically midal disease (see section 54.6, below) could be
diluted blood samples (Storch et al., 2005). The nor- considered a member of this group (Jellinger, 1986).
mal range of acanthocytes in this kind of preparation Furthermore, the relation of the familial pallidonigral
is < 6.3%. Scanning electron microscopy may be help- system degeneration with cystic damage reported by
ful in measuring the extent of the erythrocyte morpho- McCormick and Lemmi (1965) to PNLD is not
logical abnormalities in some cases (Hardie et al., known. Finally, in a small number of cases there is
1991). Apart from acanthocytosis, modestly elevated evidence of a possibly different movement disorder,
creatine kinase levels have been reported (Sakai characterized by the isolated degeneration of the
et al., 1981). Other laboratory parameters show no external pallidum or status marmoratus of the basal
abnormalities. In contrast to BassenKornzweig syn- ganglia in association with the occurrence of intraneur-
drome and to familial hypobetalipoproteinemia, onal polyglucosan (Bielschowsky) bodies (Yagishita
decreased serum lipid levels are not found in CHAC. et al., 1983).
Western blotting of patient erythrocyte membranes in In this section we focus on PNLD. In most of the
order to assess the expression level of chorein may reported families this condition appears to be of
soon become available (Dobson-Stone et al., 2004). autosomal-recessive inheritance (Jellinger, 1986).
CT revealed cortical or occasional caudate atrophy as Recently, a mutation in the microtubule-associated
significant features (Serra et al., 1987; Hardie et al., protein tau, i.e. a substitution at codon 279, has been
1991). MRI showed focal and symmetric signal found in a PNLD case (Yasuda et al., 1999; Wszolek
abnormalities in the caudate and lentiform nuclei in 3 et al., 2000). This observation and the histopathologi-
out of 4 cases (Hardie et al., 1991). In another study cal demonstration of hyperphosphorylated tau in 1
increased signal intensity accompanied by scattered PLND case indicate that this disorder may be a tauopa-
bright spots in the striatum in T2-weighted images thy (Mori et al., 2001). It is not clear whether pallidal,
was observed (Tanaka et al., 1998). PET revealed the pallidoluysian and pallidonigral degeneration are pos-
following alterations: mean posterior putamen 18F- sibly tauopathies as well. Neuropathologically, a
fluorodopa uptake was reduced to 42% of normal; degeneration of the pallidum alone or in association
depressed frontal and striatal blood flow was seen; with the substantia nigra and/or the nucleus
and a loss of caudate and putamen D2-receptors was subthalamicus (of Luys) is found. A progressive loss
observed (Brooks et al., 1991; Tanaka et al., 1998). of nerve cells and fibers accompanied by proportional
Because there is no specific treatment known, gliosis has been found in these areas (Jellinger, 1986).
therapy remains symptomatic. However, parkinsonian The changes may vary in their extent. In rare instances
symptoms did not respond to high dosages of levodopa they might be associated with further degenerative
(Spitz et al., 1985) and the response of the involuntary lesions in other extrapyramidal, motor neuron or
OTHER DEGENERATIVE PROCESSES 451
spinocerebellar systems. A pure pallidal atrophy with cal gaze palsy and bradykinesia (Wooten et al.,
gliosis and locally differing neuronal loss was reported 1993). In PNLD the most apparent symptoms are
by Lange et al. (1970). progressive akinesia and rigidity with little or no tre-
In another case of pure pallidal atrophy, morpho- mor (Jellinger, 1986). Additionally, upward gaze
metric analysis revealed a shrinkage of the globus palsy or Parinauds syndrome has been observed. A
pallidus externus to 59% and the globus pallidus inter- PNLD case with rapidly progressive hemidystonia
nus to 37% of normal, but the neuron density did not has been reported (Vercueil et al., 2000).
seem to be affected (Aizawa et al., 1991). Bilateral In conclusion, a wide spectrum of different clinical
symmetrical loss of neurons and myelin with gliosis manifestations exists, depending on the spatial and
mainly in the outer pallidum was combined with pallor temporal pattern of effect of the distinct brain areas.
of the ansa lenticularis and atrophy of subthalamic Consequently, because of their rarity, the clinical cor-
nucleus in a case of pallidoluysian degeneration (van relates of the distinct forms are still not well described.
Bogaert, 1947). Familial occurrence of the combination of progressive
These changes are consistent with the findings rigidity and choreoathethosis or torsion dystonia with
observed in the various (extended) forms of this group an early onset may suggest one of these disorders.
of neurodegenerative disorders. Other observations The disorders may be accompanied by mental dete-
include the occurrence of corpora amylacea through- rioration, but intellectual impairment may be absent,
out the CNS and brown granular deposits showing a even in advanced disease stages (Jellinger, 1986).
positive reaction to iron in the degenerated nuclei A history of psychosis was reported in several cases
and the striatum in PNLD (Kosaka et al., 1981; Kawai (Klawans, 1981; Kawai et al., 1993). The PNLD patient
et al., 1993). Furthermore, hyperphosphorylated tau with the mutation in the tau gene featured dementia
was observed in 1 patient with PNLD (Mori et al., whereas no dementia was observed in the case with
2001). No epidemiological data are available due to the accumulation of hyperphosphorylated tau (Yasuda
the small number of investigated cases. et al., 1999; Mori et al., 2001). Except for the CNS
Nosologically, PNLD and its related disorders symptoms, no other clinical manifestations are known.
should be distinguished from autosomal-dominant The diagnosis of these rare conditions can only be
striatal degeneration (ADSD), another rare basal gang- proven by means of postmortem examination. Possible
lia disease (Kuhlenbaumer et al., 2004). ADSD is differential diagnoses include juvenile parkinsonism,
characterized by onset in the fourth or fifth decade, idiopathic torsion dystonia, PKAN, dentatorubral-
slow progression and prominent dysarthria with mild pallidoluysian atrophy, progressive supranuclear palsy,
hypokinesia, manifesting mainly as gait disturbance. multiple system atrophy, corticobasal ganglionic degen-
MRI usually shows a signal increase in the striatum eration and others. Sequencing of the tau gene, if
in T2-weighted images. available, should be performed. Routine laboratory data
PNLD and its related disorders demonstrate an insi- are unremarkable. CT in younger patients was normal
dious onset and a slowly progressive course. Onset of (Jellinger, 1986); minimal atrophy of the brainstem
illness in familial cases was between ages 5 and 40 and dilatation of the sylvian fissure were seen in a sin-
and in sporadic cases between ages 30 and 64. gle case (Aizawa et al., 1991). MRI of the brain in a
Depending on the variable pattern of morphological patient with pallidoluysian degeneration showed no
alterations, there may be distinct predominant symp- abnormalities (Wooten et al., 1993). T2-weighted
toms. In pure pallidal degeneration the clinical picture MRI demonstrated increased signal intensity in palli-
is characterized by the development of progressive dum and substantia nigra in a PNLD patient with hemi-
choreoathetotic hyperkinesias with axial dystonia, fol- dystonia (Vercueil et al., 2000). Furthermore,
lowed by the appearance of progressive rigidity. contralateral cortical hyperperfusion was observed in
Finally, the involuntary movements are overcome by this patient.
permanent rigidity and the patients become bed-ridden Treatment with levodopa has produced only equivo-
(Jellinger, 1986). In contrast, Aizawa et al. (1991) cal improvement of the movement disorder (Aizawa
reported a case of pallidal degeneration with an et al., 1991; Yamamoto et al., 1991). However, 1
extreme slowness of movements without rigidity as patient with dystonic symptoms was reported to benefit
the main symptom. In pallidoluysian degeneration, from baclofen (Wooten et al., 1993).
additional symptoms are torticollis, head tremor and
distal movements with or without a ballistic component 54.6. Pallidopyramidal disease
(Jellinger, 1986). Another case of pallidoluysian
degeneration presented with 20 years of progressive Pallidopyramidal disease is thought to be an autosomal-
generalized dystonia, dysarthria, gait disorder, verti- recessive disorder with onset in the second or early third
452 J. C. MOLLER AND W. H. OERTEL
decade, with a clinical picture consisting of parkinson- KuforRakeb syndrome showed, in contrast, general-
ism and pyramidal tract signs (Davison, 1954; Horowitz ized atrophy on MRI with pronounced atrophy of the
and Greenberg, 1975; Tranchant et al., 1991; Nisipeanu lentiform nuclei and the pyramids (Najim al-Din
et al., 1994). Furthermore, a syndrome was reported that et al., 1994). PET with 18F-fluorodopa was performed
is closely related but not identical to pallidopyramidal in 2 patients with pallidopyramidal disease and
disease. It is called KuforRakeb syndrome and addi- showed marked dopaminergic denervation of the stria-
tionally characterized by supranuclear upgaze paresis tum (Remy et al., 1995). PET with 11C-flumazenil
and dementia (Najim al-Din et al., 1994). The disease demonstrated a marked decrease in benzodiazepine-
locus for KuforRakeb syndrome has been mapped to receptor density in the precentral gyrus and the mesial
chromosome 1p36 (Hampshire et al., 2001). The patho- frontal cortex (Pradat et al., 2001).
genesis of pallidopyramidal disease is unknown. Until Extrapyramidal symptoms improved with levodopa
now only one autopsy in a non-familial patient 50 years therapy. Typically, the pyramidal symptoms were not
after onset has been performed (Davison, 1954). A pal- influenced by this treatment. However, Tranchant
lor of the pallidal segments, slight shrinkage and cellu- et al. (1991) reported a worsening of pyramidal tract
lar change of the substantia nigra, a thinning of the ansa signs due to the medication regimen. Response to
lenticularis and early demyelination of the pyramids levodopa was somewhat variable; most patients
and crossed pyramidal tracts were observed. The latter responded rapidly to low doses, with improvement
extended from the lower parts of the medulla oblongata persisting for a long period. After many years of treat-
into the spinal cord. Until now 11 familial and 4 non- ment, wearing-off phenomena occurred. The daily
familial cases of pallidopyramidal disease have been dose of levodopa used in the patients reported by
described. Nisipeanu et al. (1994) was 5001000 mg.
Pallidopyramidal disease is characterized by its
CNS symptoms. In general, disease onset occurred 54.7. Rett syndrome
during the second or the early third decade. The two
siblings reported by Horowitz and Greenberg (1975) Rett syndrome is a progressive neurodegenerative dis-
developed their first symptoms in the first decade of order which is reported almost exclusively in females
life. The classical parkinsonian features were observed and characterized by a wide spectrum of motor and
in all cases. Pyramidal tract signs, consisting of hyper- behavioral abnormalities. It was first described by Rett
reflexia, spastic muscle tone and bilateral extensor (1966, 1977) and subsequently investigated by other
plantar responses, were also found. In the patients authors (Hagberg et al., 1983; Hagberg and
described by Nisipeanu et al. (1994) the occurrence Witt-Engerstrom, 1986). It has been proposed that Rett
of pyramidal tract signs preceded the appearance of syndrome is the result of an X-linked-dominant
extrapyramidal symptoms. Only a slow progression mutation with mortality for hemizygous males, with
of the disorder was noted: the 2 patients investigated each case representing a new mutation (Comings,
by Horowitz and Greenberg (1975) were still ambula- 1986). De novo mutations, possibly in a particular
tory after 1013 years of disease. No diurnal variation open reading frame, of the gene encoding X-linked
was observed. Some patients examined by Davison methyl-CpG-binding protein 2 (MeCP2) have been
(1954) showed additional symptoms such as horizontal identified as a cause of Rett syndrome (Amir et al.,
nystagmus, intention tremor, poor memory and 1999; Mnatzakanian et al., 2004). Methylation of
impaired intelligence. Psychiatric symptoms have not CpG dinucleotides in genomic DNA represents a fun-
been observed in this syndrome. However, 1 patient damental epigenetic mechanism of gene expression
reported by Davison had impaired intelligence. control. MeCP2 likely causes transcriptional repres-
Furthermore, 3 out of the 5 patients with KuforRakeb sion through an interaction with core histones since
syndrome were demented (Najim al-Din et al., 1994). the MeCP2-binding domain was shown to associate
The diagnostic possibilities are limited. Evidence of with histone deacetylases. Mutations in the MeCP2
isolated extrapyramidal and pyramidal signs and gene have been identified in 7590% of sporadic
normal laboratory and neuroradiologic investigations cases and approximately 50% of the rare familial cases
in a young adult are suggestive of pallidopyramidal (Shahbazian and Zoghbi, 2001). However, MECP2
disease. Possible differential diagnoses are juvenile mutations can also be found in individuals lacking
parkinsonism and levodopa-responsive dystonia. the clinical features of Rett syndrome (Hagberg
No biochemical criteria for diagnosis are known. et al., 2002). Genotypephenotype studies suggest that
Three out of the 4 patients examined by Nisipeanu the pattern of X-chromosome inactivation has a more
et al. (1994) were subjected to cranial CT and MRI. prominent effect on clinical severity than the type of
No abnormalities were found. Patients suffering from mutation (Shahbazian and Zoghbi, 2001). It still
OTHER DEGENERATIVE PROCESSES 453
remains to be determined why mutations of the widely (Braddock et al., 1993). Gastrointestinal complaints
expressed MECP2 gene give rise to a predominantly include constipation and weight loss. Swallowing
neuronal phenotype. difficulties are also common. Furthermore, an unusual
Recently, DLX5 was identified as one of the genes breathing pattern with central apnea intermixed with
probably targeted by MECP2 (Horike et al., 2005). hyperventilation is frequently observed. Scoliosis
Loss of MECP2 caused overexpression of this tran- often occurs. Patients with Rett syndrome have signif-
scription factor important for GABAergic neurons. icantly longer corrected QT intervals and T-wave
Autopsy studies showed diffuse cerebral atrophy with abnormalities on electrocardiograms that might
a decrease in brain weight of 1434% compared to that explain sudden death in Rett syndrome (Sekul et al.,
of age-matched controls with increased amounts of 1994).
lipofuscin and, occasionally, mild astrocytosis. Criteria for the diagnosis of Rett syndrome were
Moreover, mild but inconsistent spongy changes of initially developed by The Rett Syndrome Diagnostic
white matter were found. Most apparent was a low Criteria Work Group (1988). These criteria have been
level of pigmentation of the substantia nigra, whereas revised in the light of the recent advances in the under-
the number of nigral neurons was normal (Jellinger standing of the molecular biology of Rett syndrome
and Seitelberger, 1986). In addition, selective dendritic (Hagberg et al., 2002). Necessary criteria for the diag-
alterations in the cortex of patients with Rett syndrome nosis of Rett syndrome are apparently normal prenatal
were reported (Armstrong et al., 1995). The preva- and perinatal history; a largely normal (or delayed) psy-
lence of Rett syndrome is estimated to be about 0.44/ chomotor development through the first 6 months; nor-
10 000 (Kozinetz et al., 1993). mal head circumference at birth; postnatal deceleration
The most typical symptoms are stereotyped of head growth in the majority; loss of purposeful hand
movements and gait disturbance. A four-stage model skills at the age of 0.52.5 years; stereotypic hand
for the description of Rett syndrome was proposed movements; emerging social withdrawal, communica-
(Hagberg and Witt-Engerstrom, 1986). Stage 1 is tion dysfunction, loss of learned words and cognitive
defined by developmental stagnation, hypotonia impairment; and impaired or failing locomotion. Sup-
and deceleration of head growth (onset 6 months to portive criteria include awake disturbances of breath-
1.5 years). Stage 2 is characterized by loss of func- ing; bruxism; impaired sleep pattern from early
tional hand use, stereotypic hand-wringing, loss of infancy; abnormal muscle tone; peripheral vasomotor
expressive language, rapid developmental regression disturbances; scoliosis/kyphosis progressing through
and occasional seizures (onset 13 or 4 years). Stage childhood; growth retardation; and small hands and
3 is termed a pseudostationary period because of some feet. Diagnostic criteria for variant Rett syndrome have
restitution of communication, but increasing ataxia, also been developed (Hagberg and Skjeldal, 1994). Par-
hyperreflexia and rigidity, as well as breathing dys- kinsonism has not so far been included among the diag-
function and bruxism, are observed. After several nostic criteria of Rett syndrome or variant Rett
years stage 4 develops with the so-called late motor syndrome. Infantile autism is one of the most important
deterioration and growth retardation. With respect to differential diagnoses. Further differential diagnoses
extrapyramidal dysfunction, bruxism (97%), oculogy- include other neurodevelopmental disorders with men-
ric crises (63%) and parkinsonism and dystonia tal retardation and motor impairment. Sequencing of
(59%) are common features (FitzGerald et al., 1990). the MECP2 gene, if available, is advisable.
Myoclonus and choreoatheosis were seen only infre- It was proposed that hyperammonemia could be an
quently. In younger patients (i.e. < 4 years) hyperki- essential sign of this condition (Rett, 1966, 1977), but
netic disorders were more evident, whereas in older further investigations did not reproduce the findings of
patients (i.e. > 8 years) the bradykinetic syndrome hyperammonemia in most patients (Hagberg et al.,
tended to predominate. Drooling (75%), rigidity 1983). Significant reductions in the metabolites of
(44%) and bradykinesia (41%) were the most often norepinephrine, dopamine and serotonin, as well as
observed parkinsonian symptoms. an elevation of biopterin in the cerebrospinal fluid,
Sleep disturbances, mainly in the early stages, are constituted the first detected biochemical alterations
frequently present in Rett syndrome. They are charac- (Zoghbi et al., 1989). Additionally, there is evidence
terized by an overall increase in daytime sleep and a of elevation of lactate, pyruvate, alpha-ketoglutarate,
delayed sleep onset at night. Despite their mental malate and glutamate in the cerebrospinal fluid
retardation and their loss of expressive language, these (Hamberger et al., 1992; Matsuishi et al., 1994). MRI
patients tend to appear happy and enjoy close physical indicated a global hypoplasia of the brain and progres-
contact. Rett syndrome leads not only to neurological sive cerebellar atrophy increasing with age (Murakami
abnormalities, but also to dysfunction of other organs et al., 1992). An increased density of D2-receptors in
454 J. C. MOLLER AND W. H. OERTEL
the striatum of patients suffering from Rett syndrome Ching KH, Westaway SK, Gitschier J et al. (2002). HARP
using single-photon emission spectroscopy (SPECT) syndrome is allelic with pantothenate kinase-associated
imaging was found (Chiron et al., 1993). 18F-fluoro- neurodegeneration. Neurology 58: 16731674.
deoxyglucose PET showed several areas of hypometa- Chiron C, Bulteau B, Loch C et al. (1993). Dopaminergic D2
bolism with markedly lower metabolism in the receptor SPECT imaging in Rett syndrome: increase of
occipital lobes (Naidu et al., 1992). Furthermore, a mild specific binding in striatum. J Nucl Med 34: 17171721.
presynaptic deficit of nigrostriatal activity was demon- Comings DE (1986). The genetics of Rett syndrome: the
strated by 18F-fluorodopa and PET (Dunn et al., 2002). consequences of a disorder where every case is a new
mutation. Am J Med Genet Suppl 1: 383388.
There is no specific treatment. Naltrexone appears to
provide clinical benefit in the treatment of breathing dys- Critchley EM, Clark DB, Wikler A (1968). Acanthocytosis
and neurological disorder without betalipoproteinemia.
function and cognitive impairment (Percy et al., 1994).
Arch Neurol 18: 134140.
Furthermore, bromocriptine and l-carnitine can be tried
Crompton DE, Chinnery PF, Bates D et al. (2005). Spectrum
to improve certain disease symptoms (Zappella et al.,
of movement disorders in neuroferritinopathy. Mov Dis-
1990; Plioplys and Kasnicka, 1993). Lamotrigine and ord 20: 9599.
topiramate have been used for seizure control Curtis AR, Fey C, Morris CM et al. (2001). Mutation in the
(Kumandas et al., 2001; Goyal et al., 2004). Symptomatic gene encoding ferritin light polypeptide causes dominant
therapeutic approaches also include physiotherapy and adult-onset basal ganglia disease. Nat Genet 28: 350354.
music therapy (Armstrong et al., 1995). Davison C (1954). Pallido-pyramidal disease. J Neuropathol
Exp Neurol 13: 5059.
References Dobson-Stone C, Danek A, Rampoldi L et al. (2002). Muta-
tional spectrum of the CHAC gene in patients with chorea-
acanthocytosis. Eur J Hum Genet 10: 773781.
Aizawa H, Kwak S, Shimizu T et al. (1991). A case of adult
onset pure pallidal degeneration. I. Clinical manifestations Dobson-Stone C, Velayos-Baeza A, Filippone LA et al. (2004).
and neuropathological observations. J Neurol Sci 102: Chorein detection for the diagnosis of chorea-acanthocytosis.
7682. Ann Neurol 56: 299302.
Allen FH, Krabbe FMR, Corcoran PA (1961). A new pheno- Dooling EC, Schoene WC, Richardson EP Jr (1974). Haller-
type (McLeod) in the Kell blood group system. Vox Sang vorden-Spatz syndrome. Arch Neurol 30: 7083.
6: 555560. Dooling EC, Richardson EP Jr, Davis KR (1980). Computed
Amir RE, Van den Veyver IB, Wan M et al. (1999). Rett tomography in Hallervorden-Spatz disease. Neurology 30:
syndrome is caused by mutations in X-linked MECP2, 11281130.
encoding methyl-CpG-binding protein 2. Nat Genet 23: Dunn HG, Stoessl AJ, Ho HH et al. (2002). Rett syndrome:
185188. investigation of nine patients, including PET scan. Can J
Armstrong D, Dunn JK, Antalffy B et al. (1995). Selective Neurol Sci 29: 345357.
dendritic alterations in the cortex of Rett syndrome. J Neu- FitzGerald PM, Jankovic J, Percy AK (1990). Rett syndrome
ropathol Exp Neurol 54: 195201. and associated movement disorders. Mov Disord 5:
Boeve BF, Tremont-Lukats IW, Waclawik AJ et al. (2005). 195202.
Longitudinal characterization of two siblings with Foster NL, Wilhelmsen K, Sima AA et al. (1997). Frontotem-
frontotemporal dementia and parkinsonism linked to poral dementia and parkinsonism linked to chromosome
chromosome 17 associated with the S305N tau mutation. 17: a consensus conference. Ann Neurol 41: 706715.
Brain 128: 752772. Galvin JE, Giasson B, Hurtig HI et al. (2000). Neuro-
Braddock SR, Braddock BA, Graham JM Jr (1993). Rett degeneration with brain iron accumulation, type 1 is
syndrome. An update and review for the primary pediatri- characterized by alpha-, beta-, and gamma-synuclein
cian. Clin Pediatr (Phila) 32: 613626. neuropathology. Am J Pathol 157: 361368.
Brecher G, Bessis M (1972). Present status of spiculed red Giladi N, Fahn S (1992). Hemiparkinsonism-hemiatrophy
cells and their relationship to the discocyte-echinocyte syndrome may mimic early-stage cortical-basal ganglionic
transformation: a critical review. Blood 40: 333344. degeneration. Mov Disord 7: 384385.
Brooks DJ, Ibanez V, Playford ED et al. (1991). Presynaptic Giladi N, Burke RE, Kostic V et al. (1990). Hemiparkinson-
and postsynaptic striatal dopaminergic function in neuroa- ism-hemiatrophy syndrome: clinical and neuroradiologic
canthocytosis: a positron emission tomographic study. features. Neurology 40: 17311734.
Ann Neurol 30: 166171. Gitlin JD (1998). Aceruloplasminemia. Pediatr Res 44:
Buchman AS, Goetz CG, Klawans HL (1988). Hemiparkin- 271276.
sonism with hemiatrophy. Neurology 38: 527530. Goyal M, ORiordan MA, Wiznitzer M (2004). Effect of
Burbaud P, Rougier A, Ferrer X et al. (2002). Improvement topiramate on seizures and respiratory dysrhythmia in Rett
of severe trunk spasms by bilateral high-frequency stimu- syndrome. J Child Neurol 19: 588591.
lation of the motor thalamus in a patient with chorea- Greene PE, Bressman SB, Ford B et al. (2000). Parkinsonism,
acanthocytosis. Mov Disord 17: 204207. dystonia, and hemiatrophy. Mov Disord 15: 537541.
OTHER DEGENERATIVE PROCESSES 455

Hagberg B, Aicardi J, Dias K et al. (1983). A progressive Jellinger K, Seitelberger F (1986). Neuropathology of Rett
syndrome of autism, dementia, ataxia, and loss of purpose- syndrome. Am J Med Genet Suppl 1: 259288.
ful hand use in girls: Retts syndrome: report of 35 cases. Josephs KA, Jones AG, Dickson DW (2004). Hippocampal
Ann Neurol 14: 471479. sclerosis and ubiquitin-positive inclusions in dementia
Hagberg B, Hanefeld F, Percy A et al. (2002). An update on lacking distinctive histopathology. Dement Geriatr Cogn
clinically applicable diagnostic criteria in Rett syndrome. Disord 17:342345.
Comments to Rett Syndrome Clinical Criteria Consensus Kawai J, Sasahara M, Hazama F et al. (1993). Pallidonigro-
Panel Satellite to European Paediatric Neurology Society luysian degeneration with iron deposition: a study of three
Meeting, Baden Baden, Germany, 11 September. Eur J autopsy cases. Acta Neuropathol (Berl) 86: 609616.
Paediatr Neurol 6: 293297. Klawans HL (1981). Hemiparkinsonism as a late complica-
Hagberg B, Witt-Engerstrom I (1986). Rett syndrome: a sug- tion of hemiatrophy: a new syndrome. Neurology 31:
gested staging system for describing impairment profile 625628.
with increasing age towards adolescence. Am J Med Kornzweig AL, Bassen FA (1957). Retinitis pigmentosa,
Genet Suppl 1: 4759. acanthrocytosis and heredodegenerative neuromuscular
Hagberg BA, Skjeldal OH (1994). Rett variants: a suggested disease. AMA Arch Ophthalmol 58: 183187.
model for inclusion criteria. Pediatr Neurol 11: 511. Kosaka K, Matsushita M, Oyanagi S et al. (1981). Pallido-
Hallervorden J, Spatz H (1922). Eigenartige Erkrankung im nigro-luysial atrophy with massive appearance of corpora
extrapyramidalen System mit besonderer Beteiligung des amylacea in the CNS. Acta Neuropathol (Berl) 53:
Globus pallidus und der Substantia nigra. Z Gesamte 169172.
Neurol Psychiatr 79: 254302. Kotzbauer PT, Truax AC, Trojanowski JQ et al. (2005).
Halperin G (1931). Normal asymmetry and unilateral hyper- Altered neuronal mitochondrial coenzyme a synthesis in
trophy. Arch Intern Med 48: 676684. neurodegeneration with brain iron accumulation caused
Hamberger A, Gillberg C, Palm A et al. (1992). Elevated by abnormal processing, stability, and catalytic activity
CSF glutamate in Rett syndrome. Neuropediatrics 23: of mutant pantothenate kinase 2. J Neurosci 25: 689698.
212213. Kozinetz CA, Skender ML, MacNaughton N et al. (1993).
Hampshire DJ, Roberts E, Crow Y et al. (2001). Kufor- Epidemiology of Rett syndrome: a population-based reg-
Rakeb syndrome, pallido-pyramidal degeneration with istry. Pediatrics 91: 445450.
supranuclear upgaze paresis and dementia, maps to 1p36. Kuhlenbaumer G, Ludemann P, Schirmacher A et al.
J Med Genet 38: 680682. (2004). Autosomal dominant striatal degeneration
Hardie RJ (1989). Acanthocytosis and neurological impair- (ADSD): clinical description and mapping to 5q135q14.
menta review. Q J Med 71: 291306. Neurology 62: 22032208.
Hardie RJ, Pullon HW, Harding AE et al. (1991). Neuroa- Kumandas S, Caksen H, Ciftci A et al. (2001). Lamotrigine
canthocytosis. A clinical, haematological and pathological in two cases of Rett syndrome. Brain Dev 23: 240242.
study of 19 cases. Brain 114: 1349. Lange E, Poppe W, Scholtze P (1970). Familial progressive
Hayflick SJ, Westaway SK, Levinson B et al. (2003). Genet- pallidum atrophy. Eur Neurol 3: 265267.
ic, clinical, and radiographic delineation of Hallervorden- Levine IM, Estes JW, Looney JM (1968). Hereditary neuro-
Spatz syndrome. N Engl J Med 348: 3340. logical disease with acanthocytosis. Arch Neurol 19:
Hebb MO, Lang AE, Fletcher PJ et al. (2002). Neonatal 403409.
ablation of the nigrostriatal dopamine pathway does not Lewis J, McGowan E, Rockwood J et al. (2000). Neurofibril-
influence limb developments in rats. Exp Neurol 177: lary tangles, amyotrophy and progressive motor distur-
547556. bance in mice expressing mutant (P301L) tau protein.
Horike S, Cai S, Miyano M et al. (2005). Loss of silent-chro- Nat Genet 25: 402405.
matin looping and impaired imprinting of DLX5 in Rett Lynch T, Sano M, Marder KS et al. (1994). Clinical
syndrome. Nat Genet 37: 3140. [Epub 2004 Dec 2019]. characteristics of a family with chromosome 17-linked dis-
Horowitz G, Greenberg J (1975). Pallido-pyramidal syn- inhibition-dementia-parkinsonism-amyotrophy complex.
drome treated with levodopa. J Neurol Neurosurg Psychia- Neurology 44: 18781884.
try 38: 238240. Matsuishi T, Urabe F, Percy AK et al. (1994). Abnormal
Hutton M, Lendon CL, Rizzu P et al. (1998). Association of carbohydrate metabolism in cerebrospinal fluid in Rett
missense and 50 -splice-site mutations in tau with the syndrome. J Child Neurol 9: 2630.
inherited dementia FTDP-17. Nature 393: 702705. McCormick WF, Lemmi H (1965). Familial degeneration of
Jankovic J, Kirkpatrick JB, Blomquist KA et al. (1985). the pallidonigral system. Neurology 15: 141153.
Late-onset Hallervorden-Spatz disease presenting as Mnatzakanian GN, Lohi H, Munteanu I et al. (2004). A pre-
familial parkinsonism. Neurology 35: 227234. viously unidentified MECP2 open reading frame defines a
Jellinger K (1986). Pallidal, pallidonigral, and pallidoluysio- new protein isoform relevant to Rett syndrome. Nat Genet
nigral degenerations including association with thalamic 36: 339341.
and dendate degenerations. In: PJ Vinken, GW Brun Mori H, Motoi Y, Kobayashi T et al. (2001). Tau accumula-
(Eds.), Handbook of Clinical Neurology. Elsevier, tion in a patient with pallidonigroluysian atrophy. Neu-
Amsterdam, pp. 445463. rosci Lett 309: 8992.
456 J. C. MOLLER AND W. H. OERTEL

Morris HR, Kanh MN, Janssen JC et al. (2001). The genetic Rett A (1977). Cerebral atrophy with hyperammonaemia.
and pathological classification of familial frontotemporal In: PJ Vinken, GW Bruyn (Eds.), Handbook of Clinical
dementia. Arch Neurol 58: 18131816. Neurology. Elsevier, Amsterdam, pp. 305329.
Murakami JW, Courchesne E, Haas RH et al. (1992). Cere- Rinne JO, Daniel SE, Scaravilli F et al. (1994a). Nigral degen-
bellar and cerebral abnormalities in Rett syndrome: a eration in neuroacanthocytosis. Neurology 44: 16291632.
quantitative MR analysis. AJR Am J Roentgenol 159: Rinne JO, Daniel SE, Scaravilli F et al. (1994b). The neuro-
177183. pathological features of neuroacanthocytosis. Mov Disord
Naidu S, Wong DF, Kitt C et al. (1992). Positron emission 9: 297304.
tomography in the Rett syndrome: clinical, biochemical Rosso SM, van Swieten JC (2002). New developments in
and pathological correlates. Brain Dev 14: S75S79. frontotemporal dementia and parkinsonism linked to chro-
Najim al-Din AS, Wriekat A, Mubaidin A et al. (1994). mosome 17. Curr Opin Neurol 15: 423428.
Pallido-pyramidal degeneration, supranuclear upgaze Rutledge JN, Hilal SK, Silver AJ et al. (1987). Study of
paresis and dementia: Kufor-Rakeb syndrome. Acta movement disorders and brain iron by MR. AJR Am J
Neurol Scand 89: 347352. Roentgenol 149: 365379.
Nardone R, Lochner P, Tezzon F (2003). Hemiparkinson- Sakai T, Mawatari S, Iwashita H et al. (1981). Choreoa-
hemiatrophy syndrome: a transcranial magnetic stimulation canthocytosis. Clues to clinical diagnosis. Arch Neurol
study. Electromyogr Clin Neurophysiol 43: 235240. 38: 335338.
Schroder R, Watts GD, Mehta SG et al. (2005). Mutant valo-
Nisipeanu P, Kuritzky A, Korczyn AD (1994). Familial
sin-containing protein causes a novel type of frontotem-
levodopa-responsive parkinsonian-pyramidal syndrome.
poral dementia. Ann Neurol 57: 457461.
Mov Disord 9: 673675.
Seitelberger F (1957). Zur Morphologie und Histochemie der
Paviour DC, Lees AJ, Josephs KA et al. (2004). Front-
degenerativen Axonveranderungen vom Zentralnervensys-
otemporal lobar degeneration with ubiquitin-only-
tem. Proceedings of the 3rd International Congress of
immunoreactive neuronal changes: broadening the clinical
Neuropathology 12: 7147.
picture to include progressive supranuclear palsy. Brain
Sekul EA, Moak JP, Schultz RJ et al. (1994). Electrocardio-
127: 24412451.
graphic findings in Rett syndrome: an explanation for sud-
Penfield W, Robertson JSM (1943). Growth asymmetry due den death? J Pediatr 125: 8082.
to lesions of the post central cortex. Arch Neurol Psychia- Serra S, Xerra A, Scribano E et al. (1987). Computerized
try 50: 405430. tomography in amyotrophic choreo-acanthocytosis.
Percy AK, Glaze DG, Schultz RJ et al. (1994). Rett Neuroradiology 29: 480482.
syndrome: controlled study of an oral opiate antagonist, Sethi KD, Adams RJ, Loring DW et al. (1988). Hallervor-
naltrexone. Ann Neurol 35: 464470. den-Spatz syndrome: clinical and magnetic resonance
Plioplys AV, Kasnicka I (1993). L-carnitine as a treatment imaging correlations. Ann Neurol 24: 692694.
for Rett syndrome. South Med J 86: 14111412. Shahbazian MD, Zoghbi HY (2001). Molecular genetics of
Pradat PF, Dupel-Pottier C, Lacomblez L et al. (2001). Case Rett syndrome and clinical spectrum of MECP2 muta-
report of pallido-pyramidal disease with supplementary tions. Curr Opin Neurol 14: 171176.
motor area involvement. Mov Disord 16: 762764. Spitz MC, Jankovic J, Killian JM (1985). Familial tic
Pramstaller PP, Kunig G, Leenders K et al. (2002). Parkin disorder, parkinsonism, motor neuron disease and acantho-
mutations in a patient with hemiparkinsonism-hemiatro- cytosis: a new syndrome. Neurology 35: 366370.
phy: a clinical-genetic and PET study. Neurology 58: Storch A, Kornhass M, Schwarz J (2005). Testing for
808810. acanthocytosis A prospective reader-blinded study in
Przedborski S, Goldman S, Levivier M et al. (1993). Brain movement disorder patients. J Neurol 252: 8490.
glucose metabolism and dopamine D2 receptor analysis Swaiman KF (1991). Hallervorden-Spatz syndrome and
in a patient with hemiparkinsonism-hemiatrophy syndrome. brain iron metabolism. Arch Neurol 48: 12851293.
Mov Disord 8: 391395. Swaiman KF, Smith KA, Trock GL et al. (1983). Sea-blue
Przedborski S, Giladi N, Takikawa S et al. (1994). Metabolic histiocytes, lymphocytic cytosomes and 59Fe-studies in
topography of the hemiparkinsonism-hemiatrophy syn- Hallervorden-Spatz syndrome. Neurology 33: 301305.
drome. Neurology 44: 16221628. Swash M, Schwartz MS, Carter ND et al. (1983). Benign X-
Rampoldi L, Dobson-Stone C, Rubio JP et al. (2001). A con- linked myopathy with acanthocytes (McLeod syndrome).
served sorting-associated protein is mutant in chorea- Its relationship to X-linked muscular dystrophy. Brain
acanthocytosis. Nat Genet 28: 119120. 106: 717733.
Remy P, Hosseini H, Degos JD et al. (1995). Striatal dopa- Tanaka M, Hirai S, Kondo S et al. (1998). Cerebral hypoper-
minergic denervation in pallidopyramidal disease demon- fusion and hypometabolism with altered striatal signal
strated by positron emission tomography. Ann Neurol intensity in chorea-acanthocytosis: a combined PET and
38: 954956. MRI study. Mov Disord 13: 100107.
Rett A (1966). [On an until now unknown disease of a con- Taylor TD, Litt M, Kramer P et al. (1996). Homozygosity
genital metabolic disorder.] Krankenschwester 19: mapping of Hallervorden-Spatz syndrome to chromosome
121122. 20p12.3p13. Nat Genet 14: 479481.
OTHER DEGENERATIVE PROCESSES 457

The Rett Syndrome Diagnostic Criteria Work Group Wszolek ZK, Pfeiffer RF, Bhatt MH et al. (1992). Rapidly
(1988). Diagnostic criteria for Rett syndrome. Ann Neurol progressive autosomal dominant parkinsonism and demen-
23: 425428. tia with pallido-ponto-nigral degeneration. Ann Neurol 32:
Thomas M, Hayflick SJ, Jankovic J (2004). Clinical hetero- 312320.
geneity of neurodegeneration with brain iron accumul- Wszolek ZK, Uitti RJ, Hutton M (2000). A mutation in the
ation (Hallervorden-Spatz syndrome) and pantothenate microtubule-associated protein tau in pallido-nigro-luysian
kinase-associated neurodegeneration. Mov Disord 19: degeneration. Neurology 54: 20282030.
3642. Wszolek ZK, Tsuboi Y, Farrer M et al. (2003). Hereditary
Tomemori Y, Ichiba M, Kusumoto A et al. (2005). A gene- tauopathies and parkinsonism. Adv Neurol 91: 153163.
targeted mouse model for chorea-acanthocytosis. J Neuro- Yagishita S, Itoh Y, Nakano T et al. (1983). Pleomorphic
chem 92: 759766. intra-neuronal polyglucosan bodies mainly restricted to
Tranchant C, Boulay C, Warter JM (1991). [Pallido-pyrami- the pallidium. A case report. Acta Neuropathol (Berl) 62:
dal syndrome: an unrecognized entity]. Rev Neurol (Paris) 159163.
147: 308310. Yamamoto T, Hirose G, Shimazaki K et al. (1982). Move-
Ueno S, Maruki Y, Nakamura M et al. (2001). The gene ment disorders of familial neuroacanthocytosis syndrome.
encoding a newly discovered protein, chorein, is mutated Arch Neurol 39: 298301.
in chorea-acanthocytosis. Nat Genet 28: 121122. Yamamoto T, Kawamura J, Hashimoto S et al. (1991). Palli-
van Bogaert L (1947). Aspects cliniques et pathologiques des do-nigro-luysian atrophy, progressive supranuclear palsy
atrophies pallidales et pallido-luysiennes progressives. and adult onset Hallervorden-Spatz disease: a case of aki-
J Belge Neurol Psychiatr 47: 268286. nesia as a predominant feature of parkinsonism. J Neurol
Vercueil L, Hammouti A, Andriantseheno ML et al. (2000). Pal- Sci 101: 98106.
lido-Luysio-Nigral atrophy revealed by rapidly progressive Yasuda M, Kawamata T, Komure O et al. (1999). A muta-
hemidystonia: a clinical, radiologic, functional, and neuro- tion in the microtubule-associated protein tau in pallido-
pathologic study. Mov Disord 15: 947953. nigro-luysian degeneration. Neurology 53: 864868.
Walker RH, Morgello S, Davidoff-Feldman B et al. Young SG, Bertics SJ, Curtiss LK et al. (1987). Genetic ana-
(2002). Autosomal dominant chorea-acanthocytosis with lysis of a kindred with familial hypobetalipoproteinemia.
polyglutamine-containing neuronal inclusions. Neurology Evidence for two separate gene defects: one associated
58: 10311037. with an abnormal apolipoprotein B species, apolipopro-
Watts GD, Wymer J, Kovach MJ et al. (2004). Inclusion tein B-37; and a second associated with low plasma con-
body myopathy associated with Paget disease of bone centrations of apolipoprotein B-100. J Clin Invest 79:
and frontotemporal dementia is caused by mutant 18421851.
valosin-containing protein. Nat Genet 36: 377381. Zappella M, Genazzani A, Facchinetti F et al. (1990). Bro-
Wigboldus JM, Bruyn GW (1968). Hallervorden-Spatz dis- mocriptine in the Rett syndrome. Brain Dev 12: 221225.
ease. In: PJ Vinken, GW Bruyn (Eds.), Diseases of the Zhou B, Westaway SK, Levinson B et al. (2001). A novel
Basal Ganglia: Handbook of Clinical Neurology. Elsevier, pantothenate kinase gene (PANK2) is defective in Haller-
Amsterdam, pp. 604631. vorden-Spatz syndrome. Nat Genet 28: 345349.
Wooten GF, Lopes MB, Harris WO et al. (1993). Pallidoluy- Zoghbi HY, Milstien S, Butler IJ et al. (1989). Cerebrospinal
sian atrophy: dystonia and basal ganglia functional anat- fluid biogenic amines and biopterin in Rett syndrome. Ann
omy. Neurology 43: 17641768. Neurol 25: 5660.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 55

Hydrocephalus and structural lesions

JOHN G. L. MORRIS1*, BRIAN OWLER2, MARIESE A. HELY1 AND VICTOR S. C. FUNG1

1
Department of Neurology, 2Department of Neurosurgery,
Westmead Hospital, Sydney, NSW, Australia

55.1. Introduction and subdural hematoma. He had impaired con-


sciousness which worsened after this procedure
One of the first clues implicating the substantia nigra in and improved with drainage of the lateral ventri-
the pathogenesis of parkinsonism was the finding by cles. CSF pressure by lumbar puncture (LP)
Blocq and Marinesco (1894) of a mass in that part of the remained normal (< 200 mm of water) through-
midbrain in a patient with contralateral hemiparkinson- out his hospital stay. The second patient was a
ism. Yet structural lesions of the brain are rarely asso- 52-year-old trombone player with a 1-year his-
ciated with parkinsonism. The clinical presentation of tory of progressive apathy, slowness of thinking,
such lesions may however mimic those of parkinsonism, unsteadiness of gait and incontinence of urine.
leading to diagnostic confusion. In this chapter, we discuss CSF pressure was normal and it was noted that
the syndrome of normal-pressure hydrocephalus (NPH) the patient improved transiently after lumbar
and how it relates to parkinsonism. We also discuss puncture. A lumbar air encephalogram and right
parkinsonism in the setting of structural lesions such as occipital ventriculogram showed generalized
arteriovenous malformations (AVMs) and brain tumors. enlargement of the ventricular system. Following
the insertion of a ventriculoatrial shunt, his clin-
55.2. Normal-pressure hydrocephalus ical state again improved and this was sustained.
The third case was a 43-year-old man with fluctu-
To most neurologists, the syndrome of NPH is hard to ating conscious level associated with communi-
understand and disappointing to treat. This is a paradoxi- cating hydrocephalus complicating a major
cal disorder: the ventricles of the brain enlarge, yet head injury. CSF pressures were normal but
the pressure within is not increased. Lowering the he was noted to improve after lumbar puncture.
(normal) pressure in the ventricles by inserting a shunt He eventually showed gradual improvement
may improve symptoms, yet the size of the ventricles is following ventriculoatriostomy.
usually unchanged (Meier and Mutze, 2004). To under-
stand this curious disorder it is useful to start with the
The authors addressed the question of why the ven-
seminal descriptions written over 40 years ago.
tricles were enlarged in the face of normal CSF pres-
sure and suggested that the hydrocephalus was due to
55.2.1. Historical aspects
a transient period of raised ventricular pressure and
that, once expanded, the size of the ventricles was
In 1965, Hakim and Adams described 3 patients with
maintained by CSF pressures lower than that which
hydrocephalus associated with normal cerebrospinal
caused the dilatation in the first place. This has been
fluid (CSF) pressure.
explained by invoking the laws of physics propounded
The first patient was a 16-year-old with commu- by the French mathematicians Blaise Pascal (1623
nicating hydrocephalus (demonstrated on air 1662) and Pierre Simon de la Place (17491827).
encephalography) following a severe head injury According to Pascals law, the pressure applied to an

*Correspondence to: Professor John Morris, Department of Neurology, Westmead Hospital, Sydney, NSW 2145, Australia.
E-mail: jmorris@mail.usyd.edu.au, Tel: 61 02-9845-6793, Fax: +61 02-9635-6684.
460 J. G. L. MORRIS ET AL.
enclosed fluid is transmitted undiminished to every obstructive hydrocephalus due to a cyst of the
portion of the fluid and to the walls of the containing third ventricle. Lumbar CSF pressures were
vessel and, according to LaPlaces law, the larger the normal. He recovered after a Torkildsen (ventri-
vessel radius, the larger the wall tension required to culocisternostomy) shunting procedure.
withstand a given internal pressure. The force exerted
on the wall of a fluid-containing vessel represents the
It is important to note that the cognitive impairment
product of pressure times surface area. This explains
in cases 1 and 2 was mild until they had air encephalo-
the everyday experience that, when blowing up a
grams. The benefit of withdrawing spinal fluid on the
childs balloon, inflation occurs maximally in the body
clinical state before embarking on shunting was again
of the balloon rather than the neck. Thus, a given pres-
recorded. The authors contrasted their cases, who had
sure of CSF would be expected to have a greater force
early loss of balance, with patients with Alzheimers
when applied to the walls of enlarged ventricles than
disease, where disturbance of gait is a late feature. They
smaller ones. A pressure of 180 mm of water could
noted that the ventricles also enlarge in Alzheimers
be regarded as pathological in the presence of dilated
disease due to wasting of brain tissue (hydrocephalus
ventricles and lowering the pressure might have a ben-
ex vacuo) and correctly anticipated that differentiation
eficial effect. As we will discuss later, there are rea-
of Alzheimers disease and normal-pressure hydroce-
sons for not accepting this approach as it applies to
phalus is a problem that will recur. They noted that
the ventricles of the brain.
raised intracranial pressure, even up to 700 mm of water,
In a second paper in the same year (Adams et al.,
is often well tolerated in conditions, such as pseudotumor
1965), the authors described a further 3 cases who
cerebri (idiopathic/benign intracranial hypertension),
having become helplessly demented . . . regained full
where the ventricles are small, and suggested that the triad
mental function as a result of a shunt that reduced
of features which they described slowness of thought,
the normal pressure to even lower levels.
incontinence of the bladder and gait disturbance were
due to compression of the frontal lobes by the anterior
Case 1 was a 60-year-old woman with a 6-month
horns of the lateral ventricles. Concerning the lower-limb
history of forgetfulness, unsteadiness of gait and
hyperreflexia, they noted that the long fibers from the leg
urinary incontinence. She was noted on exami-
area of the primary motor cortex descend around the
nation to be gay and witty but her history
dilated ventricle and undergo the greatest stretching
was rambling, her stride was shortened and the
(Yakovlev, 1947). That enlargement was maximal in the
tendon reflexes, particularly in her legs, were
lateral ventricles was attributed to this being the largest
brisk. Her CSF pressure was 175 mm. Following
part of the ventricular system.
a lumbar air encephalogram, which showed
These two papers aroused great interest, not least
massive enlargement of the entire ventricular
because here was another possibly treatable cause of
system, she developed akinetic mutism. Her
dementia. Experience, however, has modified this view.
CSF pressures now rose to 300 mm water. She
remained in this state for several weeks and then
improved greatly after insertion of a ventricu- 55.2.2. Clinical features
loatrial shunt. That this improvement was
related to the shunt was supported by a marked Hakim and Adams papers encouraged neurologists to
deterioration which followed blockage of the look for hydrocephalus in patients with the triad of cog-
shunt after a fall. Case 2 was a 66-year-old nitive impairment, incontinence and gait disturbance,
woman with a history of forgetfulness and falls particularly when the hazardous procedure of air ence-
over a period of a few months. She was pleasant phalography was replaced by computed tomography
and gracious on examination but her memory (CT) and magnetic resonance imaging (MRI) scanning.
was poor. Reflexes were brisk and her stride Results of shunting were often disappointing. Numer-
length was reduced. Following an air encepha- ous papers have been written and opposing positions
logram, which showed gross dilatation of the taken (Bret et al., 2002). Editorials abound (Vanneste,
entire ventricular system, she became drowsy, 1994; Bradley, 2001; Silverberg, 2004). In one review,
incontinent, unable to walk and mute. CSF pres- 535 articles were retrieved from Medline (Hebb and
sure was 200 mm. She remained in this state for Cusimano, 2001). As a result of these studies, there is
some weeks but improved greatly after insertion now a much greater understanding of the sort of patient
of a ventriculoatrial shunt. Case 3 was a who is likely to benefit from shunting.
62-year-old man with slowness of thought, The syndrome of NPH now refers to patients with
unsteadiness of gait and incontinence from a characteristic disturbance of gait and balance
HYDROCEPHALUS AND STRUCTURAL LESIONS 461
(Fisher, 1982) which is improved by shunting of an hydrocephalus as these patients may present with
acquired, usually communicating, hydrocephalus. similar problems of gait, balance and cognition as
Impairment of cognition and incontinence of urine those with NPH but their underlying pathophysiology
may also be present. Most importantly, and contrary and management are different. For completeness,
to the interpretation widely placed on the original mention should also be made of acute/obstructive
two publications, NPH is not a treatable cause hydrocephalus where the main presenting features of
of dementia where this is the major, sole or initial headache, nausea, vomiting and visual disturbance
presenting problem (Vanneste, 1994). relate to raised intracranial pressure.
Idiopathic and secondary forms of NPH are recog- The main clinical features are discussed below.
nized. About half the cases are idiopathic with onset
most commonly in the sixth decade of life or later.
Symptomatic NPH, due to prior subarachnoid hemor- 55.2.2.1. Gait
rhage, meningitis or head trauma, presents at a younger Disturbance of gait is the principal symptom of NPH
age (Bradley, 2000). (Fisher, 1982). Of 30 patients studied by Miller
NPH must be differentiated from chronic or Fisher (Fisher, 1982), all of whom had responded to
arrested hydrocephalus, which is often non-communi- shunting or CSF removal, gait disturbance was the
cating, develops early in life but may not present until only manifestation of the disorder in 14. Ten patients
adulthood. It is not uncommonly picked up as an also had cognitive impairment but in no case did this pre-
incidental finding in a patient being investigated for cede the gait disturbance. Features of the gait included
unrelated symptoms. Here the hydrocephalus is com- staggering, falls, shuffling and slow, multistepped, pre-
pensated. The head is large and, on imaging, the cere- carious turning. Although weakness which worsened on
bral mantle attenuated (Fig. 55.1.) and the CSF exercise was a common symptom, there was no weakness
pressure is usually normal. These patients can decline, on formal examination and movement of the legs on the
usually in their 40s and 50s, probably due to the effect bed was normal. Fisher regarded it as a frontal gait
of aging in a brain with decreased neuronal reserve. It apraxia. Eleven patients had grasp or sucking reflexes.
is important to measure the diameter of the skull in Estanol (1981), in a study of 6 patients with commu-
nicating hydrocephalus, noted that patients, who could
readily make cycling movements with their legs on the
bed, locked and were barely able to walk as soon as
they bore weight on their legs. He described the patient
with this disorder as hopelessly glued to the floor,
unable to take a step. In attempting to walk he might
shuffle with short steps. His equilibrium was poor and
he might fall while attempting to move. Estanol viewed
the gait disturbance as a limb-kinetic apraxia of gait
induced by proprioceptive stimuli to the feet on standing.
He also noted that, although these patients had signs of
frontal lobe dysfunction such as grasp reflexes and perse-
veration in the Luria fist, edge and palm test, they did
not have ideomotor apraxia in the upper limbs.
Vanneste (1994) took issue with the term gait
apraxia, as it applies to NPH, on the grounds that
patients with this disorder execute nearly intact walking
movements when minimally supported or lying down
(though the ability to perform a motor sequence in one
setting but not another is surely one of the hallmarks of
apraxia). Features of the gait which he emphasized
included difficulty in initiating gait, postural instability
and shuffling. Hyperreflexia and extensor plantar
responses were often present. A similarity with the gait
seen in subcortical arteriosclerotic encephalopathy
Fig. 55.1. Computed tomography scan showing a cortical (Thompson and Marsden, 1987) was commented upon.
rim with gross enlargement of the lateral ventricles in a In a definitive review, Nutt et al. (1993) discussed
28-year-old female with a greatly enlarged head. the gait, which has variously been called frontal gait
462 J. G. L. MORRIS ET AL.
disorder, gait apraxia, frontal ataxia, marche petit pas, Clues as to where NPH fits into this scheme are
senile gait, lower-half parkinsonism and arteriosclerotic provided by a study comparing the gait disorder of
parkinsonism. They characterized the frontal gait disor- NPH with that of PD (Stolze et al., 2001). In both dis-
der as follows: variable base (narrow to wide), short orders, gait velocity was reduced due to a diminished
steps, shuffling, start and turn hesitation, and . . . dise- and variable stride length. In NPH, unlike PD, the gait
quilibrium. Widening of the base, upright posture and was broad-based and arm-swing relatively preserved.
preservation of arm-swing were features, when present, Although gait was markedly improved in PD by exter-
which helped to distinguish it from a parkinsonian gait. nal cues such as a metronome and floor stripes, this
Festination, retropulsion and propulsion were more was less apparent in NPH. The gait of NPH conforms
suggestive of parkinsonism than a frontal gait disorder. with the equilibrium apraxia designation of Liston and
The authors also disliked the term apraxia, noting that colleagues (2003), perhaps reflecting disconnection of
these patients usually have little evidence of apraxia the PMA in the periventricular white matter, though
in the upper limbs. Invoking Hughlings Jacksons sen- there are other possibilities (see below).
sorimotor hierarchy, they preferred the designation
frontal gait disorder under the general heading of 55.2.2.2. Incontinence
highest-level equilibrium and locomotor disturbance.
Urinary incontinence is a late sign of NPH (Vanneste,
(The other types of highest-level gait disturbance
1994). In Miller Fishers series (Fisher, 1982) of 30
listed by Nutt and colleagues are cautious gait, subcor-
patients, bladder symptoms were present in 12.
tical disequilibrium, frontal disequilibrium and isolated
Urgency and frequency were the early features leading
gait ignition failure.) Whatever label is applied, this
to incontinence in 8 patients. Cystometrograms showed
type of gait disturbance is most commonly associated
strong contractions to increments in volume as small as
with lesions of the frontal lobes, particularly the mesial
20 ml. In no patient was incontinence the only manifes-
parts (Ahlberg et al. 1988).
tation of NPH. Urodynamic studies in another series
Liston and colleagues (2003) reviewed the role of
(Ahlberg et al., 1988) also found hyperreflexia and
the frontal lobe in movement, dividing it into three
detrusor instability but without evidence of defective
main anatomical components: (1) primary motor cor-
sphincter control. Incontinence sans gene (unembar-
tex, which controls muscle force and direction of move-
rassed incontinence), where there is a lack of concern
ment; (2) premotor area (PMA), which couples motor
at incontinence due to frontal dementia, is not a feature
acts to environmental (external) cues; and (3) supple-
of NPH (Vanneste, 1994). Urinary symptoms in NPH
mentary motor area (SMA), which is involved in motor
are attributed to damage to the periventricular pathways
preparation and the execution of complex voluntary
to the sacral bladder center (Ahlberg et al., 1988).
movements and is internally cued by output from the
basal ganglia. In parkinsonism, it is postulated that gait
ignition failure, bradykinesia and freezing result from 55.2.2.3. Cognitive changes
disordered internal cueing of the SMA by the basal In Miller Fishers series (Fisher, 1982), the changes in
ganglia. Movement may be initiated or improved by mentation were usually difficult to separate from the
external cues. Patients with vascular highest-level gait changes that might occur in anyone in their seventies
disorders may have three types of gait abnormality: or eighties. In one patient, there was profound memory
disturbance (but this was in the era before MRI, so there
1. Ignition apraxia, characterized by gait ignition may have been other reasons for this). Cognitive impair-
failure, difficulty with turns and freezing, is asso- ment is usually mild, with forgetfulness, apathy, inatten-
ciated with infarcts in the basal ganglia/thalamus/ tion, decreased speed of information-processing and
SMA connections in the periventricular white impaired ability to manipulate acquired knowledge
matter. These patients have a failure of internal (Thomsen et al., 1986; Vanneste, 1994). Features of cor-
cueing and are helped by external cues, as with tical impairment, such as aphasia, apraxia and agnosia,
Parkinsons disease (PD). are absent. In further contradistinction to Alzheimers
2. Equilibrium apraxia, where the main problem is disease, but similar to PD, delayed recall may be mark-
loss of balance, is associated with infarcts in the edly impaired whereas delayed recognition is preserved
sensory/PMA pathways or in their connections in (Vanneste, 1994). Iddon and colleagues (1999) noted
the periventricular white matter. Such patients impairment in executive function involving reasoning,
are not helped by external cues. anticipation, goal establishment, strategy formation,
3. Mixed-gait apraxia, characterized by gait ignition shifting mental set and error monitoring in patients with
failure and disequilibrium, is associated with lesions NPH. These pointed to impaired function of the prefron-
affecting the connections of both SMA and PMA. tal cortex. Although improvement was noted in some
HYDROCEPHALUS AND STRUCTURAL LESIONS 463
aspects of cognition following shunting, patients who
fulfilled the criteria of dementia remained significantly
impaired after shunting.
In summary, where dementia is the major problem,
causes other than NPH need to be considered, and the
benefits to the patient from shunting are disappointing
(Iddon et al., 1999; Savolainen et al., 2002).

55.2.2.4. Investigations
55.2.2.4.1. Computed tomography/magnetic
resonance imaging
The diagnosis of NPH hinges upon finding ventricu-
lar enlargement out of proportion to cerebral atrophy
(Vanneste, 1994) in the appropriate clinical setting.
On CT scanning there is enlargement of the ventricu-
lar system without significant gyral atrophy
(Fig. 55.2). This is in contrast to the ventricular enlar-
gement seen in cerebral atrophy associated with
degenerative disease such as Alzheimers disease
(Fig. 55.3). In NPH there is rounding of the frontal
horns and enlargement of the temporal horns without
hippocampal atrophy, as shown by dilatation of the
perihippocampal fissures (Silverberg, 2004). In Alzhei- Fig. 55.3. Computed tomography scan showing ventricular
mers disease, the perihippocampal fissures are typically enlargement with frontal and perisylvian atrophy.
markedly dilated (Figs. 55.4 and 55.5). Measurement of
the cross-sectional volume of the hippocampus on MRI
may be useful in further distinguishing NPH from
Alzheimers disease (Golomb et al., 1994).

Fig. 55.4. Axial magnetic resonance imaging scan (flare


sequence) showing ventricular enlargement and perisylvian
Fig. 55.2. Enlargement of the ventricular system without atrophy, with frontal and occipital periventricular lucencies
gyral atrophy. (ependymitis granularis a normal finding).
464 J. G. L. MORRIS ET AL.
Periventricular lucencies on CT are a normal find-
ing around the frontal and occipital horns but are
accentuated in NPH (Figure 55.4). On MRI fluid-atte-
nuated inversion recovery (FLAIR) sequences, these
findings are more marked and in NPH may extend
around the entire perimeter of the lateral ventricles.
These changes are thought to result from transependy-
mal CSF leakage. Deep white-matter lesions, which
are discontinuous with the ventricular wall, are usually
due to infarcts (Fig. 55.6) or perivascular (Virchow
Robin) spaces. Small white-matter infarcts are very
common in the elderly and even more common in
the setting of NPH (Bradley et al., 1991), where their
presence may predispose to ventricular dilatation by
softening the brain parenchyma (see below) (Bradley,
2001). The dilemma for the clinician is that, if the
patients symptoms are due to white-matter infarcts,
shunting would not be expected to improve matters.
Fig. 55.5. Coronal magnetic resonance imaging scan (T2- On the other hand, patients with such lesions may still
weighted) showing ventricular dilatation, perisylvian atrophy benefit from the procedure (Tullberg et al., 2002), sug-
and dilatation of the perihippocampal fissures. gesting that there is a reversible component in the
pathophysiology of their symptoms (see below).

Fig. 55.6. (A) Axial magnetic resonance imaging (MRI) scan (fluid-attenuated inversion recovery (FLAIR) sequence),
showing enlarged lateral ventricles and perisylvian fissures, frontal and occipital periventricular lucencies, high signal inten-
sity lesions (due to probable gliosis not VirchowRobin spaces) and a low signal intensity lesion in the periventricular
white matter but not contiguous with the ventricular wall. (B) Axial MRI scan (T2-weighted), a little lower, in the same
patient as in (A).
HYDROCEPHALUS AND STRUCTURAL LESIONS 465
T2-weighted images may show a CSF flow void sign Rcsf represents the CSF pressure that must be applied
(Bradley et al., 1986) due to increased CSF flow velo- to the CSF system to produce an absorption rate of 1
city in the aqueduct in NPH, but the value of this sign ml of CSF per minute at equilibrium. The normal Rcsf
is disputed (Vanneste, 1994). is < 10 mmHg/ml per min (Albeck et al., 1991) but is
usually raised in NPH. The Copenhagen Symposium
55.2.2.4.2. Cerebrospinal fluid tap test
on NPH in 1990 concluded that an Rcsf > 11 mmHg/
A test for NPH that is available to all clinicians is the CSF ml per min was suggestive of NPH. Gjerris and Borgesen
tap test. A lumbar puncture is performed using a large- (1992), using the lumbar-ventricular CSF infusion
caliber spinal needle, and, after measuring CSF pressure, method, found that, of 271 patients shunted for NPH,
2040 ml of CSF is removed. If the pressure is high and no patient with an Rcsf < 12 mmHg/ml per min
the patient is relaxed, this might be considered enough responded to CSF shunting whereas 80% with Rcsf
evidence to undertake shunting. If the pressure is normal, >12.5 mmHg/ml per min did. Similar findings have
the decision regarding inserting a shunt is determined by been reported by Lundar and Nornes (1990). In a more
the clinical response to the tap. If the patients gait recent study (Boon et al., 2000) involving 95 patients
improves then the test is positive and the patient should followed for 1 year, it was suggested that the best strat-
undergo shunting. A negative result does not preclude a egy for management of patients thought to have NPH
possible benefit, since, even after implantation of a shunt, was to shunt only those patients with a Rcsf >18
there may be a delay of some days or even a week or two. mmHg/ml per min, or if the Rcsf was lower, only those
Thus, although a positive response is useful, the false- with objective clinical evidence and CT evidence
negative rate is high (Haan and Thomeer, 1988). of NPH.
55.2.2.4.3. Pressure-monitoring and infusion studies
55.2.3. Mechanisms
In specialized centers, overnight CSF pressure monitor-
ing may be done using an Ommaya or Rickham reservoir 55.2.3.1. Hydrocephalus
connected to the lateral ventricle, or a parenchymal intra- Hakim and Adams explanation of ventricular enlarge-
cranial pressure monitor. In normal subjects, waves of ment in the face of normal CSF pressure is no longer
increased pressure may be seen, particularly during accepted. An initial period of raised CSF pressure may
sleep. In NPH, these so-called Lundberg B waves occur occur in some cases of secondary NPH but there is very
with increased amplitude, duration and frequency. little evidence for this in idiopathic NPH. The laws of
Frequent B waves of > 9 mmHg amplitude are thought Pascal and LaPlace, although relevant to a balloon blown
to predict a successful response to CSF shunting up in air, are less applicable to the cerebral ventricles
(Crockard et al., 1977; Borgesen et al., 1979; Reilly, which, as they enlarge, compress the parenchyma of
2001). However their absence does not exclude a diag- the brain against the skull wall, increasing rather than
nosis of NPH or beneficial response to shunting. decreasing resistance to further expansion.
CSF dynamics can also be investigated with CSF Could enlargement of the ventricles be due to the
infusion or perfusion studies. A number of different intermittent rise in pressure that occurs with B waves?
methods have been described, including constant-rate, Probably not, for these reflect alterations in cerebral
constant-pressure and bolus methods. We (BO) have blood volume within the brain.
used the constant-rate CSF infusion study, as There is an increase in the resistance to CSF outflow
described by Czosnyka et al. (1996). This test can be or absorption in NPH. In secondary NPH this is due to
performed via the lumbar route or via a CSF reservoir. obstruction of arachnoid villi or arachnoid adhesions
Two needles are used, one through which CSF pres- resulting from previous subarachnoid hemorrhage or
sure is recorded and another through which normal meningitis. In idiopathic NPH, chronic meningeal thick-
saline is infused at a rate of 1.01.5 ml/min. After a ening has been described at autopsy (DeLand et al.,
period of 10 minutes, during which the baseline CSF 1972; Akai et al., 1987). Patients with NPH often also
pressure is recorded, the CSF infusion is started. CSF exhibit a convexity block when air, contrast or radio-
pressure rises until it reaches a new equilibrium pres- graphic tracers is injected into the subarachnoid space.
sure. The CSF pulse pressure is noted to rise as the Chronic disease and involution of the arachnoid granula-
mean CSF pressure rises. Once a stable equilibrium tions may be seen in NPH (Gilles and Davidson, 1971).
CSF pressure has been established, CSF infusion Akai et al. (1987) noted that the numbers of arachnoid
ceases and CSF pressure is noted to return to its base- villi were decreased in the lateral lacunae of patients
line. The difference between equilibrium and baseline with NPH. It is likely that the disorder of CSF circulation
pressure divided by the infusion rate provides a value in secondary and idiopathic NPH is similar, although in
for the resistance to CSF absorption (Rcsf). In addition, idiopathic NPH it is more gradual in onset.
a number of other parameters can be calculated, Impairment of CSF reabsorption however, would not
including elastance and the pressure volume index. explain why the ventricles enlarge when the pressure
466 J. G. L. MORRIS ET AL.
inside is not increased. There is increasing evidence that the gait disturbance resulted from stretching of axons
it is changes in the viscoelastic properties of brain tissue in the periventricular white matter. As shown in
with age and disease that predispose to ventricular Figures 55.7 and 55.8, the pyramidal fibers closest to
enlargement (Earnest et al., 1974). Hypertension is a the ventricles in the corona radiata are those arising
known risk factor for NPH. Akai et al. (1987) found from the leg area of the motor cortex. Although this
that patients with NPH demonstrated marked sclerosis might account for the hyperreflexia of the lower limbs,
of the small arteries and arterioles of the subependy- it would not explain the gait disturbance which, as
mal region, deep white matter, thalamus and basal described above, has the features of a higher-order gait
ganglia. Small lacunae were frequent in these regions disturbance usually associated with lesions of the
with focal softening of the tissue. Significant vascular mesial frontal lobes. Also in close proximity to the
changes have also been reported at autopsy in patients lateral ventricles is the anterior cerebral artery
with shunt-responsive NPH who died from other (Fig. 55.9A). Stretching of this artery (Fig. 55.9B)
causes (Lorenzo et al., 1974; Newton et al., 1989). may cause frontal lobe ischemia, but no evidence has
been found for this from PET studies (Brooks et al.,
55.2.3.2. Neurological features 1986). Cerebral blood flow (CBF) is maximally
It is not established how chronic hydrocephalus causes impaired in the tissue adjacent to the ventricles,
the associated neurological features. Expansion of the improving progressively with distance from the ventri-
ventricular system could impact on the function of a cles (Momjian et al., 2004). Ischemia of the basal
number of structures. Hakim and Adams proposed that ganglia interfering with the cortico-striato-pallido-tha-
lamo-cortical motor loop has been proposed (Curran
and Lang, 1994) but this might be expected to produce
a gait similar to parkinsonism with improvement with
cues, which is not the case (Stolze et al., 2000). Dis-
connection of the frontal lobes by ischemia of the
medial nuclei of the thalami (Owler et al., 2004)
would account for both the gait and frontal pattern of
cognitive impairment (Fung et al., 1997) in NPH.
Another structure implicated in the neurological fea-
tures of NPH is the posterior corpus callosum, which
is compressed against the falx cerebri as the ventricles
enlarge (Jinkins, 1991). The posterior region of the

Fig. 55.7. For full color figure, see plate section. Coronal
section of the brain showing proximity of the fibers arising
from the mesial part of the motor cortex to the lateral ventricle. Fig. 55.8. The motor homunculus. Reproduced from Penfield
Adapted from Figure 10.178, page 725, Romanes (1981). and Rasmussen (1950), with permission from Macmillan.
HYDROCEPHALUS AND STRUCTURAL LESIONS 467

Fig. 55.9. For full color figure, see plate section. (A) Proximity of the anterior cerebral artery to the lateral ventricle. Adapted
from Duus (1989). (B). Sagittal magnetic resonance imaging scan showing relationship of the anterior cerebral artery to the
enlarged lateral ventricle in normal-pressure hydrocephalus.

corpus callosum contains crossed corticostriate fibers months after surgery it had returned to normal,
and association fibers between the vestibular cortical the difference between the pre- and postshunt
areas. Similarly, involvement of the medial and lateral videos being most striking.
striae as well as the fornix is proposed as a mechanism
for disturbance of memory (Del Bigio, 1993). It is of interest that, although the ultimate result in
this patient was most gratifying, the improvement after
55.2.4. Treatment shunting was not immediate, causing the treating neu-
55.2.4.1. Some observations rologist to wonder in the immediate postoperative per-
The identification and treatment of NPH by shunting can iod whether he had made the right decision in referring
be one of the most rewarding in neurological practice: the patient for surgery. This delay in improvement
may reflect recovery of stretched axons in the vicinity
A 74-year-old man presented with a 67-year of the ventricles. Harder to explain is the improvement
history of difficulty in walking and stooped pos- which follows shunting when the ventricular size
ture, for which he had been taking levodopa, remains unchanged:
without benefit. There were occasional spells
where his feet got stuck and he had fallen on a A 73-year-old man presented with progressive
number of occasions. His wife reported a decline difficulty in walking for a year. Prior to that he
in memory commensurate with her own. For 2 had been able to ski. There was urge incontinence
years he had had urge incontinence. On exami- and no evidence of cognitive impairment. On
nation, his face lacked expression and he walked examination, he struggled to get out of the chair
with a festinating gait on a broadened base and and walked very slowly on a broad base and with
with preserved (rapid) arm-swing. He turned en a tendency to shuffle. He was able to rise from a
bloc. There was mild cogwheel rigidity in the crouching position and could stand on his toes
upper limbs but no bradykinesia. Tendon and heels. Power in the limbs was normal. The
reflexes were present but reduced. Plantars were reflexes were all hard to elicit. Tone was not
flexor. CT scan showed enlarged ventricles with- increased. There was no sensory loss. The arms
out cortical atrophy (Fig. 55.10A). Following were normal. An MRI scan showed marked dila-
insertion of a ventriculoperitoneal (VP) shunt, tation of the ventricles without gyral atrophy
the ventricles reduced in size (Figure 55.10B, (Fig. 55.11). Following ventriculostomy, he
postshunt) but there was only minimal initial became confused, incontinent of urine and bed-
improvement in his gait. Over the next few bound for several days. Over the next month, he
months his gait improved and when videoed 7 slowly improved to the point where he could walk
468 J. G. L. MORRIS ET AL.

Fig. 55.10. Computed tomography scan: dilated ventricles: (A) preshunt; (B) postshunt.

Fig. 55.11. Axial magnetic resonance imaging scan (T1-weighted) showing enlargement of the lateral ventricles: (A) preshunt;
(B) sagittal view.
HYDROCEPHALUS AND STRUCTURAL LESIONS 469

Fig. 55.12. Axial magnetic resonance imaging (T1-weighted) showing enlargement of the lateral ventricles: (A) postshunt; (B)
sagittal view.

unsupported. A postshunt MRI showed no change et al., 1999). Others reported that, although global CBF
in ventricular size (Fig. 55.12). Over the next few was not improved, the pattern of CBF was improved in
months he continued to improve and, when shunt-responders (Waldemar et al., 1993).
reviewed and revideoed after 7 months, he was More recently (Silverberg et al., 2003; Silverberg,
walking normally. 2004), it has been proposed that changes in the normal
Improvement was also delayed in this patient and circulation of CSF in NPH may result in a failure to
no less impressive than the previous patient, yet the clear toxic molecules such as amyloid--peptide, pre-
ventricular size looks much the same. This finding is disposing to the development of Alzheimers disease,
the rule rather than the exception (Meier and Mutze, which has an increased incidence in biopsy-studied
2004). This may reflect the fact that current VP shunts cases of NPH (Golomb et al., 2000). A trial of CSF
have valves in them which prevent the pressure falling shunting to slow the progression of Alzheimers disease
so low as to cause the hemispheres to collapse and is currently underway.
invite the formation of subdural hematomas. In Meier
and Mutzes study, 80% of 80 patients shunted for 55.2.4.2. Shunting
NPH had no change in ventricular size, yet 59% of Shunting is the treatment of choice in NPH. As origin-
these were judged to have made a good to excellent ally suggested by Adams et al. (1965), the aim is to
improvement and a further 17% a satisfactory decrease CSF pressure to even less than normal. The
improvement. Such findings lend support to the response to shunting varies widely between studies:
hypothesis that shunting produces its benefit by means 2580%, with a mean of 50% (Vanneste et al., 1992).
other than relieving stretch on axons adjacent to the Of 1047 patients included in this review, the response
ventricles. One such mechanism is improved blood to shunting was notably better in secondary NPH
flow to the brain parenchyma. (64%) compared to idiopathic NPH (50%). Marked
The response of CBF to shunting has been studied improvement was noted in 46% of patients with second-
using a variety of techniques. Although some investiga- ary NPH compared to 33% with idiopathic NPH.
tors found an increase in CBF after shunting (Tanaka The most common form of CSF shunt used in NPH
et al., 1997), it was often not sustained (Graff-Radford is a VP shunt, although ventriculoatrial shunting is
et al., 1987). Other investigators reported no change in also popular. Both are generally favored over lumbo-
CBF after shunting (Meixensberger et al., 1989) and peritoneal shunting, which often poses problems in
that there was no relationship to outcome (Klinge the elderly.
470 J. G. L. MORRIS ET AL.
55.2.4.2.1. Complications of shunting He improved with VP shunting but later became
The most common complications of shunting are drowsy when the shunt blocked. After shunt revi-
infection and blockage. Less common complications sion, he was noted to have tremor, rigidity and
include subdural hematoma, stroke, shunt disconnec- akinesia and Parinauds syndrome. The parkin-
tion, erosion of the distal catheter through the skin or sonian features persisted and were markedly
internal viscus, thrombosis around atrial catheters and improved by levodopa.
shunt nephritis. Vanneste et al. (1992) performed a Patient 2 was a 16-year-old man with headache,
risk/benefit study of idiopathic NPH using 1047 bilateral sixth-nerve palsies and papilledema
patients obtained from 19 studies from the literature. from hydrocephalus secondary to a pineal mass.
The benefit/harm ratio in patients with idiopathic A VP shunt was inserted and he was given radio-
NPH was 1.7 and increased to 6 if high-risk patients therapy. After repeated episodes of shunt
with significant comorbidity were excluded. This is obstruction requiring revisions, he became bra-
not a procedure to be undertaken lightly. dykinetic and rigid. These features improved
A complication of shunting that deserves special markedly with levodopa.
attention is that of subdural hematoma. This occurs Patient 3, aged 7, developed tremor, unsteadi-
in patients with large ventricles with relatively thin ness of gait, headache, vomiting and listlessness
cortical mantles when there is overdrainage of CSF. associated with aqueduct stenosis. He improved
The cortical mantle collapses, separating away from with a VP shunt. Again, after periods of repeated
the dura and tearing overstretched subdural veins. shunt obstructions, he became parkinsonian with
NPH patients are particularly prone to this complication tremor and rigidity. This too improved with levo-
(Symon et al., 1972). dopa. After some months the levodopa was
withdrawn without return of his parkinsonism.
55.2.4.2.2. Reasons for shunt failure Patient 4 presented at age 9 with headache,
As discussed previously, clinical improvement in patients papilledema and Parinauds syndrome due to
with NPH after CSF shunting is usually not immediate. aqueduct stenosis. Her symptoms improved with
Gait disorder and urinary incontinence are the first symp- a Torkildsen ventriculocisternal shunt. At age
toms to improve and may do so within days but often take 26, she presented again with personality change
several weeks. Improvement in mental function is less and generalized bradykinesia. The ventricles
predictable and is usually the last symptom to improve; were not enlarged but the shunt was noted to
it may take several months. Failure of the shunt to be wrapped around the brainstem and was
provide worthwhile benefit can be due to a number of replaced with a VP shunt. Her parkinsonism per-
factors: (1) technical problems with the pump itself; (2) sisted and was not improved by levodopa. Later
complications of shunting, such as subdural hematoma; a CT scan revealed a small left thalamic bleed.
(3) irreversible pathophysiological changes in the brain An 18F-dopa PET scan was normal, suggesting
parenchyma relating to coexisting disease, such as hyper- that her parkinsonism was postsynaptic.
tensive small-vessel disease or Alzheimers disease. Patient 5 was a 69-year-old man who developed
parkinsonism, shown at postmortem to be due to
55.2.4.3. Drugs and other treatments progressive supranuclear palsy in the setting of
Drugs have little place in the management of NPH. NPH and diffuse cerebrovascular disease.
There is no evidence that reducing CSF production Patient 6, aged 72, was similar in presentation
with drugs such as acetazolamide is beneficial. From to case 5, but with no pathological study.
the preceding discussions, it might be anticipated that Patient 7, aged 72, presented with a 10-year history
control of hypertension may lessen the predisposition of asymmetrical limb slowing, gait disturbance and
to developing NPH. incontinence associated with hydrocephalus. He
improved with shunting but later developed pro-
55.2.5. Association between parkinsonism and gressive parkinsonism with tremor and bradykine-
hydrocephalus sia. This partially improved with levodopa. Later
he developed a vertical supranuclear gaze palsy.
Curran and Lang (1994) have drawn attention to cases At pathology he was found to have typical features
of levodopa-responsive parkinsonism associated with of advanced idiopathic PD.
hydrocephalus. It is helpful to consider these:
Patient 8, aged 68, presented with gait instability,
Patient 1 was a 16-year-old boy with headaches generalized rigidity and right-arm resting tre-
and papilledema secondary to aqueduct stenosis. mor. Within a year he was incontinent and had
HYDROCEPHALUS AND STRUCTURAL LESIONS 471
cognitive changes. A CT scan showed hydroce-
phalus and all his symptoms resolved with a VP
shunt. Three months later his symptoms returned
and he was found to have small bilateral resol-
ving subdural hematomas. He initially improved
with levodopa but his parkinsonism and dementia
continued to decline over the next 2 years. An
MRI scan showed multiple areas of subcortical
high signal change. An 18F-dopa PET scan
showed decreased fluorine accumulation in the
putamen but not the caudate.
Patient 9, aged 67, with a previous history of
severe head injury, presented with parkinsonism
which responded to levodopa. Five years later
she had an episode of confusion and a CT scan
showed enlarged ventricles. Over the next 2
years she became progressively more demented
and parkinsonian.

55.2.5.1. Comment
Cases 13, all young, show that parkinsonism can
occur subacutely as a complication of obstructive Fig. 55.13. Axial T2-weighted magnetic resonance imaging
hydrocephalus involving the aqueduct. In these scan showing high signal intensity changes in the right tem-
poral lobe.
patients, the clinical picture was very different from
the cases of NPH described above: they had symptoms
and signs of raised intracranial pressure with head- with a seizure, decreased level of consciousness,
ache, drowsiness and papilledema. Their parkinsonism a dilated right pupil and left hemiparesis from a
may have resulted from direct injury to the substantia large right temporal intracerebral hemorrhage.
nigra, as there was evidence of midbrain damage The hemorrhage was evacuated together with a
(patient 1 had Parinauds syndrome and patient 2 com- small AVM. His signs included bradykinesia,
pression of the midbrain by a pineal tumor) and they rigidity, start hesitation and poor postural
responded to levodopa. Cases 59, all aged 67 or reflexes, without a resting tremor. He also had
older, illustrate that Occams razor whereby, in a signs of a Parinauds syndrome. CT and MRI
medical context, an attempt should be made to explain of the brain demonstrated changes in the right
all symptoms on the basis of a single disease is often temporal lobe associated with the hemorrhage
not a useful exercise in the elderly, where degenerative but no abnormality of the basal ganglia or mid-
parkinsonian syndromes such as idiopathic PD and brain (Fig. 55.13). Levodopa therapy produced
progressive supranuclear palsy may coexist with a dramatic improvement within a few days of
cerebrovascular disease and NPH. commencement. He still required this treatment
2 years after the bleed. Three years after the
event he died from heat stroke after jogging. At
55.3. Other structural lesions postmortem, marked neuronal loss with gliosis
was observed in the substantia nigra and super-
Dopa-responsive parkinsonism is described below in
ior colliculi. There were no areas of infarction.
association with a number of structural lesions. No Lewy bodies, neurofibrillary tangles or glial
cytoplasmic inclusion bodies were seen (M
55.3.1. Arteriovenous malformation Rodrigues, personal communication).
55.3.1.1. Intracerebral hemorrhage
Like cases 13 of Curran and Lang (1994), it seems
Our colleague Aggarwal (Ling et al., 2002) reported a
likely that the parkinsonism in this case was the result
case of dopa-responsive parkinsonism following a
of damage to the nigrostriatal tract in the midbrain by
hemorrhage into the right temporal lobe from an AVM:
compression, in this case from an expanded temporal
A 46-year-old man developed a symmetrical lobe. Similar cases have been described in association
parkinsonian syndrome 7 weeks after presenting with subdural hematoma (Trosch and Ransom, 1990).
472 J. G. L. MORRIS ET AL.
55.3.1.2. Midbrain cavernoma of levodopa wears off after about 2 hours. When
We have had a patient with dopa-responsive parkin- she is on she has no tremor and her Unified Par-
sonism associated with a cavernoma involving one kinsons Disease Rating Scale (UPDRS) score is
substantia nigra: 5; when she is off it is 30. She has never had dys-
kinesia and never had signs on the other side. MRI
She presented in 1975 at the age of 34 with tremor scans (Fig. 55.14) showed an area of increased
of the left hand. She was put on levodopa, which signal in the right cerebral peduncle with some
abolished her symptoms. When reviewed in 1991, surrounding low signal on T2-weighted images,
she had a coarse resting tremor and mild akinesia suggesting the presence of hemosiderin. No abnor-
confined to the left arm. In follow-up since that mal feeding vessels were seen on magnetic
time, she has reported that the effect of each dose resonance angiography. The features were of

Fig. 55.14. (A) T1-weighted magnetic resonance imaging (MRI) scan showing hypointense lesion in the pars compacta of the
right cerebral peduncle. (B) T2-weighted MRI scan showing low signal area (hemosiderin) around a small high signal area
(methemoglobin)in the right midbrain. (C) Coronal view of (B).
HYDROCEPHALUS AND STRUCTURAL LESIONS 473
is on the appropriate side of the midbrain to produce
hemiparkinsonism. It seems likely that the right
nigrostriatal tract has been damaged by pressure from
the cavernoma or associated bleeding.

55.3.2. Tumor
Fig. 55.15. For full color figure, see plate section. b-Carbo-
methoxy-3b-(4-iodophenyl)tropane (b-CIT) single-photon emis- Tumors rarely, if ever, cause parkinsonism. Some-
sion computed tomography (SPECT) scan showing reduced times, however, the clinical features produced by a
formation of dopamine transporter in the right caudate nucleus. tumor may be mistaken for PD:
A 52-year-old right-handed man presented with a
a cavernoma. SPECT images (Fig. 55.15) were
6-month history of difficulty using his right hand.
obtained using [123I]2b-carboxymethoxy-3b-
His writing had lost fluency and become small.
iodophenyl tropane (beta-CIT) which labels presy-
He had difficulty doing up buttons and getting
naptic dopamine transporter (DAT) and [123I]
his wallet out of his pocket. He had a tendency
iodobenzamide (IBZM), which labels the postsy-
to trip with the right foot. On examination, he
naptic D2-receptors. There was a dramatic reduc-
did not swing the right arm when he walked.
tion in presynaptic dopaminergic binding
There was minimal lag in the right arm compared
ipsilateral to the lesion (Fig. 55.15) that was more
with the left when he raised them and there was
marked in the caudate than putamen, the opposite
no weakness. Tone was increased on the right
of the putamencaudate gradient that is found in
side. He had difficulty performing piano-playing
idiopathic PD. Ipsilateral postsynaptic and con-
movements with the right hand. Reflexes were
tralateral pre- and postsynaptic dopaminergic
brisker on the right side. The plantars were
uptakes were normal.
flexor. A scan was done because of the hyperre-
It is unlikely that this patient has unrelated idio- flexia. This showed a 7  5  4 cm meningioma
pathic PD, as there has been almost no progression in indenting the left cerebral hemisphere maximally
her disability over a period of 30 years. The cavernoma over the motor strip (Fig. 55.16).

Fig. 55.16. (A) Axial postgadolinium T1-weighted magnetic resonance imaging scan showing a large meningioma indenting
the left cerebral hemiphere; (B) T2-weighted, coronal view.
474 J. G. L. MORRIS ET AL.
55.3.2.1. Comment days his speech improved, as did the power on the
Both neurologists who examined this patient at presen- right side. There was reduction in amplitude of
tation were impressed by the abnormality of finger right-hand movements involving opening and
movement of the right side which was interpreted, in closing the fingers and difficulty in making
the absence of weakness, as akinesia due to PD. The piano-playing movements involving the index and
increase in tone and loss of arm-swing lent support middle fingers, similar to what is seen in
to this. It seems likely that abnormality of finger parkinsonism. Most striking, however, was the
movement was due to a limb-kinetic apraxia, which inability to coordinate the movement of the two
may present in a very similar manner to akinesia. hands. He could clap with one hand or the other
but not both simultaneously. All of these features
55.3.3. Lesions of the striatum/globus gradually disappeared over the next few weeks,
pallidus/thalamus leaving him with normal hand function.

In Bhatia and Marsdens (1994) extensive review, iso- 55.3.4.1. Comment


lated lesions of the striatum/globus pallidus rarely, and
The relationship between the SMA and akinesia in
of the thalamus never, caused pure parkinsonism. Par-
parkinsonism has been reviewed by Williams et al.
kinsonism occurred in 9% of 240 cases, all associated
(2002): Activity in the SMA is a major contributor to
with bilateral lesions and only rarely with rest tremor.
the Bereitschaftspotential that precedes self-generated
Most cases followed toxic/metabolic lesions, making
movements and this potential is reduced in PD.
strict exclusion of nigrostriatal pathology difficult.
Imaging studies confirm impaired activation of SMA
during some movements in untreated PD; this is reversi-
55.3.4. Supplementary motor area resection
ble using the dopaminergic agonist apomorphine. It is
of interest therefore that our patient showed features
Patients undergoing surgery involving the motor path-
similar to the akinesia of parkinsonism following SMA
ways can provide insights into the mechanism of the
resection, though the more striking abnormality was in
motor deficit in parkinsonism:
loss of bimanual coordination.
A 24-year-old man with a history of intractable
seizures had the left SMA resected (Fig. 55.16). 55.4. Incidence of structural lesions
There were no clinical signs before surgery. in parkinsonism
Following surgery, he was initially mute and had
right-side weakness. An MRI scan shows the extent Most reports of structural lesions associated with
of the resection (Fig. 55.17). Over the next few parkinsonism are of single cases or of retrospective

Fig. 55.17. (A) T1-weighted magnetic resonance imaging scan: sagittal view showing high signal changes (due to blood pro-
ducts) over the mesial surface of the left cerebral hemisphere; (B) coronal view.
HYDROCEPHALUS AND STRUCTURAL LESIONS 475
studies of hospital records. Some information on the confined to the right side of his body. He was found
incidence of structural lesions in patients presenting to have a cystic lesion in the contralateral thala-
with parkinsonism is provided by the long-term Syd- mus and lentiform nucleus (Fig. 55.19), probably
ney Multicenter study of PD (Hely et al., 1994). Of secondary to a severe head injury at age 13 (for
149 de novo patients diagnosed as having PD by 26 which he underwent bilateral burrhole evacuation
Sydney neurologists and followed by author MAH of subdural hematomas). He responded well to
for up to 20 years, 13 were found in the first 2 years levodopa 300 mg/day and benztropine 15 mg/day
to be atypical, of which structural lesions relevant to and his symptoms remained confined to the right
the parkinsonism were seen in 2: side over 10 years of follow-up.
A 60-year-old man who had presented with fea-
tures of a mixed resting and postural tremor, mild 55.4.2. Comment
but definite rigidity and bradykinesia, developed a
wide-based gait soon after presentation. He was It seems likely that his hemiparkinsonism was related
found to have a pinealoma compressing the mid- to the previous head injury, though it began 21 years
brain and causing hydrocephalus (Fig. 55.18). after the injury. The symptoms were non-progressive
He responded to shunting and did not continue and contralateral to the lesion, which probably repre-
with parkinsonian medication. sented a cavity from an old hematoma. The response
to levodopa was surprising and suggests that the lesion
55.4.1. Comment also involved the nigrostriatal tract.

There were changes in the midbrain on CT scanning


and it seems likely that his transient parkinsonian
55.5. Summary and conclusions
features were due to compression of the substantia
Structural pathology of the brain can mimic the clini-
nigra or nigrostriatal tract by the pinealoma.
cal features of parkinsonism. Those involving the
A 37-year-old man presented with a 3-year history substantia nigra or the nigrostriatal tract can lead to
of resting tremor, bradykinesia and rigidity the classical features associated with idiopathic PD,

Fig. 55.18. (A) Non-contrast computed tomopgraphy (CT) scan showing a calcified mass in the pineal gland. (B) Non-contrast
CT scan in the same patient showing low-density change in the region of the aqueduct.
476 J. G. L. MORRIS ET AL.

Fig. 55.20. For full color figure, see plate section. Diffusion
tensor magnetic resonance imaging scan of a patient with
normal-pressure hydrocephalus showing thinning of the for-
ceps minor (red), forceps major (green) and compression of
the corona radiata (blue).

Fig. 55.19. Non-contrast computed tomography scan show- Acknowledgments


ing hypodense area in the left thalamus.
We thank Drs Somerville, Aggarwal, Bleasel and
Duggins for permission to describe their patients and
including asymmetry, rest tremor, akinesia, rigidity Dr Lavier Gomes for his help with the scans.
and, most importantly, responsiveness to levodopa.
Lesions of the striatum/globus pallidus/thalamus rarely References
cause pure parkinsonism. When it does occur, such
postsynaptic parkinsonism is not usually levodopa- Adams RD, Fisher CM, Hakim S et al. (1965). Symptomatic
responsive. Data from patients with parkinsonism sec- occult hydrocephalus with normal cerebrospinal-fluid pres-
sure. A treatable syndrome. N Engl J Med 273: 117126.
ondary to structural lesions lead to the conclusion that
Ahlberg J, Norlen L, Blomstrand C et al. (1988). Outcome of
the parkinsonism results from loss of dopaminergic
shunt operation on urinary incontinence in normal pres-
modulation of striatal activity, not from loss of func- sure hydrocephalus predicted by lumbar puncture. J Neu-
tion due to lesions of the striatum itself. rol Neurosurg Psychiatry 51 (1): 105108.
NPH produces a clinical picture that is very different Akai K, Uchigasaki S, Tanaka U et al. (1987). Normal pres-
from PD (Fig. 55.20). The major motor impairment is sure hydrocephalus. Neuropathological study. Acta Pathol
gait disturbance, which most closely resembles that Jpn 37 (1): 97110.
seen in multilacunar states or frontal-lobe syndromes. Albeck MJ, Borgesen SE, Gjerris F et al. (1991). Intracranial
As softening of the brain parenchyma by multiple pressure and cerebrospinal fluid outflow conductance in
infarcts predisposes to ventricular enlargement, there is healthy subjects. J Neurosurg 74 (4): 597600.
considerable overlap between NPH and the multilacunar Bhatia KP, Marsden CD (1994). The behavioural and motor
consequences of focal lesions of the basal ganglia in man.
state. Where most of the neurological deficit relates to
Brain 117 (Pt 4): 859876.
infarction, the response to shunting is likely to be poor.
Blocq P, Marinesco G (1894). Sur un cas de tremblement
Finally, cortical lesions involving the motor and Parkinsonien hemiplegique symptomatique dune tumeur
premotor cortex can cause alterations in fine finger du peduncle cerebrale. Rev Neurol (Paris) 2: 265.
movements similar to what is seen in parkinsonism. Boon AJ, Tans JT, Delwel EJ et al. (2000). The Dutch
These are best considered as limb-kinetic apraxia normal-pressure hydrocephalus study. How to select
rather than akinesia as there are no associated signs patients for shunting? An analysis of four diagnostic
such as rigidity or rest tremor. criteria. Surg Neurol 53 (3): 201207.
HYDROCEPHALUS AND STRUCTURAL LESIONS 477

Borgesen SE, Gjerris F, Sorensen SC (1979). Intracranial Gilles FH, Davidson RI (1971). Communicating hydrocepha-
pressure and conductance to outflow of cerebrospinal fluid lus associated with deficient dysplastic parasagittal
in normal-pressure hydrocephalus. J Neurosurg 50 (4): arachnoidal granulations. J Neurosurg 35 (4): 421426.
489493. Gjerris F, Borgesen SE (1992). Current concepts of measure-
Bradley WG (2000). Normal pressure hydrocephalus: new ment of cerebrospinal fluid absorption and biomechanics
concepts on etiology and diagnosis. AJNR Am J Neurora- of hydrocephalus. Adv Tech Stand Neurosurg 19:
diol 21 (9): 15861590. 145177.
Bradley WG (2001). Normal pressure hydrocephalus and deep Golomb J, de Leon MJ, George AE et al. (1994). Hippocam-
white matter ischemia: which is the chicken, and which is pal atrophy correlates with severe cognitive impairment in
the egg? AJNR Am J Neuroradiol 22 (9): 16381640. elderly patients with suspected normal pressure hydroce-
Bradley WG Jr, Kortman KE, Burgoyne B (1986). Flowing phalus. J Neurol Neurosurg Psychiatry 57 (5): 590593.
cerebrospinal fluid in normal and hydrocephalic states: Golomb J, Wisoff J, Miller DC et al. (2000). Alzheimers
appearance on MR images. Radiology 159 (3): 611616. disease comorbidity in normal pressure hydrocephalus:
Bradley WG Jr, Whittemore AR, Watanabe AS et al. (1991). prevalence and shunt response. J Neurol Neurosurg Psy-
Association of deep white matter infarction with chronic chiatry 68 (6): 778781.
communicating hydrocephalus: implications regarding Graff-Radford NR, Rezai K, Godersky JC et al. (1987).
the possible origin of normal-pressure hydrocephalus. Regional cerebral blood flow in normal pressure hydroce-
AJNR Am J Neuroradiol 12 (1): 3139. phalus. J Neurol Neurosurg Psychiatry 50 (12): 15891596.
Bret P, Guyotat J, Chazal J (2002). Is normal pressure hydro- Haan J, Thomeer RT (1988). Predictive value of temporary
cephalus a valid concept in 2002? A reappraisal in five external lumbar drainage in normal pressure hydrocepha-
questions and proposal for a new designation of the syn- lus. Neurosurgery 22 (2): 388391.
drome as chronic hydrocephalus. J Neurol Neurosurg Hakim S, Adams RD (1965). The special clinical problem of
Psychiatry 73 (1): 912. symptomatic hydrocephalus with normal cerebrospinal
Brooks DJ, Beaney RP, Powell M et al. (1986). Studies on fluid pressure. Observations on cerebrospinal fluid hydro-
cerebral oxygen metabolism, blood flow, and blood dynamics. J Neurol Sci 2 (4): 307327.
volume, in patients with hydrocephalus before and after Hebb AO, Cusimano MD (2001). Idiopathic normal pressure
surgical decompression, using positron emission tomogra- hydrocephalus: a systematic review of diagnosis and out-
phy. Brain 109 (Pt 4): 613628. come. Neurosurgery 49 (5): 11661184.
Crockard HA, Hanlon K, Duda EE et al. (1977). Hydrocepha- Hely MA, Morris JG, Reid WG et al. (1994). The Sydney
lus as a cause of dementia: evaluation by computerised multicentre study of Parkinsons disease: a randomised,
tomography and intracranial pressure monitoring. J Neurol prospective five year study comparing low dose bromo-
Neurosurg Psychiatry 40 (8): 736740. criptine with low dose levodopa-carbidopa. J Neurol Neu-
Curran T, Lang AE (1994). Parkinsonian syndromes asso- rosurg Psychiatry 57 (8): 903910.
ciated with hydrocephalus: case reports, a review of the Iddon JL, Pickard JD, Cross JJ et al. (1999). Specific patterns
literature, and pathophysiological hypotheses. Mov Disord of cognitive impairment in patients with idiopathic normal
9 (5): 508520. pressure hydrocephalus and Alzheimers disease: a pilot
Czosnyka M, Whitehouse H, Smielewski P et al. (1996). study. J Neurol Neurosurg Psychiatry 67 (6): 723732.
Testing of cerebrospinal compensatory reserve in shunted Jinkins JR (1991). Clinical manifestations of hydrocephalus
and non-shunted patients: a guide to interpretation based caused by impingement of the corpus callosum on the
on an observational study. J Neurol Neurosurg Psychiatry falx: an MR study in 40 patients. AJNR Am J Neuroradiol
60 (5): 549558. 12 (2): 331340.
Del Bigio MR (1993). Neuropathological changes caused by Klinge PM, Berding G, Brinker T et al. (1999). A positron
hydrocephalus. Acta Neuropathol (Berl) 85 (6): 573585. emission tomography study of cerebrovascular reserve
DeLand FH, James AE Jr, Ladd DJ et al. (1972). Normal before and after shunt surgery in patients with idiopathic
pressure hydrocephalus: a histologic study. Am J Clin chronic hydrocephalus. J Neurosurg 91 (4): 605609.
Pathol 58 (1): 5863. Ling MJ, Aggarwal A, Morris JG (2002). Dopa-responsive
Duus P (1989). Topical Diagnosis in Neurology, 2nd edn, parkinsonism secondary to right temporal lobe haemorrah-
Thieme Medical Publishers, New York. age. Mov Disord 17 (2): 402404.
Earnest MP, Fahn S, Karp JH et al. (1974). Normal pressure Liston R, Mickelborough J, Bene J et al. (2003). A new
hydrocephalus and hypertensive cerebrovascular disease. classification of higher level gait disorders in patients
Arch Neurol 31 (4): 262266. with cerebral multi-infarct states. Age Ageing 32 (3):
Estanol BV (1981). Gait apraxia in communicating hydroce- 252258.
phalus. J Neurol Neurosurg Psychiatry 44 (4): 305308. Lorenzo AV, Bresnan MJ, Barlow CF (1974). Cerebrospinal
Fisher CM (1982). Hydrocephalus as a cause of disturbances fluid absorption deficit in normal pressure hydrocephalus.
of gait in the elderly. Neurology 32 (12): 13581363. Arch Neurol 30 (5): 387393.
Fung VS, Morris JG, Leicester J et al. (1997). Clonic perse- Lundar T, Nornes H (1990). Determination of ventricular
veration following thalamofrontal disconnection: a distinc- fluid outflow resistance in patients with ventriculomegaly.
tive movement disorder. Mov Disord 12 (3): 378385. J Neurol Neurosurg Psychiatry 53 (10): 896898.
478 J. G. L. MORRIS ET AL.

Meier U, Mutze S (2004). Correlation between decreased which parameters respond to the CSF tap test? Clin
ventricular size and positive clinical outcome following Neurophysiol 111 (9): 16781686.
shunt placement in patients with normal-pressure hydroce- Stolze H, Kuhtz-Buschbeck JP, Drucke H et al. (2001).
phalus. J Neurosurg 100 (6): 10361040. Comparative analysis of the gait disorder of normal pres-
Meixensberger J, Brawanski A, Ullrich OW et al. (1989). sure hydrocephalus and Parkinsons disease. J Neurol
Cerebral blood flow in low pressure hydrocephalus. Psy- Neurosurg Psychiatry 70 (3): 289297.
chiatry Res 29 (3): 307308. Symon L, Dorsch NW, Stephens RJ (1972). Pressure waves in
Momjian S, Owler BK, Czosnyka Z et al. (2004). Pattern of so-called low-pressure hydrocephalus. Lancet 2 (7790):
white matter regional cerebral blood flow and autoregula- 12911292.
tion in normal pressure hydrocephalus. Brain 127 (Pt 5): Tanaka A, Kimura M, Nakayama Y et al. (1997). Cerebral
965972. blood flow and autoregulation in normal pressure hydroce-
Newton H, Pickard JD, Weller RO (1989). Normal pressure phalus. Neurosurgery 40 (6): 11611165.
hydrocephalus and cerebrovascular disease: findings of Thompson PD, Marsden CD (1987). Gait disorder of subcor-
postmortem. J Neurol Neurosurg Psychiatry 52 (6): 804. tical arteriosclerotic encephalopathy: Binswangers dis-
Nutt JG, Marsden CD, Thompson PD (1993). Human walk- ease. Mov Disord 2 (1): 18.
ing and higher-level gait disorders, particularly in the Thomsen AM, Borgesen SE, Bruhn P et al. (1986). Prognosis
elderly. Neurology 43 (2): 268279. of dementia in normal-pressure hydrocephalus after a
Owler BK, Pena A, Momjian S et al. (2004). Changes in cer- shunt operation. Ann Neurol 20 (3): 304310.
ebral blood flow during cerebrospinal fluid pressure Trosch RM, Ransom BR (1990). Levodopa-responsive par-
manipulation in patients with normal pressure hydroce- kinsonism following central herniation due to bilateral
phalus: a methodological study. J Cereb Blood Flow subdural hematomas. Neurology 40 (2): 376377.
Metab 24 (5): 579587. Tullberg M, Hultin L, Ekholm S et al. (2002). White matter
Penfield W, Rasmussen T (1950). The Cerebral Cortex of changes in normal pressure hydrocephalus and Binswan-
Man. New York, Macmillan. ger disease: specificity, predictive value and correlations
Reilly P (2001). In normal pressure hydrocephalus, intracra- to axonal degeneration and demyelination. Acta Neurol
nial pressure monitoring is the only useful test. J Clin Scand 105 (6): 417426.
Neurosci 8 (1): 6667. Vanneste J, Augustijn P, Dirven C et al. (1992). Shunting
Romanes GJ (ed.). (1981). Cunninghams Textbook of Anat- normal-pressure hydrocephalus: do the benefits outweigh
omy, 12th edn. Oxford University Press, New York. the risks? A multicenter study and literature review. Neu-
Savolainen S, Hurskainen H, Paljarvi L et al. (2002). Five- rology 42 (1): 5459.
year outcome of normal pressure hydrocephalus with or Vanneste JA (1994). Three decades of normal pressure
without a shunt: predictive value of the clinical signs, neu- hydrocephalus: are we wiser now? J Neurol Neurosurg
ropsychological evaluation and infusion test. Acta Neuro- Psychiatry 57 (9): 10211025.
chir (Wien) 144 (6): 515523. Waldemar G, Schmidt JF, Delecluse F et al. (1993). High
Silverberg GD (2004). Normal pressure hydrocephalus (NPH): resolution SPECT with [99mTc]-d,l-HMPAO in normal
ischaemia, CSF stagnation or both. Brain 127 (Pt 5): pressure hydrocephalus before and after shunt operation.
947948. J Neurol Neurosurg Psychiatry 56 (6): 655664.
Silverberg GD, Mayo M, Saul T et al. (2003). Alzheimers Williams D, Tijssen M, Van Bruggen G et al. (2002).
disease, normal-pressure hydrocephalus, and senescent Dopamine-dependent changes in the functional connectiv-
changes in CSF circulatory physiology: a hypothesis. Lan- ity between basal ganglia and cerebral cortex in humans.
cet Neurol 2 (8): 506511. Brain 125 (Pt 7): 15581569.
Stolze H, Kuhtz-Buschbeck JP, Drucke H et al. (2000). Gait Yakovlev PI (1947). Paraplegias of hydrocephalics (clinical
analysis in idiopathic normal pressure hydrocephalus note and interpretation). Am J Ment Defic 51: 561576.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 56

Calcification of the basal ganglia

JENNIFER S. HUI* AND MARK F. LEW

University of Southern California, Los Angeles, CA, USA

56.1. Introduction calcifications of the BG are found in approximately


0.31.2% of all subjects undergoing routine brain CT
Calcification of the basal ganglia (BG) has been known examination (Koller et al., 1979; Murphy, 1979; Sachs
and described since 1850 by Delacour, and histologically et al., 1979; Brannan et al., 1980; Cohen et al., 1980;
defined as cerebral blood vessel calcifications by Fenelon et al., 1993). When found, these calcifications
Bamberger in 1855. The observations of these scien- are usually punctate and restricted to the globus palli-
tists, however, are rarely credited today. In 1930, the dus, although few studies report consistent symptoms
German pathologist Fahr described a case of calcifica- related to dysfunction of BG pathways. Harrington
tion in the cerebral vessels in a patient with likely et al. (1981) reviewed the CT scans of 7000 patients
hypoparathyroidism (HP). Fahrs disease has since and found 42 patients (0.6%) with BG calcifications
become a disputed term and a relative misnomer, given and normal serum electrolytes, calcium and phosphate
that calcifications in his case occurred in predominantly levels. Clinical correlations of these patients included
white matter, sparing the BG (Klein and Vieregge, epilepsy, headache, stroke or dementia, but no symp-
1998). Confusion regarding the nomenclature and toms related to BG involvement. Likewise, Sachs
classification of BG calcifications persists, with Fahrs et al. (1979) found 14 patients out of 3800 CT exami-
disease used in varying contexts encompassing BG cal- nations (0.4%) with bilateral BG calcifications, the
cinosis of differing etiologies, symptoms and clinical majority having seizures or psychic symptoms.
significance. To clarify, the descriptive term bilateral Finally, the diversity of symptoms found in a series
striopallidodentate calcinosis (BSPDC) has been pro- of 14 of 4219 (0.3 %) consecutive patients with calci-
posed to describe idiopathic, symmetric calcifications fications and normal metabolic profiles led to the con-
of the BG resulting in characteristic clinical features clusion that further diagnostic procedures were not
and will be used throughout this chapter (Manyam, warranted if symmetric BG lesions were incidentally
1990; Manyam et al., 2001b). found on brain CT (Koller et al., 1979).
Other authors, however, have advocated an evalua-
56.2. Epidemiology
tion of calcium metabolism in cases of incidental BG
56.2.1. Incidental calcifications of the basal ganglia calcifications. In particular, Sachs et al. (1982)
reported that 10% (2/20) of incidentally revealed BG
Epidemiologic studies expanded rapidly with the calcifications revealed a diagnosis of primary HP.
widespread use of computed tomography (CT) in the However, these cases were specifically referred for
1970s, resulting in increased sensitivity of detecting evaluation of neurologic symptoms, comprising a
BG calcifications over skull plain films (Fig. 56.1). potentially higher-risk group than found with other,
Nevertheless, prevalence estimates of BG calcifica- consecutively derived CT series. In most reported
tions in the general population have varied widely cases of incidental BG calcinosis, serum calcium and
due to non-standardized radiographic and clinical phosphorus levels are normal, as seen in a collective
criteria. Authors have generally agreed that incidental total of 88 subjects reported by Koller et al. (1979),

*Correspondence to: Dr. Jennifer S. Hui, University of Southern California, 1520 San Pablo St., Suite 3000, Los Angeles, CA
90033, USA. E-mail: jhui@surgery.usc.edu
480 J. S. HUI AND M. F. LEW
Table 56.1
Etiology of basal ganglia calcifications
Idiopathic Congenital
Incidental calcifications Cockaynes syndrome
(physiologic) Mitochondrial
BSPDC myopathies
Familial Tuberous sclerosis
Autosomal dominant/ Neurofibromatosis
recessive Downs syndrome
Sporadic Hallervorden-Spatz
syndrome
Endocrine disorders Methemoglobinopathies
Hypoparathyroidism
Pseudohypoparathyroidism Toxic/anoxic
Pseudo- Lead intoxication
Fig. 56.1. Skull film demonstrating basal ganglia calcifica- pseudohypoparathyroidism Carbon monoxide
tions in a patient with hypoparathyroidism. Courtesy of Hyperparathyroidism intoxication
University of Southern California Department of Radiology. Hypothyroidism Radiation therapy
Methotrexate
Infectious Long-term
Cohen et al. (1980) and Brannan et al. (1980), except Toxoplasmosis antiepileptics
for 2 cases of previously known HP. Cystercercosis
Given the relative scarcity of pathologic conditions HIV/AIDS Autoimmune
Tuberculosis Systemic lupus
associated with incidental BG calcifications, most have
Viral encephalitis erythematosus
concluded that the radiologic finding can be regarded as
a manifestation of physiologic senescence (Murphy, HIV, human immunodeficiency virus; AIDS, acquired immunodefi-
1979; Brannan et al., 1980; Cohen et al., 1980). This ciency syndrome.
concept is supported by a population-based study show-
ing a relatively high prevalence (18.6%) of BG calcifi-
cations in healthy elderly subjects aged 85 years,
subcortical white matter, as distinguished by large,
suggesting that this finding is more common in older
confluent areas of hyperdense signal on brain CT
persons (Ostling et al., 2003). Similarly, Cohen et al.
(Fig. 56.2).
(1980) found that, among 32 patients with incidental
Conditions associated with symptomatic BG calci-
BG calcifications, 25 were above the age of 60 years.
fications can be classified into idiopathic or secondary
Together, these epidemiologic findings suggest that it
causes (Table 56.1). Of the secondary causes, primary
is reasonable to evaluate further patients with incidental
HP is the most common, with estimates of BG cal-
BG calcifications who are younger than age 40 years,
cification occurring in 6993% of those with the disor-
those with symptoms related to BG dysfunction or HP
der, as identified on brain CT imaging (Sachs et al.,
(such as tetany) or those with dense or multiple areas
1982; Illum and Dupont, 1985). Illum and Dupont
of cerebral calcification outside the globus pallidus.
(1985) found that 69% (11/16) of subjects with pri-
mary HP and 100% (8/8) of those with pseudo-HP
56.2.2. Symptomatic calcifications of the basal showed calcification in the BG. Distribution of the
ganglia lesions included the globus pallidus in all cases, with
most subjects demonstrating additional deposits in
A subset of BG calcifications exists for which there the striatum, thalamus, dentate and centrum semiovale.
are identifiable metabolic or degenerative causes Sachs et al. (1982) surveyed 14 patients with primary
(Table 56.1). In contrast to those incidental cases without HP, 13 of which demonstrated bilateral intracerebral
a known etiology, these symptomatic calcifications are calcifications (93%), 9 located in the BG. In both
often associated with a progressive syndrome of parkin- studies, patients had been diagnosed with HP for over
sonism, dystonia, chorea and tremor. Although milder 10 years, with symptoms of tetany or seizures starting
forms of symptomatic cases may occur, those who are in childhood. Parkinsonian signs of hypokinesia,
clinically affected share a radiographic presentation of rigidity or tremor were present in only 3 of 12 subjects
massive calcification of the striatum, cerebellum and in the series of Sachs et al. (1982).
CALCIFICATION OF THE BASAL GANGLIA 481

A B

C
Fig. 56.2. (A) Mild, (B) moderate and (C) severe forms of bilateral striopallidodentate calcinosis (BSPDC). Courtesy of University
of Southern California Department of Radiology.
482 J. S. HUI AND M. F. LEW
56.3. Bilateral striopallidodentate calcinosis men in the symptomatic group has been suggested in
other reviews of the literature (Ellie et al., 1989;
Although reports of parkinsonian syndromes have Kobari et al., 1997). Kobari et al. (1997) found a simi-
been relatively rare in association with HP and other lar ratio, in which over half of symptomatic cases of
secondary causes of BG calcinosis, parkinsonism is a brain calcification were men (21:11). Ellie et al.
frequent presentation of idiopathic familial cases of (1989) reported a more conservative male-to-female
massive BG calcification, popularly known as Fahrs ratio (11:13) in the symptomatic group, but, like the
disease (Lowenthal, 1986). Endocrinologic evaluations registry, noted that older patients (> 45 years) were
in these cases do not reveal abnormalities in calcium more likely to be clinically affected.
or phosphorus metabolism. Now termed BSPDC, the The clinical features of patients in the registry were
disorder can be seen in familial, autosomal-dominant, separated into 11 categories, the most common symp-
autosomal-recessive or, less commonly, sporadic tom being movement disorders (55%), with over half
patterns (Manyam, 1990; Manyam et al., 2001a). of these cases manifesting parkinsonism (57%),
Unlike incidental or some secondary causes of BG followed by chorea, tremor and dystonia (Manyam
calcinosis, familial BSPDC carries a poor prognosis. et al., 2001b). Cognitive impairment was the second
The true prevalence of the disorder remains unknown most commonly observed symptom (39%), along with
due to the evolving clinical and radiographic diag- cerebellar (36%) and speech (36%) disorders. Unlike
nostic criteria and its predominantly case-report cases of HP-associated calcifications, seizures were
description in the literature. In all likelihood relatively uncommon, accounting for 9% of all
BSPDC is rare, although cases with lesser calci- symptoms. There was considerable overlap of symp-
fication appearing in routine CT imaging cannot toms, although the number of subjects displaying more
be excluded as early presentations of BSPDC without than one symptom was not specified. Families can
a detailed family history, including radiographic demonstrate interfamilial heterogeneity.
evidence. The cognitive aspects of BSPDC have been well
documented and anatomically supported by previously
56.3.1. Clinical features described frontalsubcortical circuits (Cummings
et al., 1983; Alexander et al., 1986). Lopez-Villegas
Among multiple reports of BSPDC, a fairly uniform et al. (1996) published a case-control study of the
clinical presentation has emerged, featuring a progres- neuropsychological symptoms of subjects with BG
sive syndrome with varying symptoms of dementia, calcinosis > 5 mm, illustrating significant executive
dysarthria, parkinsonism, pyramidal and cerebellar and visuospatial dysfunction, a similar pattern to other
signs (Ellie et al., 1989; Manyam et al., 1992, 2001a, disorders affecting deep gray-matter structures
b; Kobari et al., 1997; Warren et al., 2002). Onset of (e.g. Parkinsons disease, Huntingtons disease, pro-
symptoms typically occurs between 30 and 50 years gressive supranuclear palsy). Psychiatric symptoms
and cognitive symptoms are not uncommon. Given were also more common in those with calcification
the relative uniformity of features distinct to BSPDC, compared to controls, with 22.2% meeting criteria for
Manyam et al. (2001b) established the Fahrs disease mood disorder and 33.3% for obsessive-compulsive
registry, retaining the popular name for sake of disorder (n 18). In addition, frank psychosis in
familiarity. Inclusion criteria included: (1) radiologic BSPDC has been seen alone or in conjunction with
evidence of bilateral, symmetric calcifications in the other movement disorders (Cummings et al., 1983;
BG, dentate, thalamus or cerebral white matter; (2) Rosenberg et al., 1991; Lauterbach et al., 1994), with
normal childhood development; and (3) absence of one report of three siblings, each with BG calcifica-
parathyroid disorder. All subjects were examined clini- tions and symptoms of schizophrenia, making the like-
cally and provided a detailed family history. Cases lihood of coincidental diagnoses unlikely (Rosenberg
reported in the literature that met inclusion criteria et al., 1991). Finally, Cummings et al. (1983) even
were also included (n 61). suggested that BSPDC presents with two distinct
In the combined registry and literature cases clinical patterns: an early-onset psychotic syndrome
(n 99), 67 were symptomatic and 32 were asympto- and a late-onset group with motor and cognitive
matic. The symptomatic group demonstrated a signifi- symptoms, although delayed tardive effects from early
cantly higher mean age ( sd) (47  15) compared to pharmacologic treatment of psychosis could not be
the asymptomatic group (32  20) and were more than discounted.
twice as likely to be men (45:22). Men were also more Several variations of the BSPDC clinical syndrome
likely to have intracranial calcifications compared have been published in case-report format, including
to women (57:42, P < 0.01). This predominance of a patient with subacute dementia occurring over 6
CALCIFICATION OF THE BASAL GANGLIA 483
months affecting memory, executive functioning and were documented in 1 subject over a decade with
mood without motor impairment (Benke et al., 2004). serial CT scans, showing a progressive increase in
Warren et al. (2002) reported a patient with corticoba- total calcium deposits from age 50 to 57, then a
sal degeneration manifesting as left-sided rigidity and decrease corresponding to cerebral atrophy at age 60.
apraxia, a left extensor plantar response and startle This pattern of progressive calcification correlating
myoclonus. Finally, a particularly unique case of a with worsening symptoms over time has been cross-
12-year-old girl with transient parkinsonism lasting sectionally observed in other studies, suggesting a
10 days was found after she had drunk a cup of tea, cumulative pathologic effect. In multigenerational
associated with multiple gray- and white-matter families with BSPDC, for instance, younger generations
calcifications on brain CT (Yoshikawa and Abe, 2003). are often asymptomatic with milder degrees of
calcification, whereas older relatives present with the
56.3.2. Radiography and clinical correlations progressive clinical syndrome. In a father-and-son
series by Ellie et al. (1989), a 48-year-old man presented
In virtually all reported cases of incidental, secondary with progressive speech impairment and cerebellar
and idiopathic causes of BG calcinosis, calcification ataxia with multiple areas of dense cerebral calcifica-
in the brain is nearly symmetrical and, in BSPDC, tion, with his asymptomatic 22-year-old son demon-
restricted to the central nervous system (Lowenthal, strating less extensive calcifications of the globus
1986). In cases meeting criteria for the Fahrs disease pallidus. In another familial report, Moskowitz et al.
registry, calcification was mainly seen in the dentate (1971) reported 5 patients in one family with BG
nucleus, BG, thalamus and centrum semiovale with calcifications. Of the 5 subjects, the two eldest (ages
varying severity (Figs. 56.2 and 56.3) (Manyam 47 and 58 years) presented with parkinsonism, whereas
et al., 2001b). There were no consistent patterns of the younger individuals (aged 1330 years) were
calcification within families. Symptomatic patients asymptomatic. The severity of calcifications was not
showed a significantly greater amount of calcification measured.
compared to asymptomatic individuals, suggesting a Alternatively, other familial reports of BSPDC have
doseresponse relationship. Longitudinal changes found that younger generations were more severely
affected, both clinically and radiographically, suggest-
ing a possible role for genetic anticipation (Kobari
et al., 1997). This observation has been supported by
the recent identification of a chromosomal locus for
BG calcification, showing a younger age of onset with
each genetic transmission (see below) (Geschwind
et al., 1999). As a result of these varying opinions
regarding the individual prognosis of BG calcinosis,
it is difficult to predict who will become symptomatic
from BSPDC, although it appears that more extensive
calcifications correlate with increasing severity of
symptoms.

56.3.3. Genetics of BSPDC

Variations in familial expression have revealed several


inheritance patterns of BSPDC, the most common of
which is an autosomal-dominant pattern. Autosomal-
recessive (Smits et al., 1983) and X-linked (Naderi
et al., 1993) inheritance patterns of BG calcinosis have
also been described, although these cases are often
associated with endocrine disorders such as HP or
have inadequate pedigrees consisting of only one
affected generation, limiting the certainty of genetic
transmission patterns. Of the 99 patients included in
Fig. 56.3. Dentate calcification in bilateral striopallidoden- the Fahrs disease registry, 73 patients belonged to
tate calcinosis (BSPDC). Courtesy of University of Southern 14 families, demonstrating a clear autosomal-dominant
California Department of Radiology. inheritance of BSPDC. Of the remaining patients, 12
484 J. S. HUI AND M. F. LEW
were grouped as familial without a clear pattern of corresponding to granular and stippling calcium
inheritance and 14 were sporadic cases. deposits within the parenchyma and along capillaries
Moskowitz et al. (1971) were among the first to report (Manyam et al., 2001a). Matsumoto et al. (1998) found
an entire family with autosomal-dominant BSPDC, intense calcification surrounding veins in the BG and
examining 15 members spanning at least two genera- white matter, with loss of myelin sheaths and axons in
tions, with documented male-to-male transmission. Five a patient with cerebral lupus. Fujita et al. (2003)
subjects had BG calcification and 10 did not have calci- described three histologic patterns of cerebral calcifica-
fication, as detected on skull films rather than CT, tion in a variety of neurodegenerative diseases, includ-
perhaps underestimating its prevalence. In another ing Alzheimers disease, Pick disease and diffuse
extensive familial study, Manyam et al. (2001a) evalu- neurofibrillary tangles with calcification (DNTC), a
ated 27 members of a family over four generations, with disorder demonstrating Fahrs-like calcification in
18 showing radiographic evidence of brain calcifications the BG. Type 1 histology involved diffuse deposition
in an autosomal-dominant pattern. Three patients were within the tunica media of small and medium-sized
symptomatic with parkinsonism, one, interestingly, with vessels; type 2 consisted of spherical concretions in
autopsy-confirmed Parkinsons disease and another the parenchyma; and type 3 included rows of small
without Lewy bodies on autopsy. calcifications lying along capillaries. This latter,
The chromosomal locus for one family with pericapillary pattern of calcification may reflect the
idiopathic BG calcifications has been localized to pattern most commonly seen in BPSDC, given its
chromosome 14q (Geschwind et al., 1999). Geschwind reproducibility in multiple histologic studies and its
et al. clinically and genetically examined 28 members frequent presence in DNTC (Duckett et al., 1977;
of a multigenerational family using linkage analysis, Matsumoto et al., 1998; Manyam et al., 2001a; Fujita
of which 12 had BG calcifications. The family demon- et al., 2003). Other histologic patterns, however, such
strated autosomal-dominant inheritance patterns over as intracellular deposits with minimal neuronal death,
four generations, with symptoms ranging from combi- have also been reported (Takashima and Becker,
nations of parkinsonism, dystonia and tremor. The aver- 1985; Mahy et al., 1999).
age age at onset of symptoms was 37 years, appearing to The molecular composition of cerebral calcifications
decrease over three successive generations by an aver- consists of hydroxyapatite crystals assembled from
age of > 20 years, such that the third generation had various bone matrix proteins, a structure not unlike
symptom onset from ages 512 years. physiologic calcifications found elsewhere in the body
The discovery of such a prominent pattern of anti- (Smeyers-Verbeke et al., 1975; Fujita et al., 2003).
cipation is somewhat surprising, given its lack of Detailed chemical analyses of these deposits reveal that
consistent documentation in the current literature; calcium and phosphorus are present in the greatest
however, it points to the possibility of a triplet repeat proportion, followed by trace quantities of magnesium,
mutation as a cause of some cases of BSPDC. Several zinc, aluminum and iron (Smeyers-Verbeke et al., 1975;
candidate genes within the 14q locus have been men- Duckett et al., 1977). Whether the deposits represent a
tioned, including a proteosome subunit, somatostatin primary deposition or dystrophic change from prior
receptor, kinesin receptor, paraplegin and the A kinase ischemic insult has yet to be determined.
anchor protein (Geschwind et al., 1999). It will be Several proposed mechanisms exist for the formation
important to determine whether other families with of BG calcifications, including altered vessel permeabil-
autosomal-dominant BSPDC have mutations in the ity to calcium (Lowenthal, 1986), changes in blood flow
same locus or whether there are several mutations and resulting tissue ischemia (Lowenthal, 1986; Uygur
leading to the variations in phenotype for this disorder. et al., 1995), synaptic excitotoxicity (Mahy et al.,
1999) or local shifts in calcium concentration, possibly
56.3.4. Pathophysiology due to parathyroid hormone-responsive enzymes (Low-
enthal, 1986; Kurup and Kurup, 2002). Uygur et al.
The pathogenesis of BG calcifications and BSPDC is (1995) demonstrated decreased regional blood flow in
unknown. Numerous histochemical studies and the BG and frontal cortices through a brain SPECT
experimental models have been conducted to define study of a patient with BSPDC. However, it is not
the nature of these calcifications. Several different known whether this finding is a result or cause of the
patterns of calcification have been shown histologically, cerebral calcifications. Animal models of excitotoxicity
associated with neuronal loss and gliosis in the BG and have suggested a role for glutamate in calcium deposit
dentate (Matsumoto et al., 1998; Tsuchiya et al., 2002). formation, using the excitatory amino acid, ibotenic
On gross inspection, chalky yellow mineral deposits acid, to induce calcified inclusions in astrocytes in an
in the dentate and striatum are found, microscopically in vivo rat brain (Mahy et al., 1999).
CALCIFICATION OF THE BASAL GANGLIA 485
Table 56.2
Treatment of idiopathic bilateral striopallidodentate calcinosis with dopaminergic medication

Source Number of cases Response Drug received

Klawans et al. (1976) 1 No improvement Levodopa 1


Manyam et al. (2001a) 6 Improvement Levodopa 2
No medication given 3
No improvement Levodopa 1
Berendes and Dorstelmann 1 No improvement Levodopa 1
(1978)
Harati et al. (1984) 3 Mild improvement Amantadine 1
No medication given 2

56.3.5. Treatment of parkinsonism in BSPDC forms remain predictable within a spectrum of fre-
quently disabling symptoms. The underlying patho-
The parkinsonism of BSPDC has been variable with physiology is not yet well understood. It is possible,
respect to response to dopaminergic treatment. Klawans however that clues from genetic evaluation, although
et al. (1976) described a case of levodopa-resistant par- the presentation is varied, may lead to a more detailed
kinsonism in association with BG calcifications, treated understanding of BSPDC. Additionally, data gleaned
unsuccessfully for 30 months with levodopa doses up from an ongoing registry of BSPDC patients may be
to 7 g/day, suggesting a dopaminergic postsynaptic instrumental to improving our basic knowledge of this
receptor site dysfunction. Conversely, Manyam et al. disorder over time.
(2001a) reported a levodopa-responsive case of BSPDC
at doses of 1200 mg/day. Characteristic Lewy bodies, References
however, were found on autopsy, suggesting that the
patient had levodopa-responsive Parkinsons disease Alexander GE, DeLong MR, Strick PL (1986). Parallel orga-
and BSPDC concurrently (Manyam et al., 2001a). Ver- nization of functionally segregated circuits linking basal
mersch et al. (1992) also described levodopa-responsive ganglia and cortex. Annu Rev Neurosci 9: 357381.
Bamberger H (1855). Beobachtungen und bemerkungen uber
BG calcinosis in patients who were already diagnosed
hirnkrankheiten. Verh Phy Med Ges 6: 325328.
with Parkinsons disease prior to their CT scans. In addi-
Benke T, Karner E, Seppi K et al. (2004). Subacute dementia
tion, these calcifications were mostly unilateral, without and imaging correlates in a case of Fahrs disease. J Neurol
mention of their extent or severity, opening the possibi- Neurosurg Psychiatry 75: 11631165.
lity that these cases were radiographically distinct from Berendes K, Dorstelmann D (1978). Unsuccessful treatment
the BSPDC syndrome. In a review of the literature in with levodopa of a parkinsonian patient with calcification
which treatment of BSPDC was reported, 3/6 patients of the basal ganglia. J Neurol 218: 5154.
improved with dopaminergic treatment, 3/6 did not Brannan TS, Burger AA, Chaudhary MY (1980). Bilateral
improve and 5 patients did not receive treatment basal ganglia calcifications visualised on CT scan. J Neurol
(Table 56.2). This number of publications to date is Neurosurg Psychiatry 43: 403406.
insufficient to identify a consistent pattern regarding Cohen CR, Duchesneau PM, Weinstein MA (1980). Calcifi-
cation of the basal ganglia as visualized by computed
treatment of BSPDC.
tomography. Neuroradiology 134: 9799.
Cummings JL, Gosenfeld LF, Houlihan JP et al. (1983). Neu-
56.4. Summary ropsychiatric disturbances associated with idiopathic
calcification of the basal ganglia. Biol Psychiatry 18:
Calcification of the BG occurs in many contexts, ran- 591601.
ging from incidental findings to disorders of mineral Delacour A (1850). Ossification des capillaries du cerveau.
Annu Med Psychol 2: 458461.
metabolism and familial forms of cerebral calcinosis.
Duckett S, Galle P, Escourolle R et al. (1977). Presence of
BSPDC, also known as Fahrs disease, is an idiopathic, zinc, aluminum, magnesium in striopallidodentate (SPD)
familial form of calcification of the BG presenting as a calcifications (Fahrs disease): Electron probe study. Acta
combination of parkinsonism, dementia and cerebellar Neuropathol (Berl) 38: 710.
signs. Although the terminology of BG calcifications Ellie E, Julien J, Ferrer X (1989). Familial idiopathic strio-
remains controversial, the clinical findings of severe pallidodentate calcifications. Neurology 39: 381385.
486 J. S. HUI AND M. F. LEW

Fahr I (1930). Idiopathipathische verkalking der hirume Manyam BV, Walters AS, Keller IA et al. (2001a). Parkin-
fasse. Zbl Allf Path 50: 129133. sonism associated with autosomal dominant bilateral strio-
Fenelon G, Gray F, Paillard F et al. (1993). A prospective pallidodentate calcinosis. Parkinsonism Relat Disord 7:
study of patients with CT detected pallidal calcifications. 289295.
J Neurol Neurosurg Psychiatry 56: 622625. Manyam BV, Walters AS, Narla KR (2001b). Bilateral
Fujita D, Terada S, Ishizu H et al. (2003). Immunohistologi- striopallidodentate calcinosis: Clinical characteristics of
cal examination on intracranial calcification in neurode- patients seen in a registry. Mov Disord 16: 258264.
generative diseases. Acta Neuropathol (Berl) 259264. Matsumoto R, Shintaku M, Suzuki S et al. (1998). Cere-
Geschwind DH, Loginov M, Stern JM (1999). Identificatoin of bral perivenous calcification in neuropsychiatric lupus
a locus on chromosome 14q for idiopathic basal ganglia cal- erythematosus: A case report. Neuroradiology 40: 583586.
cification (Fahr disease). Am J Hum Genet 65: 764772. Moskowitz MA, Winickoff RN, Heinz ER (1971). Familial
Harati Y, Jackson JA, Benjamin E (1984). Adult onset idio- calcification of the basal ganglions: A metabolic and
pathic familial brain calcifications. Arch Intern Med 144: genetic study. N Engl J Med 285: 7277.
24252427. Murphy MJ (1979). Clinical correlations of CT scan-detected
Harrington MG, Macpherson P, McIntosh WB et al. calcifications of the basal ganglia. Ann Neurol 6: 507511.
(1981). The significance of the incidental finding of basal Naderi S, Colakoglu Z, Luleci G (1993). Calcification of
ganglia calcification on computed tomography. J Neurol basal ganglia associated with pontine calcification in four
Neurosurg Psychiatry 44: 11681170. cases: A radiologic and genetic study. Clin Neurol Neuro-
Illum F, Dupont E (1985). Prevalences of CT-detected calcifi- surg 95: 155157.
cation in the basal ganglia in idiopathic hypoparathyroidism Ostling S, Andreasson LA, Skoog I (2003). Basal ganglia
and pseudoparathyroidism. Neuroradiology 27: 3237. calcification and psychotic symptoms in the very old. Int
Klawans HL, Lupton M, Simon L (1976). Calcification J Geriatr Psychiatry 18: 983987.
of the basal ganglia as a cause of levodopa-resistant Rosenberg DR, Neylan TC, el-Alwar M et al. (1991). Neu-
parkinsonism. Neurology 26: 221225. ropsychiatric symptoms associated with idiopathic calcifi-
Klein C, Vieregge P (1998). The confusing history of Fahrs cation of the basal ganglia. J Nerv Ment Dis 179: 4849.
disease. Neurology 50 (Suppl 4): A59. Sachs C, Ericson K, Erasmie U et al. (1979). Incidence of
Kobari M, Nogawa S, Sugimoto Y et al. (1997). Familial basal ganglia calcifications on computed tomography.
idiopathic brain calcification with autosomal dominant J Comput Assist Tomogr 3: 339344.
inheritance. Neurology 48: 645649. Sachs C, Sjoberg H, Ericson K (1982). Basal ganglia calcifi-
Koller WC, Cochran JW, Klawans HL (1979). Calcification cations on CT: Relation to hypoparathyroidism. Neurology
of the basal ganglia: Computerized tomography and clini- 32: 779782.
cal correlation. Neurology 29: 328333. Smeyers-Verbeke J, Michotte Y, Pelsmaeckers J et al.
Kurup RK, Kurup PA (2002). Hypothalamic digoxin related (1975). The chemical composition of idiopathic nonarter-
membrane Na-K ATPase inhibition and familial basal iosclerotic cerebral calcifications. Neurology 25: 4857.
ganglia calcification. Neurosci Res 42: 3544. Smits MG, Gabreels FJM, Thijssen HOM et al.
Lauterbach EC, Spears TE, Prewett MJ et al. (1994). Neu- (1983). Progressive idiopathic strio-pallido-dentate calcino-
ropsychiatric disorders, myoclonus, and dystonia in sis (Fahrs disease) with autosomal recessive inheritance.
calcification of basal ganglia pathways. Biol Psychiatry Report of three siblings. Eur Neurol 22: 5864.
35: 345351. Takashima S, Becker L (1985). Basal ganglia calcification in
Lopez-Villegas D, Kulisevsky J, Deus J et al. (1996). Neu- Downs syndrome. J Neurol Neurosurg Psychiatry 48:
ropsychological alterations in patients with computed 6164.
tomography-detected basal ganglia calcification. Arch Tsuchiya K, Nakayama H, Iritani S et al. (2002). Distribution
Neurol 53: 251256. of basal ganglia lesions in diffuse neurofibrillary tangles
Lowenthal A (1986). Striopallidodentate calcifications with calcification: A clinicopathological study of five
(chapter 24). In: PJ Vinken, GW Bruyn, HL Klawans autopsy cases. Acta Neuropathol (Berl) 103: 555564.
(Eds.), Handbook of Clinical Neurology, Vol. 5, Elsevier Uygur GA, Liu Y, Hellman RS et al. (1995). Evaluation of
Science Publishers, New York, pp. 417436. regional cerebral blood flow in massive intracerebral cal-
Mahy N, Prats A, Riveros A et al. (1999). Basal ganglia cal- cifications. J Nucl Med 36: 610612.
cification induced by excitotoxicity: An experimental Vermersch P, Leys D, Pruvo J et al. (1992). Parkinsons disease
model characterised by electron microscopy and X-ray and basal ganglia calcifications: Prevalence and clinico-radi-
microanalysis. Acta Neuropathol (Berl) 98: 217225. ological correlations. Clin Neurol Neurosurg 94: 213217.
Manyam BV (1990). Bilateral strio-pallido-dentate calcino- Warren JD, Mummery CJ, Al-Din AS et al. (2002). Cortico-
sis: A proposed classification of genetic and secondary basal degeneration syndrome with basal ganglia calcifi-
causes. Mov Disord 5: 94. cation: Fahrs disease as a corticobasal look-alike? Mov
Manyam BV, Bhatt MH, Moore WD et al. (1992). Bilateral Disord 17: 563567.
striopallidodentate calcinosis: Cerebropsinal fluid, ima- Yoshikawa H, Abe T (2003). Transient parkinsonism in
ging, and electrophysiological studies. Ann Neurol 31: bilateral striopallidodentate calcinosis. Pediatr Neurol 29:
379384. 7577.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 57

Trauma and Parkinsons disease

OSCAR S. GERSHANIK*

Laboratory of Experimental Parkinsonism, ININFA-CONICET, Buenos Aires, Argentina

57.1. Introduction responsible for the development of PD were not only


those sustained in the head, but different kinds of per-
The relationship between trauma and Parkinsons dis- ipheral trauma (cuts, bruises, skin punctures, burns,
ease (PD) and/or parkinsonism can be approached usually in the subsequently affected limb or body part).
from different perspectives. We may focus, from an Exposure to a damp environment and emotional trauma
epidemiological perspective, on the existence of pre- were also believed to be capable of inducing parkin-
ceding trauma, to either the head or other body parts, sonism. Charcot (cited by Factor et al., 1988) advanced
as a risk factor for the subsequent development of the ascending neuritis hypothesis to explain the rela-
PD; on the other hand, trauma may be the cause of tionship between peripheral trauma and PD. According
secondary parkinsonism through different mec- to this hypothesis, a peripheral injury would induce an
hanisms; or we may look at the effects of trauma on ascending inflammatory reaction along the nerves that
pre-existing PD. All these approaches have been eventually reached the central nervous system. In the
extensively dealt with in the literature and we will first part of the 20th century, several French authors
try to review them as comprehensively as possible to reported the development of parkinsonism as a conse-
bring the reader an updated discussion on the subject quence of concussion associated with mesencephalic
(for reviews, see: Factor et al., 1988; Koller et al., lesions in soldiers involved in World War I. Later on,
1989; Stern, 1991; Jankovic, 1994; Lees, 1997; Krauss in 1928, postmortem analysis of the brain of a patient
and Jankovic, 2002). who developed parkinsonism after sustaining a bullet
Trauma has long been held suspect as one of the injury to the head, showing hemorrhagic lesions at
contributing factors to the development of PD. Some the basal ganglia level, gave credence for the first time
of the earliest references in this regard date back to to the existence of true cases of posttraumatic parkin-
James Parkinson himself, who speculated, as he had sonism (Factor et al., 1988). Almost contemporary with
no solid evidence to substantiate it, that the disease this observation was the report by Patrick and Levy
might be the consequence of a direct injury to the (1922) claiming that, in 15% of the cases of PD they
brainstem or the dural sheath surrounding it. He rea- had followed and studied, trauma was a contributing
soned that, given the range of mobility that the upper factor to its development. It was not until 1934, when
segment of the spine had, this would make this region Grimberg published his thorough analysis on paralysis
more susceptible to trauma (Parkinson, 1817). In their agitans and trauma, that objective criticism of the
comprehensive historical review on the concept of hypothesis of trauma-induced parkinsonism was raised.
trauma as an etiology of parkinsonism, Factor and col- Grimberg dismissed 84 out of 86 cases reported since
laborators (1988) thoroughly discuss not only the pre- 1873 onwards as not being related to trauma based
vailing medical theories surrounding this issue in the on a number of factors (misdiagnosis, no definite
19th and early 20th century, but the socioeconomic history of trauma, pre-existing parkinsonian symptoma-
factors that may have fueled them, among them the tology antedating the causative trauma, unclear tem-
passing of the first workmens compensation legisla- poral relationship between the occurrence of the
tion. Of interest is the fact that traumatic factors held trauma and the onset of parkinsonism, lack of a clear

*Professor Oscar S. Gershanik, Department of Neurology, Centro Neurologico-Hospital Frances, Director, Laboratory of
Experimental Parkinsonism, ININFA-CONICET, La Rioja 951 (1221) Buenos Aires, Argentina. E-mail: gersha@gmail.com
488 O. S. GERSHANIK
pathophysiological mechanism to explain such an asso- and skull fracture that were subsequently followed
ciation) (Grimberg, 1934). His review, together with the for a prolonged period of time. Morbidity ratios were
one published more recently in this same Handbook by not elevated for PD or other neurodegenerative disor-
Schwab and England in 1968, concluded that it was ders such as Alzheimers and motor neurone diseases
only in cases severe enough to cause traumatic injury (Williams et al., 1991; De Rijk et al., 1996).
to the midbrain, subsequently ascertained by the post- In only seven case-control studies, five of them car-
mortem finding of hemorrhage in that same region, that ried out in North America (USA and Canada), one in
one could establish a causal relationship between the UK and one in Taiwan, was a positive association
trauma and the development of parkinsonism (Schwab found.
and England, 1968). These conclusions are only applic- In the London study, Godwin-Austen and colla-
able to the diagnosis of posttraumatic parkinsonism. A borators (1982) interviewed 350 patients and controls,
different conceptual framework has to be applied to trying to establish the relationship between smoking
the still controversial issue of trauma as a risk factor and PD and its association with several risk factors.
for the development of PD. They found that PD patients were more likely to have
had head injury associated with loss of consciousness
57.2. Trauma as a risk factor for the than controls. Unfortunately, as this was not the pri-
development of Parkinsons disease mary interest of this study, no numbers are given and
the level of significance of this finding is not provided.
Current theories on the etiology and pathogenesis Factor and Weiner (1991) interviewed 97 patients
of PD consider this disorder to be multifactorial and and 64 controls (spouses) seen consecutively in an
the result of a combination of a genetic predisposi- outpatient clinic. In 32% of PD patients and 17% of
tion possibly interacting with environmental factors. controls there was a prior history of head trauma of
That genes play a role in the etiology of PD is sup- any degree of severity (P < 0.05); when head trauma
ported at present by the discovery of at least 11 forms with alteration of consciousness was considered, the
of genetic parkinsonism that share clinical features association was maintained at the same level of signif-
and possibly pathogenetic mechanisms with the more icance (20.6% of patients and 8% of controls).
common, as yet sporadic, form of the disease (Corti In the study by Stern et al. (1991), a detailed ana-
et al., 2005; OMIM Database, 2005). The quest for lysis of various environmental exposures and early
environmental exogenous triggering factors has life experiences was conducted in 80 patients with
remained elusive and only supported through indirect old-onset PD (age older than 60), 69 young-onset
evidences gathered from numerous and extensive epide- patients (age younger than 40: young-onset Parkin-
miological studies. Age, sex, dietary habits, infections, sons disease (YOPD)) and 149 age- and sex-matched
environmental toxins and trauma are among the factors controls. They found that at least one episode of
considered by these studies (Lai et al., 2002; Allam trauma severe enough to cause vertigo, dizziness,
et al., 2003). blurred or double vision, seizures or convulsions, tran-
Although a number of epidemiological studies sient memory loss, personality changes or paralysis
addressing this question find a positive association occurred more frequently in both old and young-
between past history of trauma and later development onset patients prior to disease onset than in controls.
of PD, a larger number dispute these findings. This difference was found to be significant (odds ratio
Table 57.1 lists the majority of published epidemiolo- 2.7; P < 0.05).
gical studies that have looked at the issue of trauma as The study by Semchuk et al. (1993) was carried
a risk factor for PD. Fourteen out of 19 case-control out in Calgary, Canada, and included 130 patients
studies originating in different parts of the world, with neurologist-confirmed idiopathic parkinsonism
involving more than 1500 patients and over 2000 con- and 2 matched controls adjusted by age and sex
trols, failed to find a positive association between pre- obtained randomly from the same community
existing trauma and PD (for references, see (random-digit dialing). They used a multivariate con-
Table 57.1). In addition, two very important cohort ditional logistic regression analysis method in which
studies, the Rotterdam and the Olmsted county/Mayo they controlled for any potential confounding or
Clinic studies, were also unable to establish a link interaction between the different exposure variables
between the development of PD and past history of considered. In four of the eight logistic regression
head trauma (Williams et al., 1991; De Rijk et al., models used, having a history of head trauma was
1996). These studies included patients with reported significantly associated with the risk of developing
head trauma of enough severity to cause loss of PD (P < 0.001), second only to the existence of a
consciousness or amnesia, residual neurologic deficits positive family history.
TRAUMA AND PARKINSONS DISEASE 489
Table 57.1
Trauma and Parkinsons disease: epidemiological studies

Number of
Author Year patients/controls Type of study Association Location

Nuti et al. 2004 190/190 Case-control No Tuscany, Italy


Bower et al. 2003 196/196 Case-control Yes Minnesota, USA
P < 0.02
Tsai et al. 2002 30 YOPD/30 Case-control Yes Taiwan, China
60 LOPD P < 0.002
Preux et al. 2000 140/280 Case-control No France
Taylor et al. 1999 140/147 Case-control Yes Boston, USA
P < 0.0001
Werneck and Alvarenga 1999 92/110 Case-control No Rio de Janeiro, Brazil
Kuopio et al. 1999 123/246 Case-control No Turku, Finland
Smargiassi et al. 1998 86/86 Case-control No Emilia Romagna, Italy
McCann et al. 1998 224/310 Case-control No Queensland and NSW,
Australia
De Michele et al. 1996 116/232 Case-control No Campania/Molise, Italy
Seidler et al. 1996 380/379 Case-control No Germany
De Rijk et al. 1996 Rotterdam cohort Cohort No The Netherlands
Yang et al. 1994 90/90 Case-control No China
Morano et al. 1994 74/148 Case-control No Caceres, Spain
Semchuk et al. 1993 130/260 Case-control Yes Calgary, Canada
P < 0.001
Butterfield et al. 1993 63/68 Case-control No Oregon, USA
Stern et al. 1991 149/149 Case-control Yes USA/Canada
P < 0.05
Factor and Weiner 1991 97/64 Case-control Yes Miami/Albany, USA
P < 0.05
Williams et al. 1991 Olmsted county Cohort No Minnesota, USA
cohort (821)
Hofman et al. 1989 86/172 Case-control No The Netherlands
Tanner et al. 1987 100/200 Case-control No China
Ward et al. 1983 65/65 Case-control No USA/Canada/UK
Godwin-Austen et al. 1982 350/350 Case-control Yes London, UK
More likely

YOPD, young-onset Parkinsons disease; LOPD, late-onset Parkinsons disease.

Taylor et al. (1999) recruited 140 PD (examined and et al. (2002) and Bower et al. (2003), carried out in
followed by neurologists and movement disorders spe- Taiwan and Minnesota respectively. In Tsais study
cialists) cases from the Boston University Medical Cen- YOPD cases were compared to late-onset cases and
ter, USA. Controls were in-laws or friends of the controls. They included 30 cases with age of onset
patients (n 147). For this study a positive history of before 40, 30 young controls and 60 late-onset cases.
head trauma was considered when it was severe enough All participants in the study were administered a struc-
to cause loss of consciousness, blurred or double vision, tured questionnaire in which all the potential risk factors
dizziness, seizures or memory loss, rather than mere under analysis were included. In the case of self-
bruising, bleeding or stitches. Twenty-five percent of reported head injuries the information was verified
the cases versus 7.5% of the controls had a history of against medical records to ascertain its validity. A multi-
head trauma according to this study. This difference ple logistic regression analysis was used to examine the
was found to be highly significant (P < 0.0001). significance of all risk factors simultaneously. When
The most recent studies finding a positive associa- compared with late-onset PD patients, YOPD had a sub-
tion between head trauma and PD are those of Tsai stantially higher percentage of head injuries (37% versus
490 O. S. GERSHANIK
10%; P < 0.002). Although the difference in the different studies, both in regard to case ascertainment
percentage having head injury was not statistically and data evaluation, including definition of head injury
significant between YOPD and controls, the proportion or trauma, may be the cause of this discrepancy. All
of those having such an experience in the patient group case-control studies are retrospective and in those
was more than doubled (37% versus 17%). It is indeed reporting an association between head injury and PD,
interesting to note that this study found head trauma to the average interval between the reported trauma and
be a risk factor only for YOPD and not for PD in the onset of the disease was 33.3 years. After so many
general. The study by Bower et al. (2003) has several years it is possible that recall bias may influence
distinguishing features that differentiate it from other the outcome of these studies. Patients with chronic
case-control studies. They conducted a population-based diseases are more likely than healthy individuals
case-control study of incident PD cases and population to recall any significant event in their lives that may
controls nested within the Olmsted county cohort. The offer an explanation for their present predicament.
three key features of their study were the population- They are always looking for possible connections
based nature, the nesting within a records linkage system between life experiences and disease onset, as indivi-
and the use of information about head trauma that was duals carrying a chronic disease often feel unfairly
recorded historically before the onset of PD. They dealt with by life.
included 196 case-control pairs matched for age and Unfortunately these arguments do not necessarily
sex. Because of the peculiar design of this study I hold up to thorough scrutiny. In Factors study (Factor
believe it is interesting to analyze the results in more and Weiner, 1991) the association was still significant
detail than in the previous studies. There were 13 head whether they considered any type of trauma or severe
trauma events in the PD cases versus only 3 in the con- enough trauma to cause alteration of consciousness,
trols, this difference being statistically significant (P < suggesting that differences in definition of head injury
0.02). This association, however, was only restricted to may not be the cause of the discrepancy. If recall bias
the more severe cases. There was no difference in the was the confounding factor, older patients would have
frequency of mild head trauma with amnesia as the resi- reported more episodes of head trauma than patients
dual symptom between cases and controls. However, the 30 years their younger, which was not the case in the
frequency of mild head trauma with loss of conscious- study by Stern et al. (1991), comparing young-onset
ness combined with moderate or severe trauma was sig- with old-onset cases. In their study there was no differ-
nificantly higher in cases (5.6%) than in controls (0.5%). ence in the prevalence of head trauma prior to disease
Using the frequency of head trauma among controls esti- onset between the two groups. We are therefore left
mated in their study (1.5%), the authors computed a with an unresolved dilemma, as no single retrospective
population-attributable risk for head trauma in PD of study is free of methodological flaws or limitations in
5%. In men, head trauma was significantly more fre- its ability to detect significant differences between the
quent in cases (9.9%) than in controls (1.7%). In populations under study.
women, the frequency of head trauma was the same in It is worth noting that, even in those studies in
cases and controls (1.3%); however, the numbers were which there were no statistically significant differ-
small (one event each). When they stratified cases by ences between cases and controls in regard to preva-
median age at onset of PD, the analysis showed no sig- lence of head trauma, the PD group always had a
nificant difference between cases and controls for higher frequency of reported head injuries than the
patients with earlier age at onset. However, there was controls. However, not even in those studies finding
a significantly higher frequency of head trauma in cases a positive association was head trauma the sole predic-
(6.4%) than controls (0.0%) for patients with later onset tor, as it was usually associated with family history
(P < 0.02) (Bower et al., 2003). These findings are in and exposure to other environmental factors (well-
absolute contrast to those of Tsai et al. (2002) and no water drinking, pesticide use, etc.) (Semchuk et al.,
reasonable explanation can be advanced to account for 1993; Taylor et al., 1999).
these differences. Head trauma might therefore be one of several risk
All of these studies were published after the intro- factors acting in combination in a predisposed indivi-
duction of levodopa, thus including levodopa-respon- dual that precipitate the cascade of events leading to
siveness as one of the diagnostic criteria, and the the development of PD. Head trauma may increase the
majority of them were conducted using very strict risk of PD only in those individuals who carry a specific
methodological approaches. It is therefore difficult polymorphism of a susceptibility gene. This hypothesis
to interpret these apparently conflicting findings. Sev- refers to a model of geneenvironment interaction that
eral explanations can be offered to account for these has also been proposed for Alzheimers disease (Bower
differences. Methodological differences among the et al., 2003).
TRAUMA AND PARKINSONS DISEASE 491
In a few of the publications reporting a positive in gene expression, activation of reactive oxygen spe-
association between head trauma and an increased risk cies, activation of intracellular proteases (calpains),
of developing PD there have been attempts at finding expression of neurotrophic factors and activation of
the factors underlying this association. Three major cell death genes (apoptosis) (McIntosh et al., 1998;
hypotheses have been put forward (Factor and Weiner, Bramlett and Dietrich, 2003). In addition, elevation
1991; Bower et al., 2003), as described below. of proinflammatory cytokines has also been demon-
First, an isolated episode of head trauma could lower strated in animal models of brain trauma which could
the number of nigral cells, on top of which the normal further exacerbate the neuronal damage initially
decline of dopaminergic neurons occurring with age induced by the head injury (Liu et al., 2003). All these
would be superimposed. This concept, also designated factors may play a role in mediating delayed cell death
as the single-hit hypothesis originally proposed by after trauma. TBI, according to some, should be con-
Calne and Langston (1983), has been abandoned for sev- sidered as both an inflammatory and/or a neurodegen-
eral reasons, the most important being the difference erative disease. A research report by Uryu et al. (2003)
between the regional pattern of neuronal loss in PD com- has linked TBI to age-dependent synuclein (Syn)
pared to normal aging (Fearnley and Lees, 1991). More- pathology in mice. Of interest is the fact that both
over, studies using fluorodopa positron emission genetically induced and sporadic PD have been linked
tomography (PET) suggest that the rate of decline in to Syn deposition and pathology. Therefore any
striatal uptake is more rapid in PD than in normal aging mechanism leading to modifications in the expression
(Morrish et al., 1998). of Syn and/or its pathological deposition could be rele-
Second, it has been proposed that head injury could vant to the understanding of the pathogenesis of PD.
somehow cause a disruption of the bloodbrain barrier, In Uryus paper, at 1 week post-TBI, the aged mouse
thus allowing exogenous neurotoxins to enter the central brain showed a transient increase of a- and b-Syn
nervous system and trigger the cascade of nigral degen- immunoreactivity (IR) in the neuropil as well as an
eration (Graves et al., 1990). The finding of polymorpho- induction of g-Syn IR in subcortical axons. This was
nuclear leukocyte infiltration of the brain tissue associated with strong labeling of striatal axon bundles
following a head trauma, on the other hand, may be an by antibodies to altered or nitrated epitopes in a-Syn
indication of immune-mediated changes leading to neu- as well as by antibodies to inducible nitric oxide
rodegeneration (Bower et al., 2003). Both mechanisms synthase. The latter findings are highly suggestive, as
may be secondary to trauma-induced disruption of the it is well known that nitration of a-Syn is a common fea-
bloodbrain barrier and could eventually cause a delayed ture of pathological forms of this protein in the Lewy
neurodegenerative process. bodies of diverse synucleinopathies (Uryu et al., 2003).
Finally, it has been proposed that head trauma may
trigger an acute-phase response, causing the overex- 57.3. Posttraumatic parkinsonism
pression of one or several proteins that would eventually
lead to cytoplasmic protein aggregation and cell death. In addition to considering trauma to the head as an epi-
This is in line with current theories suggesting that PD demiological risk factor in the etiology of PD, both cen-
is the result of a complex disorder of the ubiquitin-pro- tral lesions directly affecting the nigrostriatal tract and
teasome system involved in protein degradation. By those indirectly caused by severe cranial trauma and
analogy, there is some evidence that the synthesis of peripheral trauma have been linked to the development
apolipoprotein E, of -amyloid precursor protein or of parkinsonism.
both increases markedly in the brain after head injury,
leading to the trauma-related hypothesis of Alzheimers
57.3.1. Central lesions and parkinsonism
disease (Bower et al., 2003).
At the cellular and molecular level, the mechanisms
Single trauma to the head, as well as repetitive TBI
underlying secondary or delayed cell death following
such as that seen in boxers, can be the cause of second-
traumatic brain injury (TBI) are poorly understood.
ary forms of parkinsonism.
Experimental models of TBI suggest that diffuse and
widespread neuronal damage and loss are progressive
and prolonged for months to years after the initial 57.3.1.1. Parkinsonism caused by a single trauma
insult in selectively vulnerable regions of the cortex, to the head
hippocampus, thalamus, striatum and subcortical Parkinsonism is an infrequent complication of a single
nuclei (McIntosh et al., 1998). The cellular and mole- trauma to the head, although rare instances of this syn-
cular events triggered by TBI include possible altera- drome caused by penetrating injuries to the brainstem
tions in intracellular calcium with resulting changes by either a bullet or a knife have been reported (Koller
492 O. S. GERSHANIK
et al., 1989). The majority of authors conclude that the neuropathological examination performed in 16 cases
development of a parkinsonian syndrome after trauma presenting with mild to severe parkinsonian features
to the head is more often seen in cases in which the revealed multiple lesions in the rostral brainstem with
injury is severe enough to result in a comatose or unilateral or bilateral focal lesions in the substantia
vegetative state; in these cases, the resulting parkinso- nigra. Of importance is the fact that it was not possible
nian syndrome is usually associated with mental status to establish a clear correlation between the severity of
changes, cranial nerve dysfunction, pyramidal tract the pathology and the severity of the parkinsonian symp-
dysfunction and cerebellar disorders (Factor et al., tomatology. Moreover, there have been reports of signif-
1988; Jellinger, 2003). In Grimbergs seminal review icant pathological involvement of the substantia nigra
of 86 cases of posttraumatic parkinsonism reported following head injury without the development of a par-
between 1873 and 1922, he concluded that only 2 of kinsonian syndrome (Zijlmans et al., 2002; Jellinger,
the cases could be accepted as posttraumatic in nature 2003). In the case reported by Zijlmans et al. (2002), con-
as the trauma was severe enough to produce perma- firmation of nigrostriatal damage was obtained through
nent damage to specific areas of the brain; the trauma both MRI and dopamine transporter binding at the striatal
directly involved the head; and there was an unequivo- level. Imaging studies showed in the T2-weighted
cal temporal relationship between the head injury and sequence the presence of a hypointense lesion involving
the development of parkinsonism (Grimberg, 1934; the right anteromedial substantia nigra and the right sub-
Jankovic, 1994). Nevertheless, even in survivors of thalamic nucleus indicative of a previous hemorrhage.
severe head injury, the occurrence of parkinsonism is N-omega-flouropropyl-2beta-carboxymethoxy-3beta-
rare, as was the case in the large series reported by {4-iodophenyl} tropane single-photon emission com-
Krauss and Jankovic (2002). In a follow-up of 221 puted tomography ([123I]FP-CIT SPECT) binding was
out of 264 survivors of severe head injury who had completely abolished in the right striatum and caudate
been admitted to hospital with a Glasgow Coma Score nucleus, demonstrating an almost total loss of presynap-
of 8 or less, only 2 cases of parkinsonism were tic dopamine terminals. The absence of parkinsonian
observed (0.9%). Similarly, in Jellingers (1989) per- symptomatology could be explained, according to the
sonal series of 520 autopsy cases of parkinsonism authors, by the concomitant involvement of the ipsilat-
there were only 3 cases (0.6%) in which parkinsonism eral subthalamic nucleus (Zijlmans et al., 2002).
had developed after severe head injury with or without Since Grimbergs review and in addition to Krauss
persistent vegetative state (PVS) and was associated and Jellingers reports, there have been numerous pub-
with multiple brainstem damage resulting from trans- lications documenting isolated cases or small series of
tentorial herniation; all of those brains had bilateral patients presenting with parkinsonian features after
vascular necrotic lesions or anoxic scars in substantia sustaining head injuries causing varying degrees of
nigra and other parts of the brainstem or basal ganglia. brain dysfunction. Aside from a few cases due to direct
More recently, the same author (Jellinger, 2003) in a trauma to the midbrain (knife or bullet injury) or a
letter to the editor of the Journal of Neurology, Neuro- blunt head injury causing lesions at the basal ganglia
surgery and Psychiatry, commented on his observa- level (Nayernouri, 1985; Giroud et al., 1988; Koller
tions made in a series of 125 patients with PVS et al., 1989; Rondot et al., 1994; Doder et al., 1999;
following head injury and survival times ranging from Bhatt et al., 2000; Matsuda et al., 2003; Kivi et al.,
1 to 10 years. There were 49 survivors and, in 19 of 2005) (Table 57.2), the majority of the remaining
them, a mainly symmetrical, moderate to severe, cases are related to chronic subdural hematomas
parkinsonian syndrome was present. Treatment with (CSH), indirectly causing involvement of subcortical
levodopa induced partial or full improvement of symp- structures. In a review of the literature, Wiest et al.
tomatology in 15, whereas 4 patients showed complete (1999) were able to identify 16 published cases of par-
recovery from both PVS and parkinsonism. An addi- kinsonism secondary to CSH, in addition to their own
tional 15 patients, upon recovery from the initial case; 7 additional patients were indirectly cited by the
PVS, developed a progressive parkinsonian syndrome author as reported in two of his referenced papers. The
in which rigidity and bradykinesia were present in all association between CHS and movement disorders is
cases, whereas in only 6 of them was it associated with not frequent and has been estimated to occur in about
unilateral or bilateral resting tremor. Thirty-four of 2% of patients. The majority of the patients reported
the cases underwent magnetic resonance imaging were male, with an age range of 3883 years, and
(MRI) examination; in 32 of them, unilateral or bilat- presented within days to weeks after a trivial head
eral lesions in the midbrain were present. Of the origi- injury with variable clinical symptomatology (focal
nal series, 32 cases survived for at least 2 months deficit, general deterioration with headache, nausea,
without significant improvement of PVS until death; loss of sphincter control) associated to a parkinsonian
TRAUMA AND PARKINSONS DISEASE 493
Table 57.2
Posttraumatic parkinsonism

Author Year No. of cases Mechanism of head injury Associated features

Bruetsch et al. 1935 1 A 60-year-old man fell on a concrete surface, None mentioned
striking the occiput. Parkinsonian features
developed 8 months later. On autopsy, there
were fracture of the occipital and petrous
bones; depigmentation of the nigra, petechial
hemorrhages in the striatum (cited by Koller
et al., 1989)
Morsier GDz 1960 1 Bullet injury to the substantia nigra None mentioned
(cited by Koller et al., 1989)
Nayernouri 1985 1 A 37-year-old man received a blunt head injury Coma
causing coma. After 10 days the patient
regained consciousness and developed an
akinetic-rigid syndrome. A CT scan revealed
bilateral low-density lesions of the substantia
nigra
Giroud et al. 1988 1 A 61-year-old man sustained a facial Coma
concussion and became comatose. After
recovery he developed bilateral tremor at
rest, rigidity and bradykinesia. A CT scan
performed 6 hours after the trauma revealed
bilateral striatal hematomas
Rondot et al. 1994 1 A 38-year-old man attempted suicide with a Coma and right
firearm. The bullet lodged in the left hemiplegia, residual
midbrain. He recovered from coma over 15 hemiparesis with
days, developing resting and postural tremor spasticity, brisk tendon
associated with akinesia. A fluorodopa PET reflexes, Babinski sign,
scan showed reduced uptake in the left hemidystonia
striatum
Doder et al. 1999 1 A 36-year-old man sustained a skull fracture in Abulia, forgetfulness
a road accident; he remained unconscious for
24 hours. Six weeks later he developed a
coarse resting tremor and bradykinesia on the
right side. MRI showed a lesion involving
the left caudate and lenticular nucleus
Bhatt et al. 2000 3 Three males aged 35, 39 and 46, all involved in Coma
traffic accidents. Loss of consciousness with Right hemiparesis in one;
recovery after a variable period (hours30 disorientation, right-
days). Between 20 days and 5 months they sided weakness and
developed an asymmetric akinetic-rigid slurred incoherent
syndrome with rest tremor. There was speech in another
clinical improvement with levodopa. In all
there was variable, asymmetrical,
hemorrhagic involvement of the substantia
nigra and/or putamen (MRI)
Matsuda et al. 2003 3 Three males aged 14, 27 and 51, all involved in Coma
traffic accidents. All comatose on admission. Spasticity and cognitive
Patients remained in a persistent vegetative deterioration in one, left
state for 312 months. In all 3 cases there hemiparesis and oculo-
were asymmetrical rigidity, akinesia or motor palsy in another,
tremor, with asymmetrical brainstem injury non-parkinsonian motor
on MRI examination. Levodopa improved involvement and
both the motor and cognitive status of the cognitive deterioration
patients in another

(Continued)
494 O. S. GERSHANIK

Table 57.2
(Continued)

Author Year No. of cases Mechanism of head injury Associated features

Kivi et al. 2005 1 In a pedestrian accident a 59-year-old woman Hemiparesis, hyperreflexia


sustained a severe head injury with a and Babinskis sign
predominantly left brachiofacial hemiparesis.
Several weeks after the trauma, clinical signs
of a left-side hemiparkinsonism developed
(resting tremor, hypophonia, hypomimia).
Axial MRI (FLAIR) of the midbrain of the
patient showed a hypointense lesion of the
anterior part of the red nucleus and posterior
part of the substantia nigra, indicating a
hemosiderin deposit due to a previous
hematoma

CT, computed tomography; PET, positron emission tomography; MRI, magnetic resonance imaging; FLAIR, fluid-attenuated inversion
recovery.

syndrome (in two-thirds of cases a complete parkinso- parkinsonism, except in those cases in whom damage
nian syndrome was present). In a few cases, aside from to the nigrostriatal tract can be demonstrated, either
mild headache, the only symptomatology present was through imaging techniques or at autopsy. The reasons
pure parkinsonism. Surgical evacuation of the hemato- for this delay can be diverse; the initial brain injury
mas resulted in complete remission of the symptoma- caused by the trauma can set in motion a cascade of
tology in the majority of cases. In a few cases there biochemical and metabolic changes that may lead to
was residual symptomatology (both parkinsonian and further neuronal damage; or in turn, the delay may
non-parkinsonian) after surgery. Incomplete remission be necessary for the development of sprouting, rem-
was more frequently observed in bilateral hematomas. yelinization, ephaptic transmission or central synaptic
Several mechanisms have been proposed to explain reorganization (Jankovic, 1994).
the development of secondary parkinsonism to CSH:
among them, direct mechanical compression of the 57.3.1.2. Parkinsonism caused by repetitive trauma
basal ganglia, interference with its vascular supply or to the head (boxers encephalopathy)
metabolic changes. Hemispheric displacement due to A peculiar syndrome secondary to chronic TBI asso-
the hematoma may lead to distortion and compression ciated with boxing occurs in approximately 1820% of
of the midbrain, causing potentially reversible distur- professional boxers (Friedman, 1989). It was Martland
bances at different levels of the nigrostriatal pathway. who in 1928 first described this syndrome in the medical
However, most patients with a subdural hematoma of literature, followed by numerous reports that helped
similar location do not develop parkinsonism, suggest- delineate the syndrome, including the presence of parkin-
ing, in the view of some authors, that those patients sonian symptomatology ((Critchley, 1957; Roberts,
who in fact develop a parkinsonian syndrome are espe- 1969), cited by Factor et al., 1988; Stern, 1991). It is
cially vulnerable to injury to the dopaminergic system usually referred to in the literature as boxers encephalo-
or are already in the preclinical stages of PD (Trosch pathy, punch-drunk syndrome or dementia pugilis-
and Ransom, 1990; Hageman and Horstink, 1994; tica, and includes parkinsonian features and varying
Wiest et al., 1999). degrees of cognitive and behavioral impairments in its
In the majority of cases of posttraumatic parkinson- clinical presentation (Jordan, 2000). It has been reported
ism reported in the literature, the parkinsonian sympto- to be more common in second-rate and slugging-type
matology did not develop immediately after the fighters (Guterman and Smith, 1987). Incidence and
trauma. A delay of several days, weeks or even months severity correlate with the length of the boxing career,
is usually the case, sometimes making it difficult to number of bouts and increased sparring (Guterman and
establish a clear-cut cause-and-effect relationship Smith, 1987; Jordan, 2000; Krauss and Jankovic, 2002).
between the traumatic event and the development of The parkinsonian features of this syndrome include gait
TRAUMA AND PARKINSONS DISEASE 495
disequilibrium, cogwheel rigidity, bradykinesia, hypo- may cause stretching and rupturing of vessels, focal
phonia and even a resting tremor; other neurologic man- ischemia, petechial hemorrhages, axonal injury and
ifestations frequently present are: dementia, cerebellar edema. It is possible that the long axons of the nigrostria-
dysfunction, marked dysarthria and pyramidal tract signs tal tract are particularly vulnerable to the effects of
(Stern, 1991; Krauss and Jankovic, 2002). Neurological mechanical shearing produced by trauma to the head
symptoms usually become manifest long after the end (Jankovic, 1994). Several in vivo studies have explored
of the boxing career, on average 16 years after the last the functional consequences of these lesions; proton
fight (Jankovic, 1994). The encephalopathy may run a magnetic resonance spectroscopy studies have demon-
progressive course or remain stable at any level. Three strated a significant reduction in the concentration of
stages of clinical deterioration have been described, con- N-acetylaspartate in the striatum of patients with
sisting initially of affective disturbances and additional boxers encephalopathy, suggesting the existence of
psychiatric symptoms; later on there is worsening of the terminal field involvement and striatal neuronal loss
psychiatric manifestations and parkinsonism gradually in addition to nigral damage (Davie et al., 1995).
develops. Finally in the last stage there is significant PET may help in differentiating these cases from
decrease of cognitive function together with signs of idiopathic parkinsonism, as in patients with boxers
more extensive neurological involvement (cerebellar encephalopathy mean caudate and putamen [18F]dopa
and pyramidal) (Guterman and Smith, 1987). uptake is similarly reduced by approximately 40%
The neuropathology of boxers encephalopathy was when compared to controls and mean caudate uptake
classically described by Corsellis et al. (1973) in a is much lower than that observed in idiopathic parkin-
postmortem study of 15 boxers. In addition to pete- sonism, suggesting that in boxers encephalopathy
chial hemorrhages in the cortex and brainstem and there is a uniform loss of nigrostriatal terminal
gliosis of the cerebellum, he found marked loss of pig- function (Turjanski et al., 1997).
mented neurons in the substantia nigra, especially in
the lateral and intermediate cell groups, without Lewy 57.3.2. Peripheral lesions and parkinsonism
bodies. Numerous neurofibrillary tangles without
senile plaques were present in the cerebral cortex. Cor- The role of peripheral trauma in the development of
sellis cases were more recently re-examined using secondary parkinsonism is still an unresolved issue
immunocytochemical methods and an antibody raised and evidence supporting this relationship is scarce
to the beta-protein present in Alzheimers disease pla- and inconclusive (Factor et al., 1988; Koller et al.,
ques. In all cases with substantial tangle formation 1989). In Factor et al.s (1988) review of the historical
there was evidence of extensive beta-protein immunor- concept of trauma as etiology of parkinsonism, most of
eactive deposits (plaques). These diffuse plaques the cases of peripherally induced parkinsonism
were not visible with Congo red or standard silver reported in the late 19th and early 20th centuries could
stains, which may explain Corsellis failure to detect be dismissed as not related for lack of conclusive
the presence of senile plaques. The degree of beta-pro- evidence and probably stimulated by the passing of
tein (amyloid component) deposition was comparable workmens compensation laws. Even Charcots
to that seen in Alzheimers disease (Roberts et al., ascending neuritis hypothesis of the mechanism
1990). These data suggest that Alzheimers disease by which peripheral trauma could lead to the develop-
and boxers encephalopathy may share common ment of parkinsonism, although convenient, does
pathogenetic mechanisms and perhaps its development not resist a thorough analysis (Factor et al., 1988).
is linked to the presence of a genetic predisposition Nevertheless, more recently, Schott (1986), Jankovic
(Stern, 1991; Jankovic, 1994; Jordan, 2000; Krauss (1994) and Cardoso and Jankovic (1995) contributed
and Jankovic, 2002). Recent studies in professional to the re-emergence of the hypothesis that peripheral
boxers, establishing a correlation between the presence trauma could somehow contribute to the etiology of
of the e4 allele and significantly greater scores on a parkinsonism.
scale measuring chronic encephalopathy, lend support In Schotts review (1986) of 10 patients in whom
to this hypothesis (Jordan et al., 1997). The observed various involuntary movement disorders developed
neuropathology of this syndrome is presumed to be after trauma that was predominantly or entirely periph-
the result of the cumulative effects of repeated subcli- eral, 3 of the cases corresponded to parkinsonism. The
nical concussions secondary to rotational and shearing interval between injury and onset of parkinsonian
acceleration forces induced by direct blows to the head symptomatology ranged from 48 hours to 4 weeks;
(Krauss and Jankovic, 2002). The rapid rotation of the injured and painful part was the area initially
the cranium in relation to a relatively stationary brain affected by involuntary movements, although more
496 O. S. GERSHANIK
widespread involvement subsequently occurred. These [18F]dopa PET scan showed asymmetrically reduced
clinical features, which resembled the phenomena putamenal uptake. Speculations on the mechanism of
experienced by some patients with causalgia, led the action of electrical injury as a triggering factor for
author to believe in the possibility of common the development of parkinsonism are similar to those
mechanisms. originally postulated by Jankovic (1994) and Cardoso
Cardoso and Jankovic (1995), in reviewing the and Jankovic (1995). Quinn and Maraganore (2000),
records of patients evaluated in the Movement Disor- in a letter to the editor discussing Morris et al.s
ders Clinic at Baylor College of Medicine, Houston, (1998) report, strongly question the validity of their
TX, USA between 1977 and 1993, found 28 patients assertions on the basis of epidemiological data and
in whom the onset of tremor, parkinsonism or both biological plausibility.
was antatomically and temporally related to local
injury. Eleven of these patients had parkinsonian fea- 57.4. Worsening of parkinsonism by trauma
tures in addition to tremor (both postural and resting);
in a few cases, dystonia, myoclonus and a complex Anecdotal reports referred in the literature and in the
regional pain syndrome were also present. In all cases authors own experience support the notion that
the development of parkinsonian symptoms was tem- trauma, as well as other medical conditions (such as
porally related to the occurrence of trauma (latency infections and surgery) may occasionally worsen par-
to onset 46.6  56.0 days) and the anatomical region kinsonian symptomatology in patients with an estab-
where the symptoms began was that involved in the lished diagnosis of PD (Goetz and Stebbins, 1991;
peripheral injury; nevertheless the movement disorder Wiest et al., 1999; Chou and Gutmann, 2001). Goetz
displayed a tendency to spread beyond the original site and Stebbins (1991) followed 10 parkinsonian patients
of injury. In 3 of the patients [18F]dopa PET uptake in who sustained trauma to the head in motor vehicle
the striatum was decreased, similar to what is found in accidents; in all cases disability significantly increased
classical idiopathic parkinsonism; however, only 6 of immediately after trauma, to return to baseline levels
the patients responded to levodopa therapy; a family in the ensuing weeks. This transient deterioration did
history of essential tremor was present in 2. It could not cause persistent increased disability or an altera-
be argued that the occurrence of parkinsonism after tion in the natural course of the disease. In Wiest
peripheral trauma was purely coincidental or that in et al.s (1999) and Chou and Gutmanns (2001) cases
fact peripheral trauma could unmask subclinical par- with a long-standing history of levodopa-responsive
kinsonism or worsen pre-existing subtle parkinsonian PD, the presence of rapid deterioration of pre-existing
symptoms that had been previously undetected. The symptomatology in the context of frequent falls and
possibility that peripheral injury could somehow in- occasional head trauma suggested the possibility of
duce plastic changes and central motor reorganization subdural hematoma. Imaging studies confirmed the
through modifications in spinal excitability, retrograde presence of subdural hematoma in both cases. After
axonal transport of neurotrophic factors or upregula- surgical evacuation of the hematomas the patients
tion of immediate early-response genes was discussed returned to their baseline levels in a few days.
by Cardoso and Jankovic (1995). There is, however, no clear explanation to account
Using the criteria of Jankovic (1994) and Cardoso for this phenomenon; on the other hand, there is
and Jankovic (1995) for the diagnosis of parkinsonism experimental evidence that in behaviorally normal rats
induced by a peripheral injury, Morris et al. (1998) with partial lesions of the nigrostriatal rats, a stressful
reported on a case of parkinsonism following electrical stimulus (glucose deprivation, electrical tail shock or
injury to the hand and reviewed 9 additional cases exposure to severe cold) may unmask a previously
reported in the literature. Their case was a 39-year- undetected motor deficit (akinesia) (Snyder et al.,
old man who sustained an electrical injury to his right 1985). It may be argued that in these animals the
hand; 34 weeks later he developed progressive brady- stressful stimuli triggered a series of biochemical
kinesia and rigidity initially confined to the right upper events that altered a precarious compensatory state.
extremity. Upon examination 3 years later the parkin- Similarly, the degree of clinical disability in PD
sonian symptomatology had spread, involving the ipsi- patients may be the result of a balance between the
lateral lower extremity and the contralateral arm. degree of nigrostriatal damage, compensatory en-
Levodopa significantly improved the symptomatology; dogenous mechanisms and medication. Any stress-
however, the patient could not obtain sustained benefit ful stimuli, be it trauma, infection or surgery, may
because of gastric intolerance. An MRI failed to break this delicate balance and cause transient
demonstrate the presence of a central lesion and an increased disability.
TRAUMA AND PARKINSONS DISEASE 497
57.5. Conclusions Corti O, Hampe C, Darios F et al. (2005). Parkinsons dis-
ease: From causes to mechanisms. C R Biol 328 (2):
After careful evaluation of all the evidence reviewed 131142.
thus far we may conclude: Davie CA, Pirtosek Z, Barker GJ et al. (1995). Magnetic
resonance spectroscopic study of parkinsonism related to
1. Although several case-control and two cohort studies
boxing. J Neurol Neurosurg Psychiatry 58 (6): 688691.
failed to establish that trauma to the head can influ- De Michele G, Filla A, Volpe G et al. (1996). Environmental
ence the subsequent development of PD, others have, and genetic risk factors in Parkinsons disease: A case-
in fact, found that severe head trauma increases the control study in Southern Italy. Mov Disord 11 (1): 1723.
risk of PD. It is perhaps valid to assume that history De Rijk MC, Bretleler MMB, Ott A et al. (1996). Risk fac-
of preceding head trauma, together with family his- tors for Parkinsons disease: The Rotterdam Study. J Neu-
tory and other environmental factors, plays at least rol 243 (Suppl 2): S8.
a partial role in the etiology of PD. Doder M, Jahanshahi M, Turjanski N et al. (1999). Parkin-
2. Albeit infrequent, posttraumatic parkinsonism sons syndrome after closed head injury: A single case
resulting from direct injury to the basal ganglia report. J Neurol Neurosurg Psychiatry 66 (3): 380385.
and substantia nigra or indirect involvement of Factor SA, Weiner WJ (1991). Prior history of head trauma
in Parkinsons disease. Mov Disord 6 (3): 225229.
these structures secondary to a severe cranial
Factor SA, Sanchez-Ramos J, Weiner WJ (1988). Trauma as
trauma does indeed exist. an etiology of parkinsonism: A historical review of the
3. Repetitive cranial trauma such as that experienced concept. Mov Disord 3 (1): 3036.
by boxers may result in the development of a form Fearnley JM, Lees AJ (1991). Ageing and Parkinsons dis-
of parkinsonism associated with other motor, ease: Substantia nigra regional selectivity. Brain 114:
behavioral and cognitive disorders encompassed 22832301.
in boxers encephalopathy. Friedman JH (1989). Progressive parkinsonism in boxers.
4. The possible occurrence of parkinsonism induced South Med J 82 (5): 543546.
by peripheral trauma is still controversial and Giroud M, Vincent MC, Thierry A et al. (1988). Parkinsonian
there is not enough supportive evidence to accept syndrome caused by traumatic hematomas in the basal
its existence. ganglia. Neurochirurgie 34 (1): 6163.
Godwin-Austen RB, Lee PN, Marmot MG et al. (1982).
5. Trauma, as well as infectious disorders, surgical
Smoking and Parkinsons disease. J Neurol Neurosurg
interventions and stress, may aggravate pre- Psychiatry 45 (7): 577581.
existing PD. Goetz CG, Stebbins GT (1991). Effects of head trauma from
motor vehicle accidents on Parkinsons disease. Ann Neu-
References rol 29: 191193.
Graves AB, White E, Koepsell TD et al. (1990). The asso-
Allam MF, del Castillo AS, Navajas RF (2003). Parkinsons ciation between head trauma and Alzheimers disease.
disease risk factors. Rev Neurol 36 (8): 749755. Am J Epidemiol 131 (3): 491501.
Bhatt M, Desai J, Mankodi A et al. (2000). Posttraumatic Grimberg L (1934). Paralysis agitans and trauma. J Nerv
akinetic-rigid syndrome resembling Parkinsons disease: Ment Dis 79: 1442.
a report on three patients. Mov Disord 15 (2): 313317. Guterman A, Smith RW (1987). Neurological sequelae of
Bower JH, Maraganore DM, Peterson BJ et al. (2003). Head boxing. Sports Med 4 (3): 194210.
trauma preceding PD: a case-control study. Neurology Hageman AT, Horstink MW (1994). Parkinsonism due to a
60 (10): 16101615. subdural hematoma. Mov Disord 9 (1): 107108.
Bramlett HM, Dietrich WD (2003). Synuclein aggregation: pos- Hofman A, Collette HJ, Bartelds AI (1989). Incidence and
sible role in traumatic brain injury. Exp Neurol 184: 2730. risk factors of Parkinsons disease in The Netherlands.
Butterfield PG, Valanis BG, Spencer PS et al. (1993). Envir- Neuroepidemiology 8 (6): 296299.
onmental antecedents of young-onset Parkinsons disease. Jankovic J (1994). Post-traumatic movement disorders:
Neurology 43: 11501158. Central and peripheral mechanisms. Neurology 44 (11):
Calne DB, Langston JW (1983). Etiology of Parkinsons dis- 20062014.
ease. Lancet 2: 14571459. Jellinger K (1989). Trauma as an etiologic agent in Parkinson
Cardoso F, Jankovic J (1995). Peripherally induced tremor disease. Mov Disord 4 (1): 9092.
and Parkinsonism. Arch Neurol 52: 263270. Jellinger KA (2003). Parkinsonism and persistent vegetative
Chou SM, Gutmann L (2001). Deteriorating parkinsonism state after head injury. J Neurol Neurosurg Psychiatry
and subdural hematomas. Neurology 57 (7): 1925. 75 (7): 1082.
Corsellis JA, Bruton CJ, Freeman-Browne D (1973). The Jordan BD (2000). Chronic traumatic brain injury associated
aftermath of boxing. Psychol Med 3 (3): 270303. with boxing. Semin Neurol 20 (2): 179185.
498 O. S. GERSHANIK

Jordan BD, Relkin NR, Ravdin LD et al. (1997). Apolipopro- Patrick HU, Levy PM (1922). Parkinsons disease. A clinical
tein E epsilon4 associated with chronic traumatic brain study of one hundred and forty-six cases. Arch Neurol
injury in boxing. JAMA 278 (2): 136140. Psychiat 7:711720.
Kivi A, Trottenberg T, Kupsch A et al. (2005). Levodopa- Preux PM, Condet A, Anglade C et al. (2000). Parkinsons
responsive posttraumatic parkinsonism is not associated disease and environmental factors. Matched case-control
with changes of echogenicity of the substantia nigra. study in the Limousin region, France. Neuroepidemiology
Mov Disord 20 (2): 258260. 19 (6): 3337.
Koller WC, Wong GF, Lang A (1989). Posttraumatic move- Quinn N, Maraganore D (2000). Parkinsonism following
ment disorders: A review. Mov Disord 4 (1): 2036. electrical injury to the hand. Mov Disord 15 (3): 587588.
Krauss JK, Jankovic J (2002). Head injury and posttraumatic Roberts GW, Allsop D, Bruton C (1990). The occult after-
movement disorders. Neurosurgery 50 (5): 927939. math of boxing. J Neurol Neurosurg Psychiatry 53 (5):
Kuopio AM, Marttila RJ, Helenius H et al. (1999). Environ- 373378.
mental risk factors in Parkinsons disease. Mov Disord Rondot P, Bathien N, de Recondo J et al. (1994). Dystonia-
14 (6): 928939. parkinsonism syndrome resulting from a bullet injury in
Lai BC, Marion SA, Teschke K et al. (2002). Occupational the midbrain. J Neurol Neurosurg Psychiatry 57 (5): 658.
and environmental risk factors for Parkinsons disease. Schwab RS, England AC (1968). Parkinsonism due to var-
Parkinsonism Relat Disord 8 (5): 297309. ious specific causes. In: Vinken PJ, Bruyn GW, (Eds.),
Lees AJ (1997). Trauma and Parkinson disease. Rev Neurol Handbook of Clinical Neurology, 1st. edn, Vol. 6. North
153 (10): 541546. Holland Publishing Co, Amsterdam, pp. 230233.
Liu B, Gao HM, Hong JS (2003). Parkinsons disease and Schott GD (1986). Induction of involuntary movements by
exposure to infectious agents and pesticides and the occur- peripheral trauma: An analogy with causalgia. Lancet
rence of brain injuries: Role of neuroinflammation. 2 (8509): 712716.
Environ Health Perspect 111 (8): 10651073. Seidler A, Hellenbrand W, Robra BP et al. (1996). Possible
Martland HS (1928). Punch drunk. JAMA 91:11031107. environmental, occupational, and other etiologic factors
Matsuda W, Matsumura A, Komatsu Y et al. (2003). Awa- for Parkinsons disease: A case-control study in Germany.
kenings from persistent vegetative state: Report of three Neurology 46 (5): 12751284.
cases with parkinsonism and brain stem lesions on MRI. Semchuk KM, Love EJ, Lee RG (1993). Parkinsons disease:
J Neurol Neurosurg Psychiatry 74 (11): 15711573. A test of the multifactorial etiologic hypothesis. Neurol-
McCann SJ, LeCouteur DG, Green AC et al. (1998). The ogy 43 (6): 11731180.
epidemiology of Parkinsons disease in an Australian Smargiassi A, Mutti A, De Rosa A et al. (1998). A case-con-
population. Neuroepidemiology 17 (6): 310317. trol study of occupational and environmental risk factors
McIntosh TK, Saatman KE, Raghupathi R et al. (1998). The for Parkinsons disease in the Emilia-Romagna region of
Dorothy Russell Memorial Lecture. The molecular and Italy. Neurotoxicology 19 (45): 709712.
cellular sequelae of experimental traumatic brain injury: Snyder AM, Stricker EM, Zigmond MJ (1985). Stress-
Pathogenetic mechanisms. Neuropathol Appl Neurobiol induced neurological impairments in an animal model of
24: 251267. parkinsonism. Ann Neurol 18: 544551.
Morano A, Jimenez-Jimenez FJ, Molina JA et al. (1994). Risk- Stern M, Dulaney E, Gruber SB et al. (1991). The epide-
factors for Parkinsons disease: Case-control study in the pro- miology of Parkinsons disease. A case-control study of
vince of Caceres, Spain. Acta Neurol Scand 89 (3): 164170. young-onset and old-onset patients. Arch Neurol 48 (9):
Morris HR, Moriabadi NF, Lees AJ et al. (1998). Parkinson- 903907.
ism following electrical injury to the hand. Mov Disord Stern MB (1991). Head trauma as a risk factor for Parkin-
13 (3): 600602. sons disease. Mov Disord 6 (2): 9597.
Morrish PK, Rakshi JS, Bailey DL et al. (1998). Measuring Tanner CM, Chen B, Wang WZ et al. (1987). Environmental
the rate of progression and estimating the preclinical per- factors in the etiology of Parkinsons disease. Can J Neu-
iod of Parkinsons disease with [18F]dopa PET. J Neurol rol Sci 14 (3 Suppl): 419423.
Neurosurg Psychiatry 64: 314319. Taylor CA, Saint-Hilaire MH, Cupples LA et al. (1999).
Nayernouri T (1985). Posttraumatic parkinsonism. Surg Neu- Environmental, medical, and family history risk factors
rol 24 (3): 263264. for Parkinsons disease: A New England-based case con-
Nuti A, Ceravolo R, DellAgnello G et al. (2004). Environ- trol study. Am J Med Genet 88 (6): 742749.
mental factors and Parkinsons disease: A case-control Trosch RM, Ransom BR (1990). Levodopa-responsive par-
study in the Tuscany region of Italy. Parkinsonism Relat kinsonism following central herniation due to bilateral
Disord 10: 481485. subdural hematomas. Neurology 40 (2): 376377.
OMIM Online Mendelian Inheritance in Man Database, Tsai CH, Lo SK, See LC et al. (2002). Environmental risk
2005. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db factors of young onset Parkinsons disease: A case-control
OMIM study. Clin Neurol Neurosurg 104 (4): 328333.
Parkinson J (1817). An Essay on the Shaking Palsy. Whit- Turjanski N, Lees AJ, Brooks DJ (1997). Dopaminergic
tingham and Rowland, for Sherwood Neely and Jones, function in patients with posttraumatic parkinsonism: An
London. 18F-dopa PET study. Neurology 49 (1): 183189.
TRAUMA AND PARKINSONS DISEASE 499

Uryu K, Giasson BI, Longhi L et al. (2003). Age-dependent Williams DB, Annegers JF, Kokmen E et al. (1991). Brain
synuclein pathology following traumatic brain injury in injury and neurologic sequelae: A cohort study of demen-
mice. Exp Neurol 184 (1): 214224. tia, parkinsonism, and amyotrophic lateral sclerosis. Neu-
Ward CD, Duvoisin RC, Ince SE et al. (1983). Parkinsons rology 41 (10): 15541557.
disease in 65 pairs of twins and in a set of quadruplets. Yang J, Wu Z, Lou X (1994). A case-control study on risk
Neurology 33: 815824. factors in etiology of Parkinsons disease. Zhonghua Liu
Werneck AL, Alvarenga H (1999). Genetics, drugs and Xing Bing Xue Za Zhi 15 (1): 69.
environmental factors in Parkinsons disease: A case-con- Zijlmans J, Booij J, Valk J et al. (2002). Posttraumatic tre-
trol study. Arq Neuropsiquiatr 57 (2-B): 347355. mor without parkinsonism in a patient with complete con-
Wiest RG, Burgunder JM, Krauss JK (1999). Chronic sub- tralateral loss of the nigrostriatal pathway. Mov Disord
dural haematomas and Parkinsonian syndromes. Acta 17 (5): 10861088.
Neurochir (Wien) 141 (7): 753757.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 58

Psychogenic parkinsonism

VASILIKI KOUKOUNI AND KAILASH P. BHATIA*

Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London,
London, UK

58.1. Definition the Toronto Western Hospital, over a period of 20


months, 64 patients were diagnosed with PMD and
The term psychogenic is derived from a Greek word psychogenic parkinsonism accounted for 6.1% of
meaning created by the soul and is applied to dis- these (Miyasaki et al., 2003). In another series from
eases that cannot be attributed to a certain lesion or Columbia-Presbyterian Medical Center, New York,
dysfunction of a system. Parkinsonism is defined by USA, including 131 patients, the respective figure
a constellation of features, the hallmark being brad- was 1.9% (Fahn and Williams, 1988).
ykinesia associated with tremor, rigidity and impair-
ment of postural reflexes. The most common cause is 58.3. Natural history
Parkinsons disease (PD). There are other forms of
parkinsonism, including the so-called atypical parkin- In 1988, Walters et al. described the case of a 64-
sonian conditions, like multiple systems atrophy year-old man who presented with atypical symptoms
(MSA) or progressive supranuclear palsy (PSP), and of parkinsonism that resolved spontaneously 1 year
also acquired or secondary forms of parkinsonism. after the discontinuation of levodopa. Since then,
Some of them can be accompanied by cognitive dec- two large studies have been performed regarding psy-
line and psychiatric manifestations. Although psy- chogenic parkinsonism. Of the 14 cases of psycho-
chogenic forms of a variety of different movement genic parkinsonism described by Lang et al. (1995)
disorders, such as psychogenic dystonia or psycho- in three academic movement disorders centers, the
genic tremor, have been well described in the literature average age of the patients was 47 years (range 21
over the years, little is mentioned about psychogenic 63 years). Both sexes were equally involved, in con-
parkinsonism. trast to the other PMD, where there is usually a
female predisposition (Fahn and Williams, 1988;
58.2. Epidemiology Koller et al., 1989; Monday and Jankovic, 1993;
Kim et al., 1999). The mean duration of the symp-
Psychogenic parkinsonism is perhaps the rarest of the toms prior to the diagnosis was 5 years, ranging from
different forms of the psychogenic movement disor- 4 months to 13 years. In one recent study, reported
ders (PMD). In retrospective studies conducted in only in abstract form, from three large movement dis-
large movement disorder clinics it accounted for order centers in Augusta, GA, USA and London, UK,
only 1.97% of all PMDs (Fahn and Williams, 1988; a retrospective chart review of 9 patients diagnosed
Factor et al., 1995; Miyasaki et al., 2003) and 0.5% with psychogenic parkinsonism was reported
or less of all patients presenting with parkinsonism (Morgan et al., 2004). The mean age of onset of the
(Factor et al., 1995; Lang et al., 1995). In 1995, Factor symptoms was 50 years, with a range of 3472
et al. described 28 patients with PMDs, 2 of whom years, and the disease duration varied from several
were diagnosed as having psychogenic parkinson- months to 28 years. Interestingly, there was a 2:1
ism (7%). At the Movement Disorders Clinic of male-to-female ratio.

*Correspondence to: Prof. Kailash Bhatia, Institute of Neurology, Sobell Department of Motor Neuroscience and Movement
Disorders, 7 Queen Square, London WC1N 3BG, UK. E-mail: kbhatia@ion.ucl.ac.uk, Tel: 44-20-7837-3611, Ext: 4253,
Fax: 44-20-7676-2175.
502 V. KOUKOUNI AND K. P. BHATIA
58.4. Clinical manifestations suggested that there is a co-activation of agonist and
antagonist muscles in psychogenic tremor that is sup-
The onset of the disease is abrupt in the vast majority ported by electromyographic (EMG) analysis and clin-
of patients, which is unlike typical PD. According to ical evaluation, which can produce a cogwheel-like
the studies performed by Lang et al. (1995) and Mor- resistance to passive movement, which subsides when
gan et al. (2004), there was an abrupt onset in about the patient is relaxed.
two-thirds of patients (71.2% and 78% respectively). True bradykinesia with typical arrests in movement
Precipitating factors such as work-related injuries and is never observed, although all the patients in Langs
motor vehicle accidents with minor injuries that possi- series (Lang et al., 1995) demonstrated slowness of
bly may not have involved the head are a common voluntary movements and in some of them this was
feature (Lang et al., 1995). A total of 43% of the patients their initial and main complaint. Slowness is present
studied by Lang and 77% of those studied by Morgan throughout the performance of rapid repetitive and
presented with unilateral symptoms, typically involving alternating movements, but without the fatiguing and
the dominant-hand side. decreasing amplitude that is seen in true parkinsonian
The most prominent feature is tremor, which bore patients. The patients often seem to struggle and put
all the characteristics of psychogenic tremor. Classical more effort than needed into performing the tasks. They
PD tremor is asymmetric, pill-rolling, resting tremor at are grimacing, sighing and look exhausted and may use
a rate of 34 Hz, involving the thumb and index fin- their whole body in order to do a minor movement.
ger. A faster tremor of 67 Hz can appear on keeping Despite the severe slowness, some patients may demon-
the arms outstretched. On the other hand, psychogenic strate exquisite ability in doing specific tasks, like writ-
tremor can be present at rest and persist even during ing with normal speed (Lang et al., 1995). Some of the
position and intentional movements, at the same fre- patients seen by Lang et al. (1995) demonstrated facial
quency (Koller et al., 1989). Although wrists, elbows masking, but in the majority of them it was attributed
and shoulders are commonly affected, fingers are to an underlying depression.
rarely involved, unlike typical PD (Deuschl et al., Walking can be slow and stiff, with reduced or even
1998). Another important characteristic is variability. absent arm-swing on the affected side. The arm may
The tremor may increase when the attention is drawn be held tightly extended and adducted to the side and
to the affected limb or when the patient is asked about this can persist while running. The patients usually
it and can improve dramatically or even disappear respond in a theatrical way on postural stability test-
when the patients attention is withdrawn from the ing, flinging up both their arms (even the affected
area involved (Campbell, 1979) or if the patient is dis- one) equally and symmetrically and retropulsing but
tracted by mental activity (e.g. mental arithmetic) or never falling (Lang et al., 1995). An exaggerated star-
complex motor tasks (e.g. finger-to-nose and tandem tle, when the backwards pull test is performed to test
walking) (Elble and Koller, 1990). Parkinsonian rest postural stability, is often seen.
tremor, e.g. in PD, quite often appears when the Another important feature noted by both studies
patient is walking or being distracted. Entrainment, done by Lang et al. (1995) and Morgan et al. (2004)
meaning change of the original tremor frequency to is that disability is usually maximal from the early
match the frequency of a repetitive task performed in stages of the disease and affects all aspects of every-
another limb (e.g. tapping) or side-to-side tongue day life. Most patients give up work and are some-
movements (Kim et al., 1999), is also common. times unable to perform their normal daily activities.
Koller and Biary (1989) suggested that PD patients Most patients also complain of multiple other somatic
are capable of voluntarily suppressing their tremor for symptoms, like generalized fatigue, non-specific pains,
an average of 48 seconds, if they are allowed to con- memory disturbance and impaired vision, and have signs
centrate on it, in contrast to the psychogenic patients like give-away weakness of a limb or non-anatomic
where the opposite tends to happen. The psychogenic sensory loss (Lang et al., 1995).
tremor can also vary in terms of direction (e.g. from Fahn and Williams (1988) published a classification
supinationpronation orientation to flexionextension) of psychogenic dystonia, which can be applied to all
(Koller et al., 1989) and amplitude (e.g. when a limb is PMDs. According to this, a movement disorder can
weighed) (Deuschl et al., 1998). be classified as: (1) documented, meaning symptoms
As pointed out by Lang et al. (1995), limb rigidity are relieved by psychotherapy, suggestion or placebo
is uncommon and, when it is present, the examiner or the patient is witnessed as being free of symptoms
has a sense of voluntary resistance. This also subsides when left alone; (2) clinically established, meaning
when the patient is distracted. Deuschl et al. (1998) the movement disorder is inconsistent over time or is
PSYCHOGENIC PARKINSONISM 503
incongruent with the classic condition and any of the people employed in health professions (Koller et al.,
following are present: other definitely psychogenic 1989; Fahn, 1994; Kim et al., 1999) and in those who
neurologic signs, multiple somatizations or an obvious have witnessed the disorder in other family members
psychiatric disturbance; (3) probably psychogenic, (Koller et al., 1989). Elements in clinical examination
meaning that movements are inconsistent or incongru- are also revealing as well as responses to therapeutic
ent with the classic movement disorder but no other procedures (Table 58.2). The use of placebo as diag-
features provide support for a diagnosis of psychogeni- nostic and therapeutic procedure remains controver-
city or that the movement disorder is consistent and sial, as it gives rise to ethical considerations and can
congruent with the classic disorder but other features affect the physicianpatient relationship. It is sug-
of psychogenicity are present; and (4) possibly psycho- gested by Sa et al. (2004) that the use of placebo
genic, meaning it is suspected that the movements are should be reserved for cases in which the diagnosis
psychogenic if an obvious emotional disturbance is of psychogenicity remains uncertain after a thorough
present. Most psychogenic PD patients would fall into investigation and follow-up. It can also prove useful
the documented or clinically established categories. in the differential diagnosis of complex movement dis-
orders and in cases where organic and psychogenic
58.5. Diagnosis etiologies coexist (Monday and Jankovic, 1993; Koller

The first clues of psychogenicity in a patient present- Table 58.2


ing with parkinsonism can be obtained by history
Clues suggesting that a movement disorder may be
(Table 58.1). This may include a history of a psychia-
psychogenic (Miyasaki et al., 2003; Sa et al., 2004)
tric disease or a long medical history involving multi-
ple operations, admissions to hospitals and a long Historical
catalog of diagnostic tests previously done. Events
prior to the onset of the disease, such as accidents, 1. Abrupt onset
stressful situations or work-related injuries with litiga- 2. Static course
tion or compensation pending, can be indicative of a 3. Spontaneous remissions
possible non-organic cause of the symptoms. An 4. Obvious psychiatric disturbance
5. Multiple somatizations
impressive fact is that PMDs are more frequent in
6. Employed in health profession
7. Pending litigation or compensation
Table 58.1 8. Presence of secondary gain
9. Young age
Clues suggesting psychogenic parkinsonism (Lang et al.,
1995) Clinical

Onset Often abrupt 1. Inconsistent character of movement (amplitude,


frequency, distribution, selective ability)
Course Static, maximum disability early 2. Paroxysmal movement disorder
Distribution Dominant side most affected 3. Movements increase with attention or decrease with
Tremor Rest, postural, action: reduces with distraction
distraction/ concentration, increases 4. Ability to trigger or relieve the abnormal movements
with attention with unusual or non-physiological interventions. (e.g.
Rigidity Voluntary resistance, cogwheeling trigger points of the body)
absent, may decrease with distraction 5. False weakness
Bradykinesia No true fatiguing, marked slowness, 6. False sensory complaints
bizarre features 7. Self-inflected injuries
Gait Atypical, arm held stiffly at side, 8. Deliberate slowness of movements
antalgic if pain is associated 9. Functional disability out of proportion to exam findings
Postural stability Extreme or bizarre responses to 10. Movement abnormality that is bizarre, multiple or
minimal displacement difficult to classify
Other neurologic False weakness, non-anatomic sensory
features loss Therapeutic responses
Psychiatric Varied, usually evident but accurate
features definition not always possible 1. Unresponsive to appropriate medications
Other Litigation, compensation (receiving or 2. Response to placebos
pending) 3. Remission with psychotherapy
504 V. KOUKOUNI AND K. P. BHATIA
et al., 2002). When there is strong evidence for psy- 1989). Also drug treatment can reduce the tremor
chogenic parkinsonism, sodium amytal can be used amplitude but it will not affect the frequency (Koller
alternatively, as it may ameliorate psychogenic tremor. et al., 1989). Zeuner et al. (2003), used accelerometry
Furthermore, spontaneous remissions and improve- to measure frequency changes during tapping in a
ment of symptoms with psychiatric consultation and group of psychogenic tremor patients and a group with
psychotherapy are strongly suggestive of their psy- parkinsonian and essential tremor patients. They con-
chogenic origin (Fahn, 1994). Admission to hospital cluded that a significant absolute change in tremor
can prove beneficial, as it allows continuous observa- frequency and marked variability in tapping were
tion of the patient, as well as psychiatric estimation noted in psychogenic tremor patients and suggested
and intervention. Investigations including imaging that the change in frequency is more characteristic
(computed tomography (CT) and magnetic resonance than entrainment in those patients.
imaging (MRI)) may be useful in order to exclude a It should be emphasized that patients with organic
structural lesion. Relevant blood tests should always PMDs can demonstrate additional psychogenic symp-
be performed in order to exclude possible organic con- toms, which usually affect the same part of the body as
ditions and also convince the patient that the physi- the underlying organic movement disorder (Ranawaya
cian pays the appropriate attention to the patients et al., 1990). This, along with the fact that psychogenic
complaints (Fahn, 1994; Sa et al., 2004). parkinsonism is mainly a diagnosis of exclusion, makes
As PD and other parkinsonian disorders have a pre- the diagnosis a challenging issue that should be estab-
synaptic dopaminergic defect, dopamine transporter lished with extreme caution, preferably by a movement
(DAT) single-photon emission computed tomography disorders specialist or an experienced neurologist, with
(SPECT) and fluorodopa (18F-dopa) positron emission the assistance of a psychiatrist.
tomography (PET) scans may be useful to differentiate
psychogenic parkinsonism from organic parkinsonian 58.6. Psychiatric diagnosis
conditions. It should be kept in mind that DAT SPECT
and 18F-dopa PET scans can be normal in dopa- It would be useful to define some of the conditions
responsive dystonia-parkinsonism, manganese and seen in patients with psychogenic disorders, according
neuroleptic-induced parkinsonism, akinetic-rigid Hun- to the American Psychiatric Associations (1995)
tingtons disease, X-linked parkinsonism and other Diagnostic and Statistical Manual of Mental Disor-
conditions (Lang et al., 1995). There are also a small ders (DSM-IV). In somatoform disorders, the symp-
number of PD patients in whom scans were found to toms suggest a general medical condition but they
be apparently normal (Whone et al., 2003). In Langs cannot be fully explained by the presence of a specific
series (Lang et al., 1995) 4 patients underwent 18F-dopa medical disorder, exposure to a substance or by any
PET; the scan was abnormal in only 1 patient and this other mental disorder and cause significant distress
raised the suspicion of an underlying organic PD. In or impairment in social, occupational or other areas
Morgans series (Morgan et al., 2004), DAT SPECT of functioning. The symptoms are not intentionally
scans were performed in 2 patients and they both produced or feigned. They include somatization and
proved to be normal. In any case, an abnormal result conversion disorders. Somatization disorders begin
on DAT or 18F-dopa PET would be in favor of organic before the age of 30 and occur over a period of many
parkinsonism (Tolosa et al., 2003). years, resulting in medical treatment or significant im-
Electrophysiological studies, including EMG, pairment in social, occupational or other areas of
accelerometry (Zeuner et al., 2003) and frequency ana- functioning. Conversion disorder is characterized by
lysis (Brown and Thompson, 2001), have been used in symptoms or deficits affecting voluntary motor or sen-
the literature for the establishment of the psychogenic sory function that suggest a neurological or other
origin of the tremor, but they do not have much use in medical condition and is usually preceded by conflicts
the day-to-day clinical service. Psychogenic tremor or other stressors. On the other hand, factitious disor-
has a frequency of less than 6 Hz, as hardly anyone ders, such as Munchausens syndrome, are intention-
can produce a voluntary tremor over 7 Hz (Elble and ally produced and the motivation is for the patient to
Koller, 1990). Large fluctuations in tremor amplitude assume the sick role. In malingering the symptoms
(e.g. with weighing) and frequency and also improve- are again intentionally produced and motivated by
ment of tremor with distractibility are noted during external incentives (e.g. avoiding work or military
the examination. It should, however, be kept in mind duty or obtaining compensation).
that patients with PD or essential tremor can demon- The establishment of the underlying psychiatric dis-
strate variations in tremor amplitude with stress or order in psychogenic parkinsonism and all other psy-
emotion, but these are only minimal (Koller et al., chogenic disorders, although crucial for diagnosis
PSYCHOGENIC PARKINSONISM 505
and treatment, is not always possible. In Langs series a persistence in abnormal movements in more than
(Lang et al., 1995) there was evidence of somatoform 90% of patients. In Langs series (Lang et al., 1995),
disorders (35.7%), especially conversion disorders 2 patients had spontaneous long remissions, 1 had
and depression (14.3%), in some patients and litigation complete but short remission and one had marked but
or compensation issues in a few others (21.4%), incomplete response to psychotherapy and haloperidol.
whereas the psychiatric disorder was not clearly iden- In Morgans series (Morgan et al., 2004), 5 patients
tified in 3 of them (21.4%). In other series, regarding attended follow-up; 2 of them had abrupt resolution
other PMDs, the most common psychiatric diagnoses of their symptoms, 1 was cured after intensive inpati-
were depression, anxiety and conversion and somatiza- ent psychotherapy and 2 remained convinced that they
tion disorders (Koller et al., 1989; Factor et al., 1995; had PD. Generally, in all PMDs, the prognosis depends
Kim et al., 2003). on the underlying psychiatric disorder, the acute or
insidious onset and the course of the disease, the pre-
58.7. Treatment morbid functioning and the presence of an identifiable
trigger. The prognosis is supposed to be better when
When the diagnosis of psychogenic parkinsonism is the onset of the disease is acute, with a short dura-
established, it should be presented to the patient tion of symptoms and when there is a specific emo-
with extreme caution and a non-judgmental character, tional event that preceded the onset of the disease of
to gain trust and acceptance. It should be pointed out a previously healthy individual (Koller et al., 2002).
that the parkinsonism is not due to a lesion or a Conversion disorders with recent onset are considered
severe dysfunction of the nervous system and that it to have better prognoses than factitious disorders
can be fully resolved with the appropriate approach. and malingering, which usually respond poorly and
A neuropsychiatric evaluation should be suggested, unpredictably (Miyasaki et al., 2003).
especially to those who had a long-standing disease
References
with multiple investigations and ineffective treatments
(Sa et al., 2004). It is important that the patient is
American Psychiatric Association (1995). Diagnostic and
approached in a way that does not make him/her feel
Statistical Manual of Mental Disorders, 4th edn. (DSM-
that he/she is crazy. It was suggested by Ford et al. IV). International Version with ICD-10 Codes, Washing-
(1995) that a possible neurobiological explanation for ton DC, pp. 457487.
the patients symptoms could be presented. It should Brown P, Thompson PD (2001). Electrophysiological aids to
perhaps be explained that the mind and the body are the diagnosis of psychogenic jerks, spasms, and tremor.
in strong association with each other and that dysfunc- Mov Disord 16 (4): 595599.
tion of the one can affect the other, as happens with Campbell J (1979). The shortest paper. Neurology 29: 1633.
numerous other diseases (Fahn, 1994). A psychiatrist Crimlisk HL, Bhatia KP, Cope H et al. (1998). Slater revis-
should be involved, preferably early in the course ited: 6 year follow-up study of patients with medically
of the investigation, providing psychotherapy and unexplained motor symptoms. BMJ 316: 582586.
Deuschl G, Koster B, Lucking CH et al. (1998). Diagnostic
positive reinforcement. Cognitive-behavioral therapy
and pathophysiological aspects of psychogenic tremors.
should also be considered. As discussed earlier, the
Mov Disord 13: 294302.
use of placebo is controversial and should only be used Elble RJ, Koller WC (1990). Unusual forms of tremor. In:
to establish the diagnosis (Fahn, 1994). Alternatively, RJ Elble, WC Koller (Eds.), Tremor. The Johns Hopkins
mild antidepressants and anxiolytics can be used to University Press, Baltimore, pp. 154157.
manage the underlying depression and anxiety, along Factor SA, Podskalny GD, Molho ES (1995). Psychogenic
with a periodical neurological assessment (Koller movement disorders: frequency, clinical profile and char-
et al., 2002). acteristics. J Neurol Neurosurg Psychiatry 59: 406412.
Fahn S (1994). Psychogenic movement disorders. In: CD
58.8. Prognosis Marsden, S Fahn (Eds.), Movement Disorders.3rd edn. -
Buttenw Heinem, Oxford, pp. 359372.
Fahn S, Williams PJ (1988). Psychogenic dystonia. Adv
The prognosis for functional recovery is variable and
Neurol 50: 431455.
relatively poor. In a follow-up study of patients with
Feinstein A, Stergiopoulos V, Fine J et al. (2001). Psychiatric
medically unexplained movement disorders, the pre- outcome in patients with a psychogenic movement disor-
senting symptom remained unchanged in 14% of der: A prospective study. Neuropsychiatry Neuropsychol
patients, although in 38% of them it had worsened Behav Neurol 14 (3): 169176.
(Crimlisk et al., 1998). A prospective study done by Ford B, Williams DT, Fahn S (1995). Treatment of psychogenic
Feinstein et al. (2001) regarding the psychiatric out- movement disorders. In: R Kurlan, (Ed.). Treatment of Move-
come in patients with PMDs revealed that there was ment Disorders. JP Lippincott, Philadelphia, pp. 475485.
506 V. KOUKOUNI AND K. P. BHATIA

Kim YJ, Pakiam ASI, Lang AE (1999). Historical and clini- Ranawaya R, Riley D, Lang A (1990). Psychogenic dyskine-
cal features of psychogenic tremor: review of 70 cases. sias in patients with organic movement disorders. Mov
Can J Neurol Sci 26: 190195. Disord 5: 127133.
Koller WC, Biary NM (1989). Volitional control of involun- Sa DS, Galvez-Jimenez N, Lang AE (2004). Psychogenic
tary movements. Mov Disord 4 (2): 153156. movement disorders. In: R Watts, WC Koller (Eds.),
Koller WC, Lang AE, Vetre-Overfield B et al. (1989). Psycho- Movement Disorders, Neurologic Principles and Prac-
genic tremors. Neurology 39: 10941099. tice, 2nd edn. McGraw Hill, New York, pp. 891914.
Koller WC, Marjama-Lyons J, Troster AJ (2002). Psychogenic Tolosa E, Coehlo M, Gallardo M (2003). DAT Imaging in
movement disorders. In: JJ Jankovic, E Tolosa (Eds.), drug-induced and psychogenic parkinsonism. Mov Disord
Parkinsons Disease and Movement Disorders. Lippincott, 18 (Suppl 7): S28S33.
Philadelphia, pp. 546552. Whone AL, Watts RL, Stoessl AJ et al. (2003), REAL-PET
Lang AE, Koller WC, Fahn S (1995). Psychogenic parkin- Study Group. Slower progression of Parkinsons disease
sonism. Arch Neurol 52: 802810. with ropinirole versus levodopa: The REAL-PET study.
Miyasaki JI, Sa DS, Galvez-Jimenez N et al. (2003). Psychogenic Ann Neurol 54: 93101.
movement disorders. Can J Neurol Sci 30 (Suppl 1): S94S100. Walters AS, Boudwin J, Wright D et al. (1988). Three hys-
Monday K, Jankovic J (1993). Psychogenic myoclonus. Neu- terical movement disorders. Psychol Rep 62: 979985.
rology 43: 349352. Zeuner KE, Shoge RO, Goldstein SR et al. (2003). Accelerome-
Morgan JC, Mir P, Mahapatra RK et al. (2004). Psychogenic try to distinguish psychogenic from essential or parkinsonian
parkinsonism: clinical features of a large case series. Mov tremor. Neurology 61: 584550.
Disord 19 (Suppl 9): S345S346.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 59

Parkinsonism and dystonia

RUTH H. WALKER*

Movement Disorders Clinic, Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY
and Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA

59.1. Introduction in patients with generalized dystonia from a variety of


causes, symptoms often develop asymmetrically and
59.1.1. The dopamine connection
remain worse on one side throughout the course of the
disease. A number of the pediatric metabolic genetic
The commonalities and distinctions between parkin-
disorders cause specifically orofacial dyskinesias.
sonism and dystonia have long intrigued clinicians
Some authors have described the movements seen in
and researchers in the field of movement disorders.
dystonia as bradykinetic, but the slowness of actions in
The clinical features of both can be seen in conditions
dystonia is of a different quality to that seen in PD, being
in which dopaminergic neurotransmission is impaired;
due to an increase in muscle activation in a pattern which
however, dystonia can also be seen in a wide variety of
counteracts the intended movement, hypothesized to be a
other conditions, in which the pathophysiology is far
consequence of decreased inhibition of unwanted motor
from clear. Dysfunction of the dopaminergic system
activity (Mink, 1996, 2003; Perlmutter et al., 1997; Kaji,
in parkinsonism has been known since the 1960s; in
2001). This is in contrast to difficulty in initiating move-
dystonia this has been inferred from empirical evi-
ment (akinesia), due to inadequate and discoordinated
dence for many years and is slowly becoming clarified
muscle activity and prolongation of movement time,
from scientific studies.
superimposed upon muscle rigidity, as seen in Parkinsons
disease (PD) (Hallett, 2003).
59.1.2. Phenomenology of dystonia

Phenomenologically, dystonia is classified as a hyper- 59.1.3. Classification of dystonia


kinetic movement disorder, in which involuntary mus-
cle contractions result in abnormal postures and The classification of dystonia is in a state of flux, as var-
movements (Fahn et al., 1998). An imbalance of flexor ious genes become identified as causes of what used to be
and extensor activity may cause hyperextension or called idiopathic dystonia. The term primary is now
flexion at a joint, which is often, although not invari- used for cases in which dystonia is the major, and often
ably, dynamic. In some cases, alternation of flexion the sole, symptom. These are typically due to genetic
and extension results in tremor, rather than abnormal causes and other etiologies, such as a structural lesion,
posture, which may confuse the diagnosis. In other must be excluded by clinical, radiological and laboratory
cases, particularly those due to secondary causes (e.g. evaluations (Fahn et al., 1998). In addition, there must be
structural brain lesions), the abnormal posture is fixed no response to levodopa, excluding dopa-reponsive dys-
and does not resolve during relaxation or even sleep. tonia (DRD) (Fahn et al., 1998; Bressman, 2004).
Dystonia can affect any part of the body and often Secondary dystonias are those in which dystonia is due
has a characteristic distribution in different diseases. to another disease process (Fahn et al., 1998; Bressman,
Axial involvement may be present in combination with 2004). This may be one of the inherited dystonia-plus syn-
axial parkinsonism. A strictly unilateral presentation dromes, in which dystonia is part of a symptom complex,
may indicate a contralateral structural lesion; however, such as myoclonusdystonia or DRD; acquired, such as a

*Correspondence to: Ruth H. Walker, Department of Neurology, James J. Peters Veterans Affairs Medical Center (127), 130 W.
Kingsbridge Road, Bronx NY 10468, USA. E-mail: ruth.walker@mssm.edu, Tel: 1-718-584-9000-x5915, Fax: 1-718-741-4708.
508 R. H. WALKER
structural lesion, for example stroke, tumor or head trauma 59.1.4. Pathophysiology
or drug-induced; or heredodegenerative, in which dystonia
may be a manifestation of an inherited disease such as According to the commonly accepted model of the
Huntingtons disease (HD), spinocerebellar ataxia (SCA) basal ganglia (Albin et al., 1989; DeLong, 1990), the
type III, Lubag (Filipino X-linked dystoniaparkinsonism) direct and indirect pathways from the striatum have
or chromosome 17 frontotemporal dementia and parkin- opposing effects upon the activity of the subthalamic
sonism. In addition, secondary dystonia may be present nucleus (STN) and its projection targets, the internal
in the apparently sporadic (and rarely inherited) parkinso- segment of the globus pallidus (GPi) and the substantia
nian syndromes, including idiopathic PD, multiple system nigra pars reticulata (SNpr) (Fig. 59.1A). These nuclei
atrophy (MSA), corticobasal degeneration (CBD) and pro- are viewed as the main source of output of the basal
gressive supranuclear palsy (PSP). ganglia and send inhibitory projections to a number
In this review, the disorders have been classified as of targets, including the thalamus centromedian pars
parkinsonian disorders with dystonia, dystonic dis- fascicularis (CM-pf), the pedunculopontine nucleus
orders with parkinsonism and mixed movement dis- (PPN) and the substantia nigra pars compacta (SNpc).
orders, in which the phenomenology may vary and The projection from the striatum to the GPi the direct
there may be variety of neurologic features, including pathway is GABAergic and inhibits the activity of
dystonia and parkinsonism. GPi neurons. The projection from the striatum to the
PARKINSONISM AND DYSTONIA 509

Fig. 59.1. For full color figure, see plate section. (A) During normal function of the basal ganglia, the direct pathway from the
striatum inhibits neurons of the internal segment of the globus pallidus (GPi), disinhibiting the motor pattern generator, con-
sisting of the motor thalamic nuclei and their projections to the cortex. The neurons which select the motor program are repre-
sented as being surrounded by a network, controlled by the indirect pathway, which reduces the generation of unwanted
movements. (B) In Parkinsons disease, with loss of the dopaminergic input to the striatum, the direct pathway is less active,
with increased activity of the inhibitory neurons of the GPi, which further inhibit the motor pattern generators. The indirect
pathway is disinhibited by the loss of dopamine, resulting in a decreased inhibition of the subthalamic nucleus (STN) and
the GPi. The surround-inhibitory neurons of the GPi also have increased activity and unselected movements are also
decreased. Dashed lines, decreased activity; thicker lines, increased activity. (C) In dystonia, there is increased activity of
the direct pathway, with decreased inhibition of the thalamocortical motor pattern generators. There is decreased surround inhi-
bition via the indirect pathway, with loss of inhibition of unselected movements. GABA, gamma-aminobutyric acid;
SNcSNpc, substantia nigra pars compacta; GPe, globus pallidus pars externa. Adapted from Mink (2003) with permission.
Copyright # 2003, American Medical Association. All rights reserved.

globus pallidus pars externa (GPe), the indirect path- Alterations in regional blood flow in various brain
way, is also GABAergic and an increase in activity areas reflect changes in neuronal activity in PD and
of this pathway inhibits the GABAergic GPe and disin- correlate to some extent with the above model (Eidel-
hibits the STN. The STN sends an excitatory projection berg et al., 1990; Jenkins et al., 1992; Playford et al.,
to the GPi and SNpr, thus the neuronal activity of these 1992). A correspondence is seen with disease severity
structures usually changes in parallel. (Eidelberg et al., 1995) and these alterations can be
In PD a decrease in the dopaminergic input to the normalized by various therapeutic interventions (Jen-
striatum results in decreased activation of the direct kins et al., 1992; Playford et al., 1992; Jahanshahi
pathway by dopamine D1-receptors and decreased et al., 1995; Eidelberg et al., 1996; Fukuda et al.,
inhibition of the GPi. The indirect pathway is inhibited 2001). However, these metabolic studies represent
by dopamine D2-receptors, thus dopamine depletion levels of synaptic activity and do not distinguish
results in increased activity of the inhibitory striato- between excitatory or inhibitory neurons, or afferents
GPe pathway. These neurons inhibit the GPe, which or interneurons, so do not necessarily help us under-
reduces its inhibition of the glutamatergic STN neu- stand the precise mechanism of dysfunction in PD at
rons (Bergman et al., 1994) (Fig. 59.1B). The STN a neuronal level.
shows an increase in activity, resulting in increased sti- In another model which has been proposed, based
mulation by STN neurons of the internal segment of upon a center-surround pattern, the direct pathway
the GPi and the SNpr (Carpenter et al., 1981; Kitai serves to activate a motor program and the indirect path-
and Kita, 1987) and increased activity in the projec- way focuses and selects movements by inhibiting
tions from these structures. This model has been borne unwanted motor programs (Mink, 1996, 2003; Perlmutter
out by data from a variety of animal models of dopa- et al., 1997; Kaji, 2001). Thus, in PD, there is less acti-
mine depletion and intraoperative recordings in PD vation of motor programs via the direct pathway and
(Wichmann et al., 1994; Eidelberg et al., 1997; Rodriguez excessive focusing via the indirect pathway, resulting in
et al., 1998; Vitek et al., 1998). inadequate movement (Fig. 59.1B).
510 R. H. WALKER
In dystonia, the converse may be proposed: increased hyperactivity as the etiology of dystonia (Goto et al.,
D1-mediated stimulation of the direct pathway and 2005) (discussed in more detail on p. 514).
increased D2-mediated inhibition of the indirect pathway Other studies suggest changes in the sensorimotor
to the GPe, with disinhibition of the GPe, an increase in cortex in dystonia. For example, an increase in size
the inhibitory GPe-subthalamic pathway, with resultant in receptive fields of cortical sensory neurons and a
decreased STN activity and thus decreased GPi neuronal loss of specificity of cortical motor topography has been
activity (Mink, 1996, 2003; Berardelli et al., 1998; Kaji, observed in dystonia (Byl et al., 1996; Lenz et al., 1999;
2001) (Fig. 59.1C). According to the model, this would Bara-Jimenez et al., 2000; Quartarone et al., 2003).
cause increased activation and loss of selectivity of motor Decreased cortical inhibition in focal hand dystonia
pattern generators and decreased inhibitory surround (Ridding et al., 1995; Ikoma et al., 1996; Sohn and Hal-
(Mink, 1996, 2003), resulting in the excessive muscle lett, 2004), likely due to a reduction in cortical gamma-
contractions and the overflow of movements which we aminobutyric acid (GABA) (Levy and Hallett, 2002)
see clinically. suggests that decreased focusing is present in the sen-
Dysfunction of the direct pathway in dystonia is sorimotor cortex, either as a primary or secondary phe-
suggested by an increase in the striatal-GPi pathway nomenon. There is evidence for aberrant neuronal
activity seen on functional imaging (Eidelberg et al., plasticity in dystonia in experimental situations (Byl
1995). This would result in a decrease in GPi activity et al., 1996) and in clinical practice, when focal dystonia
and disinhibition of the thalamus. develops as the consequence of excessive performance
There are data supporting dysfunction of the indir- of the same task, as is seen in musicians and other
ect pathway; however these are harder to interpret task-specific dystonia. The presence of a sensory trick
and to translate to the model. Whatever the mechan- or geste antagoniste, when the patient can correct
ism, the changes must ultimately be explicable in the movement with a light touch, certainly suggests
terms of loss of a component of the basal ganglia, as involvement of sensory circuits.
dystonia can be seen in a number of neurodegenerative When dystonia is due to a structural lesion of the
disorders, in addition to those cases in which there is putamen, the model would predict that neurons of
no evidence of neurodegeneration, for example in the indirect pathway were preferentially damaged, as
DYT1 dystonia or when is a medication side-effect. decreased function of the first connection of the indir-
There is evidence of decreased dopamine D2-receptor ect pathway is postulated (as opposed to increased
binding (Perlmutter et al., 1998). However, decreased activity in the striato-GPi direct pathway). There is
D2-receptor binding was also found in non-manifesting as yet no neuropathological evidence to support this,
DYT1 mutation carriers (Asanuma et al., 2005). One thus we do not know whether the model fits these clin-
explanation was that this was to a lesser extent than in ical situations. In addition, the model does not explain
symptomatic carriers. Alternatively, additional factors why pallidotomy should reduce the symptoms of dys-
may be required for the motor symptoms to manifest tonia. However, intraoperative recordings from small
clinically. Other functional imaging (Eidelberg, 1998; numbers of patients with dystonia undergoing surgery
Meunier et al., 2001) and neuropsychological studies suggest that neuronal firing patterns show abnormal
(Heiman et al., 2004) of non-manifesting DYT1 carriers bursting patterns rather than an absolute reduction in
also suggested subtle changes in brain networks which rate (Vitek et al., 1999; Silberstein et al., 2003) and
do not manifest as a movement disorder. that it is the information thus encoded which causes
In contrast to DYT1 dystonia, in DRD an increase the loss of movement selectivity.
in dopamine D2-receptors was found, with unchanged The model proposes different mechanisms for the
D1 and dopamine transporter labeling (Rinne et al., different clinical manifestations of basal ganglia dys-
2004). The model (Figure 59.1C) predicts decreased function. The presence of both simultaneously due to
activity of the striato-GPe connection of the indirect a single disease process suggests a topographical var-
pathway in dystonia, which could be as a consequence iation in signal processing, possibly within a single
of inhibition from increased postsynaptic dopamine structure of the basal ganglia.
D2 stimulation. However, it is not known whether
the tracer imaging results demonstrate a primary or 59.1.5. The function of dopamine in the striatum
compensatory change and whether the D2-receptors
are pre- or postsynaptic. Additionally the DRD The precise function of dopamine within the striatum at
patients were being treated with levodopa, making it the synaptic level still remains to be fully elucidated.
difficult to interpret the data and fit them to the model. The relationship between dopamine and glutamate
Recent work upon the neuropathology of Lubag release from corticostriatal afferents is complex and inter-
(DYT3) suggests a possible mechanism for dopaminergic dependent. Glutamatergic corticostriatal axons synapse
PARKINSONISM AND DYSTONIA 511
on the heads of spines of striatal medium spiny neurons, dopamine levels were decreased and turnover increased.
with nigrostriatal dopaminergic terminals synapsing on Clearly further study is necessary to determine the
the spine shafts, thus the release of dopamine is ideally precise nature of dopaminergic dysfunction in dystonia.
positioned anatomically to modulate the effect of inputs
from sensorimotor cortex (Smith et al., 1994).
59.2. Parkinsonian disorders with dystonia
The effect of dopamine upon the postsynaptic med-
(Table 59.1)
ium spiny neurons which bear dopamine receptors
depends upon a number of variables. These include dopa- 59.2.1. Parkinsons disease (sporadic)
mine receptor subtype (Calabresi et al., 1987, 1997),
recent activation by corticostriatal glutamate release in Dystonia may be a feature of PD in a variety of cir-
a single spike or burst (long-term potentiation or long- cumstances. A frequent early manifestation of PD is
term depression (Calabresi et al., 1996; Arbuthnott with limb, typically leg, dystonia. This may persist as
et al., 2000)), hyperpolarization by GABAergic inputs a feature during off periods, particularly during the
(from globus pallidus or local axon collaterals) and night and early morning, when patients are not medi-
probably other factors as yet undetermined. cated for an extended period. Such dystonia is distinct
Release of dopamine itself is also modified by a from the dystonia seen during on periods, in that it is
number of factors, including recent neuronal activity fixed and painful. Characteristically this is described
(Cragg, 2003), corticostriatal afferents (Avshalumov as a painful cramp of the calf muscles and foot, with
et al., 2003), inputs from cholinergic interneurons the foot and toes plantar-flexed. It is possible that the
(Partridge et al., 2002; Rice and Cragg, 2004), nitric foot region of the putamen is particularly vulnerable
oxide synthase-containing interneurons (Saka et al., to disruptions of dopaminergic function. In addition
2002) and GABAergic afferents (Juranyi et al., 2003). to the foot dystonia seen in early PD, both child-
In addition, dopamine modulates glutamate release hood-onset DYT1 dystonia (Bressman et al., 1998)
from corticostriatal terminals via dopamine D2-recep- and DRD (Segawa and Nomura, 1993) typically pre-
tors (Bamford et al., 2004a, b). Evidence from these sent with foot dystonia, before spreading more ros-
studies suggests that dopamine facilitates more active, trally. The body is represented topographically in the
but inhibits less active, corticostriatal inputs (Bamford putamen, with the foot region being located dorsally
et al., 2004b). Dopamine also facilitates the effects of (Gerardin et al., 2003); therefore, there may be an ana-
glutamatergic inputs upon medium spiny neurons, via tomic reason for the location of this symptom.
postsynaptic D1-receptors (Flores-Hernandez et al., Truncal dystonia, resulting in forward and/or lateral
2002). In combination, these mechanisms serve to flexion of the spine (camptocormia) is seen in
reinforce selection of more active signals and to filter advanced PD (Djaldetti et al., 1999) and may respond
out weaker ones. This may be represented in the cir- to dopaminergic agents or deep brain stimulation
cuit diagram (Fig. 59.1) by the center-surround (DBS), but often becomes fixed. This can be a
model being present in the striatum as well as further severely disabling feature of the disease. Dystonia of
downstream in the globus pallidus. other regions has also been reported in PD, including
When dopamine is decreased, as in PD, there may cervical, cranial and upper limb (Katchen and Duvoi-
be a loss of facilitation of corticostriatal signals from sin, 1986; LeWitt et al., 1986; Poewe et al., 1988;
the motor cortex, with resultant difficulty in initiation Morrison and Patterson, 1992).
and slowness of movement. In dystonia, there appears Dystonia is often a component of levodopa-induced
to be an excess of movement, often following the dyskinesias. Levodopa-induced dyskinesias can be
initiation of a specific action. This may be the conse- seen as blood levels of medication are increasing or
quence of a loss of signal selection, with unwanted corti- wearing off (onoff dyskinesias) or at the peak dose of
costriatal signals being transmitted as discussed above. medication. Peak-dose dyskinesias involve the arms,
There is evidence from functional imaging studies of trunk and neck and are usually choreiform but may
alterations in various indicators of striatal dopaminergic incorporate more sustained involuntary muscle contrac-
neurotransmission in DYT1 dystonia (Playford et al., tions, either alone or in combination with rapid, flowing
1993; Perlmutter et al., 1998) and DRD (Rinne et al., movements. Onoff dyskinesias tend to involve the
2004); however, these results should be interpreted with legs and often have a dystonic component. The dyskine-
caution. Preliminary evidence from transgenic mice car- sias which occur as levodopa or dopamine levels are
rying a mutation of torsinA, the cause of DYT1 dystonia changing are hypothesized to be the result of an
(Shashidharan et al., 2002, 2005; Bao et al., 2006) and imbalance of dopamine transmission between different
from a small number of humans with DYT1 dystonia pathways or possibly between adjacent striatal micro-
(Augood et al., 2002; Rostasy et al., 2003), suggested that circuits. Therapies which smooth out serum/brain
512 R. H. WALKER
Table 59.1
Parkinsonian disorders in which dystonia may occur

Other
neurological Other
Disease Inheritance Dystonia Parkinsonism features Genetic tests useful tests

Parkinsons disease Sporadic, , off, Subcortical Parkin, DJ-1, Response to


AR, AD peak- dementia PINK1, levodopa,
dose a-synuclein, dopamine
LRRK2 transporter
imaging
Postencephalitic Sporadic Dementia
parkinsonism
Progressive Sporadic Dementia,
supranuclear vertical gaze
palsy palsy
Corticobasal Sporadic Focal cortical
degeneration signs, alien
limb
Multiple system Sporadic Autonomic, Fe in putamen on
atrophy cerebellar MRI
Primary pallidal Sporadic Chorea
degeneration
Frontotemporal AD, Dementia tau
dementia and sporadic
parkinsonism
Neuronal Sporadic Dementia,
intermediate corticospinal
filament disease

AD, autosomal dominant; AR, autosomal recessive; absent; mild; moderate; marked; MRI, magnetic resonance imaging.

levodopa/dopaminergic levels are usually beneficial, of onset rather than the presence of the causative muta-
such as catechol-O-methyltransferase inhibitors or the tion (Quinn et al., 1987; Khan et al., 2003; Lohmann
use of longer-acting dopaminergic agents. DBS of the et al., 2003; Tan et al., 2003b). The response to levo-
STN is also helpful as it enables less dopaminergic dopa is usually striking and this feature, in combina-
medication to be used. tion with prominent dystonia, may even suggest a
diagnosis of DRD (Tassin et al., 2000).
59.2.2. Inherited Parkinsons disease Autosomal-dominant inheritance, for example of a-
synuclein (Polymeropoulos et al., 1997) or LRRK2
There is debate as to whether disease due to some of (Zimprich et al., 2004) mutations, tends to result in more
the causative mutations identified to date, such as of typical disease. Autosomal-recessive inheritance of
parkin and PARK8, can truly be called PD, as the mutations of other genes causing early-onset PD, DJ-1
pathologic hallmark, the Lewy body, is often (but not (Bonifati et al., 2003) and PINK1 (Valente et al., 2004),
invariably) absent in these cases (Singleton, 2004). do not appear to be specifically characterized by dysto-
However, the clinical criteria in terms of symptom- nia, although the rarity of these mutations as a cause of
atology, disease progression and response to levodopa either sporadic or familial early-onset PD (Clark et al.,
are otherwise identical to idiopathic PD, so for the 2004), means that a typical phenotype is not yet fully
purpose of classification they are included here. characterized.
Mutation of the parkin gene typically results in dis-
ease onset in the 20s and 30s and may be the result of 59.2.3. Postencephalitic parkinsonism
either heterozygosity or homozygosity for mutant
alleles. Dystonia, sometimes exercise-induced (Khan Postencephalitic parkinsonism was the consequence of
et al., 2003), is often a presenting feature in these von Economos encephalitis, which affected many
patients, but appears to be more related to young age people in the earlier part of the 20th century. Although
PARKINSONISM AND DYSTONIA 513
this pandemic was temporally coincident with the comprises the syndrome. Typically, the parkinsonism is
influenza pandemic, it has not been confirmed that midline with marked axial rigidity and falling and cervi-
encephalitis lethargica was due to the influenza virus. cal antecollis is often seen (Boesch et al., 2002). Limb
This condition is often understood to be a hypoki- dystonia may also be present (Boesch et al., 2002).
netic-rigid syndrome; however, dystonia and other
hyperkinetic movements were quite prominent (Evi- 59.2.7. Frontotemporal dementia and
dente et al., 1998; Albanese, 2003). Dystonia usually parkinsonism
appeared to be cranial segmental in distribution,
although the limbs were also involved (Morrison and This disease can be seen in an autosomal-dominant
Patterson, 1992; Evidente et al., 1998). familial form, associated with heterozygous mutation
Contemporary encephalitides may occasionally of the gene for tau protein on chromosome 17 (Hutton
cause this syndrome. For example, parkinsonism and et al., 1998), or may be sporadic. Neuropathologically,
dystonia have been reported following Japanese ence- mutations of tau result in typical deposits of this protein
phalitis (Murgod et al., 2001). (Taniguchi et al., 2004). In cases due to tau mutations,
parkinsonism, rather than frontotemporal dementia,
59.2.4. Progressive supranuclear palsy may be associated with a particular genetic haplotype
(Walker et al., 2002a) and dystonia, particularly cranial,
The dystonias seen in this parkinsonian neurodegen- may be present (Walker et al., 2002a).
erative condition are strikingly different from those
seen in PD. Blepharospasm is a frequent feature and 59.2.8. Primary pallidal degeneration
may be functionally very limiting, but responds well
to botulinum toxin injection. Extension of the trunk Rarely, cases of bradykinesia and dystonia have been
and neck is characteristic, with retropulsion, unlike in reported in which neuropathological changes were
PD, where there tends to be forward flexion. Although limited to the globus pallidus (Aizawa et al., 1991).
usually associated with a diagnosis of CBD, limb The onset was in adulthood and the disease was dis-
dystonia has been reported in neuropathologically tinct from PD in that there was no increase in muscle
confirmed PSP (Barclay and Lang, 1997; Oide et al., tone. Onset at younger age has also been reported,
2002). Diagnostic pathological changes are neuro- associated with marked rigidity and more choreiform
fibrillary tangles in the basal ganglia and midbrain. and dystonic movements.
Dystonic dyskinesias are rare as a side-effect of
levodopa therapy in this condition, but may occur 59.2.9. Neuronal intermediate filament inclusion
(Barclay and Lang, 1997; Tan et al., 2003a). Func- disease
tional benefits from dopaminergic medications in
PSP and the following conditions are usually minimal. This rare sporadic disorder may present with a variety of
neurologic features, including frontotemporal dementia,
59.2.5. Corticobasal degeneration behavioral change, corticospinal signs and parkinsonism
in young adulthood (Cairns et al., 2004). Limb dystonia
CBD is characterized by an asymmetric presentation of may also be seen, along with supranuclear palsy and buc-
parkinsonism, dystonia and cortical deficits, due to cal apraxia. The diagnosis is made neuropathologically
pathology of the basal ganglia and cortex. Limb dystonia with the finding of characteristic neuronal inclusions
may result in flexion contractures of the hand and arm. which are immunoreactive for phosphorylated intermedi-
The leg tends to be extended at the knee and the foot ate filament (Cairns et al., 2004).
plantar-flexed and inverted. The alien-limb phenom-
enon may be seen, in which the patients hand performs 59.3. Dystonic disorders with parkinsonism
involuntary semipurposeful movements and is presumed
to be due to involvement of cortical motor areas. Only dystonic conditions with parkinsonian features are
described in detail here. These and the other genetically
59.2.6. Multiple system atrophy defined dystonias are summarized in Table 59.2

The parkinsonism of MSA is due to degeneration of the 59.3.1. Lubag (DYT3)


GABAergic medium spiny striatonigral neurons, which
receive inputs from the nigrostriatal dopaminergic X-linked dystonia-parkinsonism is found solely amon-
neurons. In addition, degeneration of either cerebellar gst Filipinos from the province of Capiz on the island
outputs or neurons of the autonomic nervous system of Panay (Lee et al., 1976). Although mostly males are
514 R. H. WALKER
Table 59.2
The genetically defined dystonias with genes and gene products (where known)

Name Inheritance Gene Location Protein Parkinsonism

DYT1 Primary torsion dystonia AD DYT1 9q34 torsinA


DYT2 Generalized early onset AR ? ? ?
DYT3 Lubag X-linked DYT3 Xq31.1 Multiple transcript
system
DYT4 Whispering dysphonia AD ? ATP7B ? 13q14.3 ? Copper-transporting
ATPase 2
DYT5 Dopa-responsive AD GCH-1 14q22.1 GTP cyclohydrolase-1
dystonia AR TH 11p15.5 Tyrosine hydroxylase
DYT6 Adolescent-onset mixed AD 8p ?
DYT7 Adult cervical and AD 18p ?
upper-limb dystonia
DYT8 Paroxysmal non- AD 2q23 ?
kinesogenic
dyskinesia
DYT9 Episodic AD 1p21 ?
choreoathetosis/ataxia
spasticity
DYT10 Paroxysmal kinesogenic AD 16p Sodium channels?
choreoathetosis/
dystonia
DYT11 Myoclonus-dystonia AD SGCE 7q21 E-sarcoglycan
DYT12 Rapid-onset dystonia- AD ATP1A3 19q13 Na/K-ATPase
parkinsonism a3 subunit
DYT13 Cranal, cervical and AD 1p36.13 ?
upper-limb dystonia 32
DYT14 Dopa-responsive AD 14q13 ?
dystonia
DYT15 Myoclonus dystonia AD 18p.11 ? 

AD, autosomal dominant; AR, autosomal recessive; absent; mild; moderate; marked; ATPase, adenosine triphosphatase; GTP,
guanosine triphosphate.

affected, symptomatic carrier females have occa- An elegant study comparing the neuropathology
sionally been reported, but tend to have a milder of parkinsonian cases with dystonic cases found, in dys-
phenotype (Waters et al., 1993b; Evidente et al., tonic cases, predominant loss of striatal projection neu-
2004). Onset is from the teens to the mid-40s, with rons in striosomes and patchy loss in the surrounding
progression over 57 years and then stabilization. matrix (Goto et al., 2005). This work supports the
Presentation is often with focal dystonia (lubag three-pathway model of basal ganglia function, which
describes the twisting movements and wa-eg, the emphasizes the relative roles of striosomes and matrix
abnormal sustained postures, in the local Illongo dia- (Graybiel et al., 2000). According to this model, loss of
lect) and may vary widely (Evidente et al., 2002). the inhibitory striosomal projection to the SNpc results
Parkinsonism (sud-sud, named for the shuffling gait) in increased dopaminergic activity, thus causing
may be an early or presenting feature and most increased activity of the neurons of the direct pathway
patients have mixed features. There is minimal and hence the decreased activity of the GPi, in ac-
response to levodopa and antidystonic agents (Evi- cordance with the traditional model (Fig. 59.1C). As the
dente et al., 2002). A benefit has been reported with disease progresses, striatal neuronal loss becomes more
the GABA-A receptor agonist zolpidem (Evidente, widespread and loss of the striato-GPi neurons, which
2002). were previously hyperactive, results in parkinsonism.
The causative gene has been identified as coding Other previous reports have also shown mosaic
for a multiple transcript system whose function is not degeneration of the striatum in Lubag (Waters et al.,
yet known (Nolte et al., 2003). 1993a; Singleton et al., 2004).
PARKINSONISM AND DYSTONIA 515
This diagnosis should be considered in any Filipino, instability. From a single case report there are no
even women, presenting with any movement disorder neuropathological abnormalities (Pittock et al., 2000).
and a detailed family history should be taken so that
appropriate counseling can be given (Evidente et al., 59.4. Mixed movement disorders
2002).
An Italian family has been reported with parkinson- 59.4.1. Autosomal-dominant (Table 59.3)
ism and dystonia affecting males in an apparently X- 59.4.1.1. Huntingtons disease
linked manner (Fabbrini et al., 2005). Linkage to the
The juvenile form of this genetic disorder, with age of
DYT3 locus was demonstrated, but no disease-causing
onset younger than 20 years, known as the Westphal
mutations were detected in the gene.
variant, is characterized by a parkinsonian presenta-
tion, often with marked dystonia. These tend to result
59.3.2. Dopa-responsive dystonia (Segawa disease) from paternal inheritance, when there is greater
(DYT5, DYT14) instability of the trinucleotide repeat expansion.
In adult-onset HD, as the disease advances, chorea
DRD is due to a variety of mutations of the gene for gua- often gives way to more parkinsonian and dystonic
nosine triphosphate cyclohydrolase-1 (GTP-CH1) (Ichi- features, with severe bradykinesia or akinesia, rigidity
nose et al., 1999) or for tyrosine hydoxylase (TH). and postural impairment (Penney et al., 1990; Kremer
GTP-CH1 is a key enzyme involved in the synthesis of et al., 1992). Rarely, HD may even present as levo-
tetrahydrobiopterin, which is a necessary cofactor for dopa-responsive parkinsonism with very mild chorea
the synthesis of TH. DRD is typically autosomal-domi- (Reuter et al., 2000). The juvenile form may also
nant with incomplete penetrance, affecting females more respond to both levodopa (Low and Allsop, 1973) and
than males (Furukawa et al., 1998). Autosomal-recessive dopamine agonists (Bonelli et al., 2002), suggesting that
inheritance of mutations of the gene for TH may also be the deficit is of presynaptic nigral dopaminergic neu-
responsible for a very similar disease (Hoffmann et al., rons. Although degeneration of striatal medium spiny
2003). Another locus responsible for autosomal-domi- neurons is well documented as the primary neuropatho-
nant DRD has been identified on chromosome 14, logic abnormality, loss of neurons from the SN, both
although the gene product is not yet known (Nygaard pars compacta and pars reticulata, has also been
et al., 1993). Onset is in childhood, with mixed features described (Bugiani et al., 1984; Oyanagi et al., 1989).
of parkinsonism and lower-limb dystonia. Very young One clue to the diagnosis of HD is, of course, a
onset with severe spasticity may be mistaken for cerebral positive family history, although in consideration of
palsy (Nygaard et al., 1994). This disorder is characteris- late-onset disease or non-paternity, a negative pedigree
tically exquisitely responsive to low or moderate doses of should not pre-empt genetic testing.
levodopa and even young children who are misdiagnosed
for a number of years may recover significant function. 59.4.1.2. Huntingtons disease-like 2
Neuropathologically, there is decreased immunoreac-
Huntingtons disease-like 2 (HDL2) (Margolis et al.,
tivity for TH in the SNpc, but no signs of neuronal loss or
2001) is a trinucleotide repeat expansion disease
Lewy bodies (Ichinose et al., 1999; Grotzsch et al.,
(Holmes et al., 2001), which bears a striking resem-
2002).
blance to HD in many clinical, genetic and neuro-
pathological aspects. The genetic mutation associated
59.3.3. Rapid-onset dystonia-parkinsonism with HDL2 has been characterized as a CTG/CAG tri-
(DYT12) nucleotide repeat expansion within the junctophilin-3
(JPH3) gene on chromosome 16q24.3 (Holmes et al.,
This disorder is inherited in an autosomal-dominant 2001). In the normal population the repeat length
manner (Brashear et al., 1998) and is due to muta- ranges from 6 to 27 CTG/CAG triplets, whereas
tion of ATP1A3 which encodes for the a3 subunit affected individuals have 4158 triplets. The repeat
of Na/K-ATPase (de Carvalho et al., 2004). The lies within an alternatively spliced exon of JPH3 and
onset may be in the teens or 20s and is often seen fol- because of variable splice acceptor sites may encode
lowing a period of physiologic stress, such as pro- either polyalanine or polyleucine or may be untrans-
longed physical exertion, fever or childbirth, when lated. Whether the expanded repeat is translated or
symptoms evolve over a period of hours or days and even transcribed remains unknown.
then plateau. Dystonia typically affects the cranial There can be marked variations in phenotype
musculature or the limbs and patients have addition- and parkinsonism, chorea or dystonia may be present
ally bradykinesia, dysphagia, dysarthria and gait (Walker et al., 2003a). The initial symptoms are often
516 R. H. WALKER
Table 59.3
Mixed movement disorders in which parkinsonism and dystonia may both occur; autosomal-dominant (AD), X-linked
inheritance and sporadic

Other neurolo-
Disease Inheritance Dystonia Parkinsonism gical features Affected protein Other useful tests

Huntingtons disease AD Chorea Huntingtin


Huntingtons disease-like 2 AD Chorea Junctophilin-3 Acanthocytosis
(10%)
Spinocerebellar ataxia III AD Chorea Ataxin 3
Spinocerebellar ataxia 17 AD Chorea TATA-binding
protein
Dentatorubropallidoluysian AD Chorea, Atrophin
atrophy ataxia
Neuroferritinopathy AD Chorea Ferritin light Ferritin, Fe in basal
chain ganglia on MRI
Fahrs disease AD, other Ataxia, chorea, ? Calcification on
(idiopathic basal cognitive, neuroimaging;
ganglia calcification) behavioral rule out
changes parathyroid
disease
McLeod syndrome X-linked Chorea, XK Kell and Kx ag,
seizures, acanthocytosis,
peripheral CK, LFTs
neuropathy
Neuroleptic-induced Sporadic
(acute)
Tardive dyskinesia Sporadic ? Chorea
Alzheimers disease Sporadic, Dementia, Presenilin,
AD rigidity amyloid
precursor
protein

absent; mild; moderate; marked; CK, creatine kinase; LFTs, liver function tests; MRI, magnetic resonance imaging.

a change in personality and cognitive functioning, evol- can be found on peripheral blood smear, resulting in
ving over 10 years or more to frank dementia. The neuro- confusion of the diagnosis with neuroacanthocytosis
logic presentation varies within families and in some (Walker et al., 2002b, 2003b).
cases changes with evolution of the disease. The relation-
ship between clinical features and size of the trinucleo-
tide repeat expansion at this point remains uncertain, 59.4.1.3. Spinocerebellar ataxias and
although, as in HD, the size of the repeat correlates dentatorubropallidoluysian atrophy
inversely with the age of onset (Margolis et al., 2004). The phenotypes of the SCAs can involve movement
The range of phenotypes is similar to that observed in disorders, in addition to signs and symptoms of cere-
HD, although parkinsonism can be prominent in adult- bellar neurodegeneration. These neurodegenerative
onset cases, as compared with HD when this presentation disorders are inherited in an autosomal-dominant man-
is usually associated with juvenile onset with very long ner and are usually due to expanded trinucleotide
repeat expansions. The mechanism of the phenotypic repeats within various proteins. The size of the expan-
variation in HDL2 remains to be determined. sions does not in general appear to correlate with the
All patients reported to date have been of African phenotype.
ancestry (Holmes et al., 2001; Margolis et al., 2001, The most common SCA in most populations, SCA
2004) and no patients of Caucasian ancestry have yet III (MachadoJoseph disease) can present with parkin-
been found (Andrew et al., 1994; Bauer et al., 2002; sonism, dystonia and chorea, usually in association
Stevanin et al., 2002). Occasionally, acanthocytosis with the typical cerebellar signs and eye findings.
PARKINSONISM AND DYSTONIA 517
A variety of movement disorders may be seen in 59.4.2. X-linked recessive (Table 59.3)
SCA 17, including parkinsonism, dystonia and chorea
59.4.2.1. McLeod syndrome
(Stevanin et al., 2003; Zuhlke et al., 2003), in addition
to the typical phenotype of ataxia, dementia and The X-linked McLeod neuroacanthocytosis syndrome
hyperreflexia. (Symmans et al., 1979; Takashima et al., 1994; Danek
SCA II has been reported to present with levodopa- et al., 2001) is similar to autosomal-recessive chorea-
responsive parkinsonism (Shan et al., 2001; Furtado acanthocytosis (ChAc) (see below) in its neurological
et al., 2004) but not dystonia. presentation, but other organ systems are also involved.
Dentatorubropallidoluysian atrophy (DRPLA) is Onset of clinical symptoms is typically in mid to late
found more often in Japanese populations, but rarely adulthood (Danek et al., 2001; Jung et al., 2001a).
in Caucasian (Le Ber et al., 2003; Martins et al., 2003) Patients may present with subtle neurobehavioral
or African-American (Burke et al., 1994) families and changes, particularly subcortical dementia, which are
may present with movement disorders, including dys- sometimes only fully recognized when the movement
tonia, myoclonus and chorea. Usual features are ataxia disorder develops. In addition to chorea, dystonia and
and dementia. Seizures are common in patients with parkinsonism may also be seen. Facial hyperkinesias
onset below the age of 20, but tend to decrease with time with dysarthria and involuntary vocalizations are fre-
and are rare in older-onset patients. quent, although the self-mutilating lip-biting seen in
autosomal-recessive ChAc is not characteristic. Periph-
eral sensorimotor neuropathy and areflexia are typical.
59.4.1.4. Neuroferritinopathy
Seizures are seen in 50% and can usually be controlled
This is one of the disorders that falls under the classi- with standard anticonvulsant medications.
fication of neurodegeneration with brain iron accu- Cardiomyopathy is present in approximately two-
mulation (NBIA), as the mutation of ferritin light thirds of patients and cardiac arrhythmias may be a
chain results in iron deposition in the basal ganglia significant source of morbidity and mortality (Danek
(Curtis et al., 2001). However, unlike the other disor- et al., 2001). Elevated serum creatine kinase (often
ders (panthothenate kinase-associated neurodegenera- into the 1000s) is an invariable finding and frank myo-
tion, aceruloplasminemia; see below), most of which pathy is common (Kawakami et al., 1999; Danek et al.,
are inherited in an autosomal-recessive fashion, this 2001). Liver enzymes are elevated and hepatospleno-
is an autosomal-dominant disorder. This may result megaly is present in 40% of patients and may lead to
in a variety of movement disorders, including chorea, evaluation for liver disease prior to neurologic presen-
dystonia and parkinsonism (Crompton et al., 2004; tation (Walker et al., 2005).
Mir et al., 2004), with onset at age 4055 years. Cog- Often patients are identified, as was the eponymous
nitive impairment is not normally seen, although it can subject McLeod, at blood banks by blood antigen typ-
occasionally be a feature (Wills et al., 2002). Serum ing. Rare patients with the McLeod red blood cell
ferritin values tend to be below, or at the lower end antigen phenotype, but without any neurological or
of, the normal range. systemic abnormalities, have been reported (Jung
et al., 2003; Walker et al., in press). Occasionally car-
59.4.1.5. Fahrs Disease rier females have been symptomatic, presumably due
Fahrs disease (idiopathic basal ganglia calcifica- to X-chromosome inactivation (Hardie et al., 1991;
tion; IBGC) refers to a heterogeneous group of disor- Ueyama et al., 2000; Jung et al., 2001a).
ders in which there is deposition of calcium in the Neuroimaging typically shows a decrease in basal
basal ganglia and other cerebral regions, particularly ganglia volume (Danek et al., 2001), although white-
the deep cerebellar nuclei. The clinical picture may matter changes have also been reported (Nicholl
include dystonia, parkinsonism, chorea, ataxia, cog- et al., 2004). Neuropathologic findings appear non-
nitive impairment and behavioral changes. In one specific, with neuronal loss and reactive gliosis (Brin
family, linkage to 14q was demonstrated (IBC1) et al., 1993; Geser et al., 2006).
(Geschwind et al., 1999) although the gene has not The phenotype is defined by the characteristics of
yet been identified. In several other families with auto- the red blood cell (RBC) Kell antigen system (Allen
somal-dominant inheritance, linkage to this locus was et al., 1961), the third most important erythrocyte anti-
excluded (Brodaty et al., 2002; Oliveira et al., 2004). gen system after ABO and rhesus. Mutation of the XK
In other families (Reske-Nielsen et al., 1988; gene causes reduced or absent XK protein. As the XK
Younes-Mhenni et al., 2002), the pattern of inheritance protein is closely associated with the Kell proteins and
and additional clinical features suggest mitochondrial determines Kell antigen expression, this results in a
inheritance. reduction in the Kell protein on the RBC membrane
518 R. H. WALKER
(Russo et al., 1998). Patients thus have absent Kx anti- to the clinical appearance of parkinsonism (Rinne
gen expression and reduced Kell antigen expression. et al., 1994).
The diagnosis can only be confirmed at specialized
laboratories which have the requisite panel of anti-Kell 59.4.3.2. Wilsons disease
and anti-Kx antibodies. XK may function as a membrane
This autosomal-recessive disorder of copper metabo-
transport protein (Ho et al., 1994) and has been found in
lism may present with a variety of abnormal move-
muscle and brain in addition to RBCs (Ho et al., 1994;
ments. The causative mutation is of the ATP7B
Jung et al., 2001a, b; Russo et al., 2000). This abnormal-
gene, resulting in an inability to transport copper on to
ity of RBC membrane proteins is presumably the cause
ceruloplasmin, with resultant copper accumulation in
of the characteristic acanthocytosis.
brain, liver, cornea and elsewhere. The typical neurolo-
gical presentation is with an asymmetric, flapping arm
59.4.3. Autosomal-recessive (Table 59.4) tremor, which may vary in amplitude, distribution and
59.4.3.1. Chorea-acanthocytosis the position in which it is elicited. The gait is often
affected and may be parkinsonian or cerebellar. Dysto-
ChAc, previously known as neuroacanthocytosis or
nia of the face, tongue and pharynx can be seen, which,
LevineCritchley syndrome (Levine et al., 1960;
in combination with cerebellar dysfunction, may result
Critchley et al., 1968), is an autosomal-recessive disor-
in dysarthria. The classic risus sardonicus of Wilsons
der due to a mutation of the VPS13A gene (formerly
disease is due to dystonia affecting the lower facial mus-
CHAC) (Velayos-Baeza et al., 2004) located on chro-
cles. A hereditary whispering dysphonia (DYT4) is
mosome 9q21 (Rubio et al., 1999; Ueno et al., 2001).
associated with Wilsons disease (Elwes and Saunders,
Similar to XK (see above), this protein appears to be
1986).
involved in membrane processing and sorting, which
Limb and trunk dystonia can also be a feature,
may account for the RBC membrane abnormalities.
although these are extremely uncommon as a presen-
The movement disorder in this neurodegenerative
tation of Wilsons disease in the absence of hepatic
disease is typically chorea, presenting in young to
features. In a series of patients with cervical dystonia
mid-adulthood with marked lingual-buccal-facial dyski-
as the sole symptom, no cases of Wilsons disease
nesia and self-mutilation (Rampoldi et al., 2002). As
were detected (Risvoll and Kerty, 2001).
with several other basal ganglia disorders described
On neuroimaging and neuropathological evaluation,
here, these patients may present with psychiatric
the basal ganglia and brainstem are consistently
(Rovito and Pirone, 1963; Bruneau et al., 2003), beha-
affected by abnormal copper deposition (Magalhaes
vioral or cognitive problems. Dystonia is found in 50%
et al., 1994; Sener, 2003; Page et al., 2004), thus the
of patients (Rampoldi et al., 2002). Other abnormal
presence of movement disorders is not surprising.
movements are often seen, including vocal and motor
The importance of awareness of Wilsons disease as
tics (Saiki et al., 2004). Parkinsonism is reported in
part of the differential diagnosis of movement disor-
32% of patients (Rampoldi et al., 2002) and may be a
ders is that it is one of the few etiologies that can be
presenting feature (Bostantjopoulou et al., 2000).
effectively treated, thus a high level of suspicion is
Widespread involvement of the nervous system is sug-
always warranted. Testing for serum ceruloplasmin
gested by the presence of seizures, cognitive impair-
levels, 24-hour copper excretion and slit-lamp exami-
ment and peripheral neuropathy. Outside the nervous
nation for KayserFleischer rings are all necessary to
system, the liver and spleen may be enlarged, with
exclude the diagnosis.
abnormal liver function tests, although this is less com-
mon than in McLeod syndrome (Rampoldi et al.,
2002). Unlike McLeod syndrome, cardiomyopathy is 59.4.3.3. Aceruloplasminemia
very rare. Deficiency of ceruloplasmin, due to autosomal-reces-
A Japanese family with apparent autosomal-domi- sive inheritance of mutations of the gene for cerulo-
nant inheritance of this disorder, documented muta- plasmin, results in iron deposition in the retina,
tions of VPS13A and an apparently identical pancreas, cerebellum and basal ganglia (Miyajima,
phenotype to the autosomal-recessive form has been 2003; Xu et al., 2004). Typical presentation is of ret-
reported (Saiki et al., 2003). Diagnosis of this disorder inal degeneration and diabetes mellitus in the 20s. In
can be confirmed by a quantitative test for chorein the 40s and 50s neurological signs appear, usually
(Dobson-Stone et al., 2004). ataxia. Subsequently dystonia, especially orofacial, par-
Neuropathologically, there is degeneration of the kinsonism and chorea, may develop. Dementia may
caudate nucleus and putamen and there may be manifest in later years. Symptomatic heteroplasmic
marked loss of substantia nigra neurons, corresponding carriers have also been reported.
Table 59.4
Mixed movement disorders in which parkinsonism and dystonia may both occur; autosomal-recessive (AR) inheritance

Inheritance Dystonia Parkinsonism Other neurological features Affected protein Other tests

Chorea-acanthocytosis AR Chorea, peripheral neuropathy, Chorein Acanthocytosis, CK, LFTs,


seizures chorein assay
Wilsons disease AR Tremor, ataxia ATP7B Ceruloplasmin, Cu
excretion, Kayser
Fleischer rings
Aceruloplasminemia AR Ataxia, chorea Ceruloplasmin Fe on MRI in basal ganglia
Pantothenate kinase- AR Chorea, spasticity, dementia, Pantothenate kinase 2 Acanthocytes (8%)
associated retinal degeneration
neurodegeneration

PARKINSONISM AND DYSTONIA


GM1 gangliosidosis type 3 AR Cerebellar, dysarthria, dementia b-galactosidase Bone marrow foam or
Gaucher-like cells
GM2 gangliosidosis late- AR / Cerebellar, spasticity, seizures, Hexosaminidase A High T2 in basal ganglia on
onset peripheral neuropathy, MRI
supranuclear vertical gaze palsy
NiemannPick type C AR Supranuclar vertical gaze palsy, Sphingomyelinase Bone marrow sea-blue
cerebellar, intention tremor histiocytes
Gaucher disease type III AR (facial) Seizures, myoclonus, horizontal Glucocerebroside Bone marrow Gaucher
gaze palsy cells
Glutaric aciduria AR Encephalopathy Glutaryl-coenzyme A Urinary amino acids
dehydrogenase
2-Hydroxyglutaric AR Encephalopathy, seizures 2-hydroxyglutaric acid Urinary amino acids
aciduria dehydrogenase (l or d
isoform)
Homocystinuria AR / Vascular thrombosis, mental Cystathione beta-synthase Urinary amino acids
retardation
Biotin-responsive basal AR Encephalopathy, quadriparesis, ?Biotin transporter Biotin-responsiveness
ganglia disease spasicity, seizures
Juvenile neuronal ceroid AR Dementia, seizures, visual loss CLN3 Skin biopsy
lipofuscinosis
Adult neuronal ceroid AR (facial) Epilepsy, myoclonus dementia; Skin biopsy
lipofuscinosis extrapyramidal signs, dementia
Neuronal intranuclear AR (also Cerebellar, corticospinal Neuropathology
(hyaline) inclusion sprodic,
disease AD?)

AD, autosomal dominant; AR, autosomal recessive; absent; mild; moderate; marked; CK, creatine kinase; LFTs, liver function tests; MRI, magnetic resonance imaging.

519
520 R. H. WALKER
Copper metabolism is not disturbed by the lack enzyme function (Hayflick et al., 2003). The distribu-
of ceruloplasmin. However, as ceruloplasmin func- tion of the neurological lesions is thought to relate to
tions as a ferroxidase, iron oxidation from Fe2 to the accumulation of iron and other neurotoxic sub-
Fe3 is impaired and neurons are also more vulnerable stances and to local tissue demand for coenzyme A
to oxidative stress. Neuropathologically, astrocytes (Zhou et al., 2001).
and neurons laden with iron are found in the cerebel-
lum, basal ganglia and cortex (Miyajima, 2003; Xu 59.4.3.5. GM1 gangliosidosis
et al., 2004). The late-onset, chronic form (adult; type 3) of GM1
gangliosidosis is slowly progressive and may present
during childhood or as late as the fourth decade. This
59.4.3.4. Pantothenate kinase-associated
form affects only the central nervous system and does
neurodegeneration
not affect the skeletal system. Dystonia is often
The disorder now known as pantothenate kinase-asso- marked, especially orofacial, and dysarthria and dys-
ciated neurodegeneration (PKAN) is due to mutations phagia may be severe. Features of parkinsonism are
of pantothenate kinase 2 (PANK2) located on chromo- also reported (Goldman et al., 1981; Uyama et al.,
some 20p13 (Zhou et al., 2001). The course in typical 1992; Yoshida et al., 1994; Muthane et al., 2004).
cases is of disease onset by the age of 10 years with MRI frequently shows bilateral putaminal hyperinten-
dystonia and a rapid progression over the next 10 years sities on T2 sequences (Uyama et al., 1992; Muthane
(Hayflick et al., 2003). Orofacial and limb dystonia, et al., 2004).
choreoathetosis and spasticity are characteristic early
features. Approximately one-third of typical cases 59.4.3.6. GM2 gangliosidosis
develop cognitive impairment and two-thirds have
The late-onset forms of GM2 gangliosidosis may
retinopathy. The typical magnetic resonance imaging
occur in childhood, adolescence or adulthood as either
(MRI) eye of the tiger pattern of iron deposition in
subacute or chronic forms. Dystonia and choreoatheto-
the globus pallidus was seen in the majority of these
sis have been reported in the subacute forms, along
patients (Hayflick et al., 2003; Hartig et al., 2006).
with dysarthria, speech loss, ataxia, spasticity, seizures
8% of typical PANK patients have acanthocytosis.
and behavioral changes (Meek et al., 1984; Hardie
The occasional reported association of decreased pre-
et al., 1988; Nardocci et al., 1992). In general, cerebel-
betalipoprotein levels (hypoprebetalipoproteinemia,
lar and corticospinal abnormalities tend to predomi-
acanthocytosis, retinitis pigmentosa and pallidal degen-
nate over basal ganglia-related symptomatology.
eration: HARP syndrome) (Orrell et al., 1995; Malan-
Parkinsonism has rarely been reported (Inzelberg and
drini et al., 1996) in patients with PANK2 mutations
Korczyn, 1994). Supranuclear ophthalmoplegia and
(Ching et al., 2002) does not appear to be of clinical sig-
oculomotor apraxia may occur (Specola et al., 1990)
nificance and may be observed in normal subjects
and thus this disorder should be considered in atypical
(Houlden et al., 2003; Danek and Hegele, 2004).
presentations of PSP.
In atypical cases disease onset is after the age of 20,
with dystonia, rigidity and gait freezing, but slower
59.4.3.7. NiemannPick type C
progression. Early speech difficulty, with pallilalia or
dysarthria, is common, as are cognitive decline and This disorder may present in childhood or even late into
personality change. These patients do not tend to have adulthood with cerebellar signs, intention tremor and
retinopathy. In one-third of these there is a mutation dysarthria. Dystonia and chorea-athetosis may be pre-
of PANK2 and the diagnostic MRI finding, whereas sent and, as with GM2 gangliosidosis, the presence of
in two-thirds PANK2 mutations were not found, nor supranuclear gaze palsy, specifically loss of vertical
was there the typical MRI image (Hayflick et al., gaze, may lead this disorder to resemble PSP (Coleman
2003). These are classified as NBIA without PANK2 et al., 1988; Cardoso and Camargos, 2000).
mutation.
The majority of clinically typical cases are due to 59.4.3.8. Gaucher disease type III
mutations of PANK2 causing protein truncation. Pan- The neuropathic form of this disease presents from
tothenate kinase catalyzes the rate-limiting step in the childhood until early adulthood, with action myo-
synthesis of coenzyme A from vitamin B5 (pantothe- clonus and seizures and a supranuclear palsy of hori-
nate). The amount of active enzyme correlates with zontal gaze. Occasionally facial grimacing may be
the disease phenotype, as typical patients have no observed. An association between heterozygous car-
active enzyme but atypical patients, in whom there is riers of the mutation and variably treatment-responsive
a missense mutation of PANK2, may have some parkinsonism has been reported (Tayebi et al., 2003;
PARKINSONISM AND DYSTONIA 521
Aharon-Peretz et al., 2004; Goker-Alpan et al., 2004). of microcephaly and hypotonia, with atrophy of the
It is hypothesized that mutations of the glucocere- caudate nuclei, due to a mutation of CLN2 (Simonati
brosidase gene confer susceptibility to parkinsonism, et al., 2000). This encodes for a lysosomal enzyme
possibly by interfering with protein degradation in tripetidyl peptidase.
the SNpc. In the juvenile form (late-onset Batten disease;
SpielmeyerVogt disease), usually due to mutations
59.4.3.9. Glutaric aciduria of CLN3, a gene for a membrane protein of unknown
Glutaric acidura typically presents with generalized dys- function, involvement of the extrapyramidal system
tonia and parkinsonism may also be a feature (Gascon is typically a late feature, following intellectual
et al., 1994), in addition to the other features of encepha- decline, visual loss and seizures (Aberg et al., 2001).
lopathy. Often the presentation is catastrophic in early Rigidity and impaired postural reflexes usually occur
infancy, but occasionally it may present more gradually late, although occasionally may be presenting features.
in later childhood or even adulthood. On MRI, dilation Dystonia may sometimes be seen (Boustany et al.,
of the sylvian fissures and lesions of the putamen can 1988).
be seen. The adult form (Kufs disease) may present in one of
2-Hydroxyglutaric aciduria is considerably rarer two forms, with epilepsy, myoclonus dementia and
and has been reported as a cause of parkinsonism with extrapyramidal signs (Yoshioka et al., 1999; Nijssen
dystonia (Owens and Okun, 2004). et al., 2002) or with behavioral disturbances, dementia
and facial dyskinesias. Parkinsonism has been reported
59.4.3.10. Homocystinuria (Nijssen et al., 2002), but not dystonia. The gene has
been assigned the name CLN4, but it has not as yet
Movement disorders are uncommon in this disorder,
been identified.
which is characterized by marfanoid features, cataracts,
vascular thrombosis and variable mental retardation.
59.4.3.13. Neuronal intranuclear hyaline inclusion
Occasionally severe dystonia may be seen (Hagberg
disease
et al., 1970; Davous and Rondot, 1983; Kempster
et al., 1988; Berardelli et al., 1991). Parkinsonism has This is a heterogeneous disorder diagnosed neuro-
occasionally been reported (Keskin and Yurdakul, pathologically with widespread intranuclear inclusions
1996), in one case in the brother of a patient with severe throughout the central, peripheral and autonomic ner-
dystonia (Ekinci et al., 2004). vous system (Takahashi-Fujigasaki, 2003). The onset
may be at any age and those with young onset tend
59.4.3.11. Biotin-responsive basal ganglia disease to have parkinsonism and dystonia, in addition to
behavioral changes, corticospinal, cerebellar signs
This disorder, probably of autosomal-recessive inheri-
and seizures. Autosomal-recessive inheritance is sug-
tance, was reported in a series of 10 patients, who had
gested by several affected sib-pairs, although in one
onset in infancy of a subacute encephalopathy, pro-
family autosomal-dominant inheritance was apparent
gressing to dystonia, quadriparesis, cogwheel rigidity,
(Takahashi-Fujigasaki, 2003).
cranial neuropathies and seizures (Ozand et al.,
1998). Symptoms improved remarkably with the
59.4.4. Sporadic (Table 59.3)
administration of biotin, but recurred when it was
discontinued. 59.4.4.1. Drug-induced syndromes
Treatment with the classic dopamine receptor-blocking
59.4.3.12. Neuronal ceroid lipofuscinoses antipsychotic agents, such as thorazine, chlorproma-
The combination of dystonia and parkinsonism has not zine, haloperidol, perphenazine and many others, may
been reported in the various forms of neuronal ceroid result in an acute dystonic crisis characterized by
liposcinosis; however, they are individually rare fea- extension of the neck and rolling up of the eyes in
tures at different stages of the disease, indicating the the head, known as an oculogyric crisis. This happens
potential for basal ganglia involvement. Mutations of particularly in young males following initiation of
a number of genes (CLN1, CLN2, CLN3, CLN5, therapy with an intramuscular depot injection. These
CLN6, CLN8) for endosomal-lysosomal proteins have episodes respond well to intravenous anticholinergic
been reported to be associated with different disease agents and these medications are often given orally
phenotypes (Mole, 2004). prophylactically.
Dystonia has occasionally been reported in the The dopamine receptor-blocking effects of the neu-
infantile form, along with an unusual phenotype roleptics are well-recognized as a cause of iatrogenic
522 R. H. WALKER
parkinsonism, but are becoming less widespread as the are present is to improve both aspects of the movement
atypical antipsychotics, such as clozapine, olanzepine, disorder, without the treatment for one exacerbating
quetiapine, aripiprazole and ziprasidone, are used the other.
more extensively. Although risperidone was initially In a small number of disorders, therapy is definitive
classified as an atypical antipsychotic, clinical experi- and essential to minimize disease progression. It is
ence demonstrates that it may often cause parkin- important to consider the diagnosis of dopa-responsive
sonism, especially in patients with an underlying dystonia, especially in pediatric cases with an atypical
neurodegenerative process, and thus should be avoided presentation which may resemble cerebral palsy
in these patients. Once the offending medication (Nygaard et al., 1994). A diagnostic trial of levodopa
is removed, the symptoms of parkinsonism should is obligatory in most cases of dystonia and definitely
resolve. in cases with concomitant parkinsonism. The diagnosis
There is less awareness of the potential side-effects of Wilsons disease is also important to consider in
of other dopamine receptor-blocking agents not used almost every unusual presentation of a movement dis-
as neuroleptics, such as metoclopramide, compazine, order, in order to institute the appropriate therapy if
flunarizine and cinnarizine, but these medications can indicated.
also be responsible for the generation of the same Parkinsonism may respond to dopaminergic agents,
range of movement disorders. especially if there is impairment of the nigrostriatal
The more serious and long-lasting consequence of pathway, rather than of the striatal medium spiny pro-
classic neuroleptic use is tardive dyskinesia. Chorei- jection neurons which bear the postsynaptic dopamine
form orofacial dyskinesias are the most typical form, receptors. Levodopa/carbidopa is usually the first-line
but tardive dystonias are not uncommon, including medication and should be increased very cautiously,
blepharospasm, spasmodic dysphonia, cervical and with careful monitoring for worsening of psychiatric
truncal dystonia. The movements may emerge as the symptoms and hyperkinetic movements. The usual
medication is discontinued or whilst it is still being strategy is gradually to increase the levodopa over a
used. In the latter situation, patients may manifest both number of weeks to approximately 2000 mg, if toler-
dystonia and parkinsonism. ated, and then slowly to decrease the dose. If there is
no benefit from this, then dopamine receptor agonists
should be tried. Benefits have been reported in patients
59.4.4.2. Alzheimers disease with secondary parkinsonism of a variety of etiologies
A variety of movement disorders can be seen in (Low and Allsop, 1973; Bonelli et al., 2002; Tayebi
Alzheimers disease. Myoclonus may be seen in spora- et al., 2003; Goker-Alpan et al., 2004).
dic cases and in patients with presenilin mutations. Treatment of dystonia may be frustrating and the
Occasional families with mutations of amyloid precur- use of systemic medications may result in exacerbation
sor protein may have prominent parkinsonism and of neuropsychiatric symptoms. Local injections of
myoclonus (Edwards-Lee et al., 2005), but this is not botulinum toxin may be helpful. The armamentarium
typical. Generalized rigidity is often a feature of late- of potentially effective medications includes agents
stage disease and facial masking may give a parkinso- with a wide variety of mechanisms of action. Anticho-
nian appearance. Retrocollis can also occur and linergics, benzodiazepines, anticonvulsants and baclo-
responds well to injection of botulinum toxin. fen (oral or intrathecal (Ford et al., 1996; Walker
Patients with Alzheimers disease are particularly et al., 2000)) may all be useful, but results can be quite
vulnerable to side-effects of medications, in particular variable. The newer atypical antipsychotics, such as
to parkinsonism or dystonia induced by neuroleptics, quetiapine, clozapine, aripiprazole and ziprasidone,
including risperidone (Magnuson et al., 2000). Truncal may also have benefits, with less potential for exacer-
dystonia (Pisa syndrome) has been reported following bating parkinsonism or causing tardive dyskinesias as
the use of acetylcholinesterase inhibitors (Kwak can be seen with the typical antipsychotics.
et al., 2000; Miyaoka et al., 2001). DBS of either the GPi or the STN has become
accepted as treatment for advanced PD and can also
have good results in primary (Toda et al., 2004;
59.5. Therapy Vidailhet et al., 2005) or tardive (Trottenberg et al.,
2005) dystonia. In general, secondary parkinsonian or
Therapy for these disorders ideally would be directed dystonic (Eltahawy et al., 2004) disorders do not tend
towards the etiologic causes; however, in the majority to benefit from surgical interventions, probably due
of cases this is not possible. The challenge in sympto- to more extensive disruption of neuronal pathways
matic treatment when both parkinsonism and dystonia than in primary dystonia or PD. Occasional cases
PARKINSONISM AND DYSTONIA 523
may respond (Wohrle et al., 2003; Umemura et al., References
2004). A case report of DBS of the GPi in HD (Moro
et al., 2004) was promising, although in a case of ChAc Aberg LE, Rinne JO, Rajantie I et al. (2001). A
it was not beneficial (Wihl et al., 2001). The motor thala- favorable response to antiparkinsonian treatment in juve-
mus has also been proposed as a potentially promising nile neuronal ceroid lipofuscinosis. Neurology 56:
site for DBS and has been reported as having positive 12361239.
results in a patient with ChAc (Burbaud et al., 2002). Aharon-Peretz J, Rosenbaum H, Gershoni-Baruch R (2004).
It is suspected that the mechanisms of action of Mutations in the glucocerebrosidase gene and Parkinsons
DBS in PD and dystonia are different as the stimula- disease in Ashkenazi Jews. N Engl J Med 351:
19721977.
tion parameters at which benefits are seen may differ
Aizawa H, Kwak S, Shimizu T et al. (1991). A case of adult
significantly in the two conditions. It is postulated that onset pure pallidal degeneration. I. Clinical manifestations
blocking disordered neuronal signaling from the out- and neuropathological observations. J Neurol Sci 102:
put nuclei of the basal ganglia, the GPi and SNpr, is 7682.
the critical aspect of DBS; however, this therapy is Albanese A (2003). Dystonia in parkinsonian syndromes.
still poorly understood. Adv Neurol 91: 351360.
A number of the disorders described here may pre- Albin RL, Young AB, Penney JB (1989). The functional
sent initially with subtle psychiatric or neurobeha- anatomy of basal ganglia disorders. Trends Neurosci 12:
vioral features. The use of neuroleptics may mask the 366375.
correct diagnosis, being presumed to be the cause of Allen FH, Krabbe SM, Corcoran PA (1961). A new pheno-
any movement disorders that develop. A high degree type (McLeod) in the Kell blood-group system. Vox Sang
of suspicion is necessary as well as alertness for the 6: 555560.
presence of additional neurologic or medical features, Andrew SE, Goldberg YP, Kremer B et al. (1994). Hunting-
a family history or an unusual disease course that ton disease without cag expansionphenocopies or errors
may provide important clues to the correct diagnosis. in assignment. Am J Hum Genet 54: 852863.
Arbuthnott GW, Ingham CA, Wickens JR (2000). Dopamine
and synaptic plasticity in the neostriatum. J Anat 196:
59.6. Conclusion
587596.
Asanuma K, Ma Y, Okulski J et al. (2005). Decreased striatal
The phenomenology and pathophysiology of diseases in D2 receptor binding in non-manifesting carriers of the
which parkinsonism and dystonia coexist may lead us to DYT1 dystonia mutation. Neurology 64: 347349.
important insights into the function of the basal ganglia Augood SJ, Hollingsworth Z, Albers DS et al. (2002). Dopa-
and of dopamine specifically. An anatomically related mine transmission in DYT1 dystonia: A biochemical and
vulnerability to dopamine dysfunction, presumably autoradiographical study. Neurology 59: 445448.
within the putamen, is suggested by the pattern of onset, Avshalumov MV, Chen BT, Marshall SP et al. (2003). Gluta-
for example, the prominence of foot dystonia in early mate-dependent inhibition of dopamine release in striatum
PD, childhood-onset DYT1 dystonia and DRD. Distur- is mediated by a new diffusible messenger, H2O2.
bances in dopaminergic neurotransmission are clearly J Neurosci 23: 27442750.
Bamford NS, Robinson S, Palmiter RD et al. (2004a). Dopa-
implicated in parkinsonism and dystonia. However,
mine modulates release from corticostriatal terminals.
the benefits of mere replacement are limited, particu- J Neurosci 24: 95419552.
larly in dystonia, and it is evident that dopamine is more Bamford NS, Zhang H, Schmitz Y et al. (2004b). Heterosyn-
than an onoff switch for movement. The partial aptic dopamine neurotransmission selects sets of corticos-
positive effects seen with the wide variety of medica- triatal terminals. Neuron 42: 653663.
tions used therapeutically in dystonia suggest complex Bao L, Patel JC, Walker RH et al. (2006). Alterations in
interactions of a number of different neurotransmitters. striatal dopamine release in DYT1 mice: implications for
Sound scientific methodology requires that we do early onset dystonia. Neuroscience Meeting Planner 78.8/
not infer the function of a specific component in a sys- JJ1 Society for Neuroscience online.
tem by the consequences of its dysfunction. However, Bara-Jimenez W, Shelton P, Hallett M (2000). Spatial discri-
it is impossible to resist speculating about the role of mination is abnormal in focal hand dystonia. Neurology
55: 18691873.
dopamine in motor programs and how its deficiency
Barclay CL, Lang AE (1997). Dystonia in progressive supra-
may result in both impaired and excessive movement. nuclear palsy. J Neurol Neurosurg Psychiatry 62:
Models of basal ganglia function are invaluable in 352356.
designing experiments and potential therapeutic strate- Bauer I, Gencik M, Laccone F et al. (2002). Trinucleotide
gies. The limitation of the models in explaining the repeat expansions in the junctophilin-3 gene are not found
coexistence of hypo- and hyperkinetic disorders sug- in Caucasian patients with a Huntingtons disease-like
gests that yet another layer of complexity is required. phenotype. Ann Neurol 51: 662.
524 R. H. WALKER

Berardelli A, Thompson PD, Zaccagnini M et al. (1991). sentation of the hand in the primary somatosensory cortex
Two sisters with generalized dystonia associated with in adult monkeys. Neurology 47: 508520.
homocystinuria. Mov Disord 6: 163165. Cairns NJ, Grossman M, Arnold SE et al. (2004). Clinical
Berardelli A, Rothwell JC, Hallett M et al. (1998). The patho- and neuropathologic variation in neuronal intermediate
physiology of primary dystonia. Brain 121: 11951212. filament inclusion disease. Neurology 63: 13761384.
Bergman H, Wichmann T, Karmon B et al. (1994). The Calabresi P, Mercuri N, Stanzione P et al. (1987). Intracellu-
primate subthalamic nucleus. II. Neuronal activity in the lar studies on the dopamine-induced firing inhibition of
MPTP model of parkinsonism. J Neurophysiol 72: neostriatal neurons in vitro: Evidence for D1 receptor
507520. involvement. Neuroscience 20: 757771.
Boesch SM, Wenning GK, Ransmayr G et al. (2002). Calabresi P, Pisani A, Mercuri NB et al. (1996). The corti-
Dystonia in multiple system atrophy. J Neurol Neurosurg costriatal projection: From synaptic plasticity to dysfunc-
Psychiatry 72: 300303. tions of the basal ganglia [see comments]. Trends
Bonelli RM, Niederwieser G, Diez J et al. (2002). Pramipex- Neurosci 19: 1924.
ole ameliorates neurologic and psychiatric symptoms in a Calabresi P, Saiardi A, Pisani A et al. (1997). Abnormal
Westphal variant of Huntingtons disease. Clin Neuro- synaptic plasticity in the striatum of mice lacking dopa-
pharmacol 25: 5860. mine D2 receptors. J Neurosci 17: 45364544.
Bonifati V, Rizzu P, Squitieri F et al. (2003). DJ-1( PARK7), Cardoso F, Camargos S (2000). Juvenile parkinsonism: a
a novel gene for autosomal recessive, early onset parkin- heterogeneous entity. Eur J Neurol 7: 467471.
sonism. Neurol Sci 24: 159160. Carpenter MB, Carleton SC, Keller JK et al. (1981). Connec-
Bostantjopoulou S, Katsarou Z, Kazis A et al. (2000). Neu- tions of the subthalamic nucleus in the monkey. Brain Res
roacanthocytosis presenting as parkinsonism. Mov Disord 224: 129.
15: 12711273. Ching KHL, Westaway SK, Gitschier J et al. (2002). HARP
Boustany RM, Alroy J, Kolodny EH (1988). Clinical classi- syndrome is allelic with pantothenate kinase-associated
fication of neuronal ceroid-lipofuscinosis subtypes. Am neurodegeneration. Neurology 58: 16731674.
J Med Genet Suppl 5: 4758. Clark LN, Afridi S, Mejia-Santana H et al. (2004). Analysis
Brashear A, Butler IJ, Ozelius LJ et al. (1998). Rapid-onset of an early-onset Parkinsons disease cohort for DJ-1
dystonia-parkinsonism: a report of clinical, biochemical, mutations. Mov Disord 19: 796800.
and genetic studies in two families. Adv Neurol 78: Coleman RJ, Robb SA, Lake BD et al. (1988). The diverse
335339. neurological features of Niemann-Pick disease type C: a
Bressman SB (2004). Dystonia genotypes, phenotypes, and report of two cases. Mov Disord 3: 295299.
classification. Adv Neurol 94: 101107. Cragg SJ (2003). Variable dopamine release probability and
Bressman SB, de Leon D, Raymond D et al. (1998). Clinical- short-term plasticity between functional domains of the
genetic spectrum of primary dystonia. Adv Neurol 78: primate striatum. J Neurosci 23: 43784385.
7991. Critchley EM, Clark DB, Wikler A (1968). Acanthocytosis
Brin MF, Hays A, Symmans WA et al. (1993). Neuropathol- and neurological disorder without betalipoproteinemia.
ogy of McLeod phenotype is like chorea-acanthocytosis Arch Neurol 18: 134140.
(CA). Can J Neurol Sci 20 (Suppl 4): 234. Crompton DE, Chinnery PF, Bates D et al. (2004). Spectrum
Brodaty H, Mitchell P, Luscombe G et al. (2002). Familial of movement disorders in neuroferritinopathy. Mov
idiopathic basal ganglia calcification (Fahrs disease) Disord 20: 9599.
without neurological, cognitive and psychiatric symptoms Curtis AR, Fey C, Morris CM et al. (2001). Mutation in
is not linked to the IBGC1 locus on chromosome 14q. the gene encoding ferritin light polypeptide causes domi-
Hum Genet 110: 814. nant adult-onset basal ganglia disease. Nat Genet 28:
Bruneau MA, Lesperance P, Chouinard S (2003). Schizophre- 350354.
nia-like presentation of neuroacanthocytosis. J Neuropsy- Danek A, Hegele RA (2004). Compound heterozygous
chiatry Clin Neurosci 15: 378380. PANK2 mutations confirm HARP and Hallervorden-Spatz
Bugiani O, Tabaton M, Cammarata S (1984). Huntingtons syndromes are allelic. Neurology. On-line correspondence
disease: Survival of large striatal neurons in the rigid http://www.neurology.org/cgi/eletters/61/10/1423.
variant. Ann Neurol 15: 154156. Danek A, Rubio JP, Rampoldi L et al. (2001). McLeod
Burbaud P, Vital A, Rougier A et al. (2002). Minimal tissue neuroacanthocytosis: Genotype and phenotype. Ann
damage after stimulation of the motor thalamus in a case Neurol 50: 755764.
of chorea-acanthocytosis. Neurology 59: 19821984. Davous P, Rondot P (1983). Homocystinuria and dystonia.
Burke JR, Wingfield MS, Lewis KE et al. (1994). The Haw J Neurol Neurosurg Psychiatry 46: 283286.
River syndrome: dentatorubropallidoluysian atrophy de Carvalho AP, Sweadner KJ, Penniston JT et al. (2004).
(DRPLA) in an African-American family. Nat Genet 7: Mutations in the Na/K-ATPase alpha3 gene ATP1A3
521524. are associated with rapid-onset dystonia parkinsonism.
Byl NN, Merzenich MM, Jenkins WM (1996). A primate Neuron 43: 169175.
genesis model of focal dystonia and repetitive strain DeLong MR (1990). Primate models of movement disorders
injury: I. Learning-induced dedifferentiation of the repre- of basal ganglia origin. Trends Neurosci 13: 281285.
PARKINSONISM AND DYSTONIA 525

Djaldetti R, Mosberg-Galili R, Sroka H et al. (1999). Cam- Ford B, Greene P, Louis ED et al. (1996). Use of intrathecal
ptocormia (bent spine) in patients with Parkinsons baclofen in the treatment of patients with dystonia. Arch
diseasecharacterization and possible pathogenesis of an Neurol 53: 12411246.
unusual phenomenon. Mov Disord 14: 443447. Fukuda M, Mentis MJ, Ma Y et al. (2001). Networks mediat-
Dobson-Stone C, Velayos-Baeza A, Filippone LA et al. ing the clinical effects of pallidal brain stimulation for
(2004). Chorein detection for the diagnosis of chorea- Parkinsons diseasea PET study of resting-state glucose
acanthocytosis. Ann Neurol 56: 299302. metabolism. Brain 124: 16011609.
Edwards-Lee T, Ringman JM, Chung J et al. (2005). An African Furtado S, Payami H, Lockhart PJ et al. (2004). Profile of
American family with early-onset Alzheimer disease and an families with parkinsonism-predominant spinocerebellar
APP (T714I) mutation. Neurology 64: 377379. ataxia type 2 (SCA2). Mov Disord 19: 622629.
Eidelberg D (1998). Abnormal brain networks in DYT1 dys- Furukawa Y, Lang AE, Trugman JM et al. (1998). Gender-
tonia. Adv Neurol 78: 127133. related penetrance and de novo GTP-cyclohydrolase I
Eidelberg D, Moeller JR, Dhawan V et al. (1990). The meta- gene mutations in dopa-responsive dystonia [see com-
bolic anatomy of Parkinsons disease: complementary ments]. Neurology 50: 10151020.
fluorodeoxyglucose and fluorodopa positron emission Gascon GG, Ozand PT, Brismar J (1994). Movement dis-
tomographic studies. Mov Disord 5: 203213. orders in childhood organic acidurias. Clinical, neuroima-
Eidelberg D, Moeller JR, Isikawa T et al. (1995). Assessment ging, and biochemical correlations. Brain Dev 16 (Suppl):
of disease severity in parkinsonism with fluorine-18-fluor- 94103.
odeoxyglucose and PET. J Nucl Med 36: 378383. Gerardin E, Lehericy S, Pochon JB et al. (2003). Foot, hand,
Eidelberg D, Moeller JR, Ishikawa T et al. (1996). Regional face and eye representation in the human striatum. Cereb
metabolic correlates of surgical outcome following unilat- Cortex 13: 162169.
eral pallidotomy for Parkinsons disease. Ann Neurol 39: Geschwind DH, Loginov M, Stern JM (1999). Identification
450459. of a locus on chromosome 14q for idiopathic basal ganglia
Eidelberg D, Moeller JR, Kazumata K et al. (1997). Meta- calcification (Fahr disease). Am J Hum Genet 65:
bolic correlates of pallidal neuronal activity in Parkinsons 764772.
disease. Brain 120: 13151324. Geser F, Prokop S, Glatzel M et al. (2006). The neuropathol-
Ekinci B, Apaydin H, Vural M et al. (2004). Two siblings ogy of McLeod syndrome: a case study. Mov Disord
with homocystinuria presenting with dystonia and parkin- 21 (Suppl 14): S357.
sonism. Mov Disord 19: 962964. Goker-Alpan O, Schiffmann R, LaMarca ME et al. (2004).
Eltahawy HA, Saint-Cyr J, Giladi N et al. (2004). Primary Parkinsonism among Gaucher disease carriers. J Med
dystonia is more responsive than secondary dystonia to Genet 41: 937940.
pallidal interventions: Outcome after pallidotomy or palli- Goldman JE, Katz D, Rapin I et al. (1981). Chronic GM1
dal deep brain stimulation. Neurosurgery 54: 613619. gangliosidosis presenting as dystonia: I. Clinical and
Elwes R, Saunders M (1986). Generalized dystonia, whisper- pathological features. Ann Neurol 9: 465475.
ing dysphonia and Wilsons disease in members of the same Goto S, Lee LV, Munoz EL et al. (2005). Functional anat-
family [letter]. J Neurol Neurosurg Psychiatry 49: 107. omy of the basal ganglia in X-linked recessive dystonia-
Evidente VG (2002). Zolpidem improves dystonia in parkinsonism. Ann Neurol 58: 717.
Lubag or X-linked dystonia-parkinsonism syndrome. Graybiel AM, Canales JJ, Capper-Loup C (2000). Levodopa-
Neurology 58: 662663. induced dyskinesias and dopamine-dependent stereoty-
Evidente VG, Gwinn KA, Caviness JN et al. (1998). Early pies: a new hypothesis. Trends Neurosci 23: S71S77.
cinematographic cases of postencephalitic parkinsonism Grotzsch H, Pizzolato GP, Ghika J et al. (2002). Neuro-
and other movement disorders [see comments]. Mov Dis- pathology of a case of dopa-responsive dystonia asso-
ord 13: 167169. ciated with a new genetic locus, DYT14. Neurology 58:
Evidente VG, Advincula J, Esteban R et al. (2002). Phenom- 18391842.
enology of Lubag or X-linked dystonia-parkinsonism. Hagberg B, Hambraeus L, Bensch K (1970). A case of
Mov Disord 17: 12711277. homocystinuria with a dystonia neurological syndrome.
Evidente VG, Nolte D, Niemann S et al. (2004). Phenotypic Neuropadiatrie 1: 337343.
and molecular analyses of X-linked dystonia-parkinsonism Hallett M (2003). Parkinson revisited: Pathophysiology of
(lubag) in women. Arch Neurol 61: 19561959. motor signs. Parkinsons disease Adv Neurol 91: 1928.
Fabbrini G, Brancati F, Vacca L et al. (2005). A novel family Hardie RJ, Young EP, Morgan-Hughes JA (1988). Hexosami-
with an unusual early-onset generalized dystonia. Mov nidase A deficiency presenting as juvenile progressive
Disord 20: 8186. dystonia [letter]. J Neurol Neurosurg Psychiatry 51:
Fahn S, Bressman SB, Marsden CD (1998). Classification of 446447.
dystonia. Adv Neurol 78: 110. Hardie RJ, Pullon HW, Harding AE et al. (1991). Neuroa-
Flores-Hernandez J, Cepeda C, Hernandez-Echeagaray canthocytosis. A clinical, haematological and pathological
E et al. (2002). Dopamine enhancement of NMDA currents study of 19 cases. Brain 114: 1349.
in dissociated medium-sized striatal neurons: role of D1 Hartig MB, Hortnagel K, Garavaglia B et al. (2006). Genoty-
receptors and DARPP-32. J Neurophysiol 88: 30103020. pic and phenotypic spectrum of PANK2 mutations in
526 R. H. WALKER

patients with neurodegeneration with brain iron accumula- Kaji R (2001). Basal ganglia as a sensory gating devise for
tion. Ann Neurol 59: 248256. motor control. J Med Invest 48: 142146.
Hayflick SJ, Westaway SK, Levinson B et al. (2003). Genet- Katchen M, Duvoisin RC (1986). Parkinsonism following
ic, clinical, and radiographic delineation of Hallervorden- dystonia in three patients. Mov Disord 1: 151157.
Spatz syndrome. N Engl J Med 348: 3340. Kawakami T, Takiyama Y, Sakoe K et al. (1999). A case of
Heiman GA, Ottman R, Saunders-Pullman RJ et al. McLeod syndrome with unusually severe myopathy. J
(2004). Increased risk for recurrent major depression in Neurol Sci 166: 3639.
DYT1 dystonia mutation carriers. Neurology 63: 631637. Kempster PA, Brenton DP, Gale AN et al. (1988). Dystonia
Ho M, Chelly J, Carter N et al. (1994). Isolation of the gene in homocystinuria. J Neurol Neurosurg Psychiatry 51:
for McLeod syndrome that encodes a novel membrane 859862.
transport protein. Cell 77: 869880. Keskin S, Yurdakul F (1996). Parkinsonian manifestations
Hoffmann GF, Assmann B, Brautigam C et al. (2003). Tyro- in a patient with homocystinuria. J Child Neurol 11:
sine hydroxylase deficiency causes progressive encephalo- 235236.
pathy and dopa-nonresponsive dystonia. Ann Neurol Khan NL, Graham E, Critchley P et al. (2003). Parkin dis-
54 (Suppl 6): S56S65. ease: a phenotypic study of a large case series. Brain
Holmes SE, OHearn E, Rosenblatt A et al. (2001). A repeat 126: 12791292.
expansion in the gene encoding junctophilin-3 is asso- Kitai ST, Kita H (1987). Anatomy and physiology of the
ciated with Huntington disease-like 2. Nat Genet 29: subthalamic nucleus: a driving force of the basal ganglia.
377378. In: MB Carpenter, A Jayaraman (Eds.), The Basal Ganglia
Houlden H, Lincoln S, Farrer M et al. (2003). Compound IIStructure and Function: Current Concepts, Advances
heterozygous PANK2 mutations confirm HARP and Hal- in Behavioral Biology. Plenum Press, New York, pp.
lervorden-Spatz syndromes are allelic. Neurology 61: 357373.
14231426. Kremer B, Weber B, Hayden MR (1992). New insights into
Hutton M, Lendon CL, Rizzu P et al. (1998). Association of the clinical features, pathogenesis and molecular genetics
missense and 50 -splice-site mutations in tau with the inher- of Huntington disease. Brain Pathol 2: 321335.
ited dementia FTDP-17. Nature 393: 702705. Kwak YT, Han IW, Baik J et al. (2000). Relation between cho-
Ichinose H, Suzuki T, Inagaki H et al. (1999). Molecular linesterase inhibitor and Pisa syndrome. Lancet 355: 2222.
genetics of dopa-responsive dystonia. Biol Chem 380: Le Ber I, Camuzat A, Castelnovo G et al. (2003). Prevalence
13551364. of dentatorubral-pallidoluysian atrophy in a large series of
Ikoma K, Samii A, Mercuri B et al. (1996). Abnormal cortical white patients with cerebellar ataxia. Arch Neurol 60:
motor excitability in dystonia. Neurology 46: 13711376. 10971099.
Inzelberg R, Korczyn AD (1994). Parkinsonism in adult- Lee LV, Pascasio FM, Fuentes FD et al. (1976). Torsion dys-
onset GM2 gangliosidosis. Mov Disord 9: 375377. tonia in Panay, Philippines. Adv Neurol 14: 137151.
Jahanshahi M, Jenkins IH, Brown RG et al. (1995). Self- Lenz FA, Jaeger CJ, Seike MS et al. (1999). Thalamic single
initiated versus externally triggered movements I. An neuron activity in patients with dystonia: Dystonia-related
investigation using measurement of regional cerebral activity and somatic sensory reorganization. J Neurophy-
blood flow with PET and movement-related potentials in siol 82: 23722392.
normal and Parkinsons disease subjects [see comments]. Levine IM, Yettra M, Stefanini M (1960). A hereditary
Brain 118: 913933. neurological disorder with acanthocytosis. Neurology 10:
Jenkins IH, Fernandez W, Playford ED et al. (1992). 425.
Impaired activation of the supplementary motor area in Levy LM, Hallett M (2002). Impaired brain GABA in focal
Parkinsons disease is reversed when akinesia is treated dystonia. Ann Neurol 51: 93101.
with apomorphine. Ann Neurol 32: 749757. LeWitt PA, Burns RS, Newman RP (1986). Dystonia in
Jung HH, Hergersberg M, Kneifel S et al. (2001a). McLeod untreated parkinsonism. Clin Neuropharmacol 9: 293297.
syndrome: a novel mutation, predominant psychiatric Lohmann E, Periquet M, Bonifati V et al. (2003). How much
manifestations, and distinct striatal imaging findings. phenotypic variation can be attributed to parkin genotype?
Ann Neurol 49: 384392. Ann Neurol 54: 176185.
Jung HH, Russo D, Redman C et al. (2001b). Kell and XK Low PA, Allsop JL (1973). Huntingtons choreathe rigid
immunohistochemistry in McLeod myopathy. Muscle form (Westphal variant) treated with L-DOPA: a case
Nerve 24: 13461351. report. Proc Aust Assoc Neurol 10: 4546.
Jung HH, Hergersberg M, Vogt M et al. (2003). McLeod Magalhaes AC, Caramelli P, Menezes JR et al. (1994). Wil-
phenotype associated with a XK missense mutation with- sons disease: MRI with clinical correlation. Neuroradiol-
out hematologic, neuromuscular, or cerebral involvement. ogy 36: 97100.
Transfusion 43: 928938. Magnuson TM, Roccaforte WH, Wengel SP et al. (2000).
Juranyi Z, Zigmond MJ, Harsing LG Jr (2003). [3H] Dopa- Medication-induced dystonias in nine patients with
mine release in striatum in response to cortical stimulation dementia. J Neuropsychiatry Clin Neurosci 12: 219225.
in a corticostriatal slice preparation. J Neurosci Methods Malandrini A, Cesaretti S, Mulinari M et al. (1996). Acantho-
126: 5767. cytosis, retinitis pigmentosa, pallidal degeneration. Report
PARKINSONISM AND DYSTONIA 527

of two cases without serum lipid abnormalities. J Neurol Nolte D, Niemann S, Muller U (2003). Specific sequence
Sci 140: 129131. changes in multiple transcript system DYT3 are associated
Margolis RL, OHearn E, Rosenblatt A et al. (2001). A dis- with X-linked dystonia parkinsonism. Proc Natl Acad Sci
order similar to Huntingtons disease is associated with a USA 100: 1034710352.
novel CAG repeat expansion. Ann Neurol 50: 373380. Nygaard TG, Wilhelmsen KC, Risch NJ et al. (1993). Link-
Margolis RL, Holmes SE, Rosenblatt A et al. (2004). Hun- age mapping of dopa-responsive dystonia (DRD) to chro-
tingtons disease-like 2 (HDL2) in North America and mosome 14q. Nat Genet 5: 386391.
Japan. Ann Neurol 56: 670674. Nygaard TG, Waran SP, Levine RA et al. (1994). Dopa-
Martins S, Matama T, Guimaraes L et al. (2003). Portuguese responsive dystonia simulating cerebral palsy. Pediatr
families with dentatorubropallidoluysian atrophy (DRPLA) Neurol 11: 236240.
share a common haplotype of Asian origin. Eur J Hum Oide T, Ohara S, Yazawa M et al. (2002). Progressive supra-
Genet 11: 808811. nuclear palsy with asymmetric tau pathology presenting
Meek D, Wolfe LS, Andermann E et al. (1984). Juvenile with unilateral limb dystonia. Acta Neuropathol (Berl)
progressive dystonia: a new phenotype of GM2 gangliosi- 104: 209214.
dosis. Ann Neurol 15: 348352. Oliveira JR, Spiteri E, Sobrido MJ et al. (2004). Genetic het-
Meunier S, Garnero L, Ducorps A et al. (2001). Human erogeneity in familial idiopathic basal ganglia calcifica-
brain mapping in dystonia reveals both endophenotypic tion (Fahr disease). Neurology 63: 21652167.
traits and adaptive reorganization. Ann Neurol 50: Orrell RW, Amrolia PJ, Heald A et al. (1995). Acantho-
521527. cytosis, retinitis pigmentosa, and pallidal degeneration:
Mink JW (1996). The basal ganglia: focused selection and a report of three patients, including the second reported
inhibition of competing motor programs. Prog Neurobiol case with hypoprebetalipoproteinemia (HARP syndrome).
50: 381425. Neurology 45: 487492.
Mink JW (2003). The basal ganglia and involuntary move- Owens WE, Okun MS (2004). Dystonia, tremor, and par-
mentsimpaired inhibition of competing motor patterns. kinsonism in a 54 year old man with 2-hydroxyglutaric
Arch Neurol 60: 13651368. aciduria. J Neurol Neurosurg Psychiatry 75: 13621363.
Mir P, Edwards MJ, Curtis AR et al. (2004). Adult-onset Oyanagi K, Takeda S, Takahashi H et al. (1989). A quantita-
generalized dystonia due to a mutation in the neuroferriti- tive investigation of the substantia nigra in Huntingtons
nopathy gene. Mov Disord 20: 243245. disease. Ann Neurol 26: 1319.
Miyajima H (2003). Aceruloplasminemia, an iron metabolic Ozand PT, Gascon GG, Al Essa M et al. (1998). Biotin-
disorder. Neuropathology 23: 345350. responsive basal ganglia disease: A novel entity. Brain
Miyaoka T, Seno H, Yamamori C et al. (2001). Pisa syn- 121: 12671279.
drome due to a cholinesterase inhibitor (donepezil): a case Page RA, Davie CA, Macmanus D et al. (2004). Clinical cor-
report. J Clin Psychiatry 62: 573574. relation of brain MRI and MRS abnormalities in patients
Mole SE (2004). The genetic spectrum of human neuronal with Wilson disease. Neurology 63: 638643.
ceroid-lipofuscinoses. Brain Pathol 14: 7076. Partridge JG, Apparsundaram S, Gerhardt GA et al. (2002).
Moro E, Lang AE, Strafella AP et al. (2004). Bilateral globus Nicotinic acetylcholine receptors interact with dopamine
pallidus stimulation for Huntingtons disease. Ann Neurol in induction of striatal long-term depression. J Neurosci
56: 290294. 22: 25412549.
Morrison PJ, Patterson VH (1992). Cranial dystonia (Meige Penney JB, Young AB, Shoulson I et al. (1990). Huntingtons
syndrome) in postencephalitic parkinsonism. Mov Disord disease in Venezuela: 7 years of follow-up on sympto-
7: 9091. matic and asymptomatic individuals. Mov Disord 5:
Murgod UA, Muthane UB, Ravi V et al. (2001). Persistent 9399.
movement disorders following Japanese encephalitis. Neu- Perlmutter JS, Tempel LW, Black KJ et al. (1997). MPTP
rology 57: 23132315. induces dystonia and parkinsonismclues to the patho-
Muthane U, Chickabasaviah Y, Kaneski C et al. (2004). Clin- physiology of dystonia. Neurology 49: 14321438.
ical features of adult GM1 gangliosidosis: Report of three Perlmutter JS, Stambuk MK, Markham J et al. (1998).
Indian patients and review of 40 cases. Mov Disord 19: Decreased [18F]spiperone binding in putamen in dystonia.
13341341. Adv Neurol 78: 161168.
Nardocci N, Bertagnolio B, Rumi V et al. (1992). Progressive Pittock SJ, Joyce C, OKeane V et al. (2000). Rapid-onset
dystonia symptomatic of juvenile GM2 gangliosidosis [see dystonia-parkinsonismA clinical and genetic analysis
comments]. Mov Disord 7: 6467. of a new kindred. Neurology 55: 991995.
Nicholl DJ, Sutton I, Dotti MT et al. (2004). White matter Playford ED, Jenkins IH, Passingham RE et al. (1992).
abnormalities on MRI in neuroacanthocytosis. J Neurol Impaired mesial frontal and putamen activation in Parkin-
Neurosurg Psychiatry 75: 12001201. sons disease: A positron emission tomography study. Ann
Nijssen PC, Brusse E, Leyten AC et al. (2002). Autosomal Neurol 32: 151161.
dominant adult neuronal ceroid lipofuscinosis: Parkin- Poewe WH, Lees AJ, Stern GM (1988). Dystonia in Parkin-
sonism due to both striatal and nigral dysfunction. Mov sons disease: Clinical and pharmacological features. Ann
Disord 17: 482487. Neurol 23: 7378.
528 R. H. WALKER

Polymeropoulos MH, Lavedan C, Leroy E et al. (1997). responses to dopaminergic stimulation. Proc Natl Acad
Mutation in the alpha-synuclein gene identified in families Sci USA 99: 90049009.
with Parkinsons disease. Science 276: 20452047. Segawa M, Nomura Y (1993). Hereditary progressive dysto-
Quartarone A, Bagnato S, Rizzo V et al. (2003). Abnormal nia with marked diurnal fluctuation. Pathophysiological
associative plasticity of the human motor cortex in wri- importance of the age of onset. Adv Neurol 60: 568576.
ters cramp. Brain 126: 25862596. Sener RN (2003). Diffusion MR imaging changes associated
Quinn N, Critchley P, Marsden CD (1987). Young onset with Wilson disease. AJNR Am J Neuroradiol 24: 965967.
Parkinsons disease. Mov Disord 2: 7391. Shan DE, Soong BW, Sun CM et al. (2001). Spinocerebellar
Rampoldi L, Danek A, Monaco AP (2002). Clinical features ataxia type 2 presenting as familial levodopa-responsive
and molecular bases of neuroacanthocytosis. J Mol Med parkinsonism. Ann Neurol 50: 812815.
80: 475491. Shashidharan P, Sandu D, Walker RH (2002). Expression of
Reske-Nielsen E, Jensen PK, Hein-Sorensen O et al. (1988). mutant torsinA in transgenic mice interferes with dop-
Calcification of the central nervous system in a new her- aminergic system. Soc Neurosci Abstr. Abstract/Itinerary
editary neurological syndrome. Acta Neuropathol (Berl) Planner CD-ROM: 523.11.
75: 590596. Shashidharan P, Sandu D, Potla U et al. (2005). Transgenic
Reuter I, Hu MT, Andrews TC et al. (2000). Late onset levo- mouse model of early-onset DYT1 dystonia. Hum Mol
dopa responsive Huntingtons disease with minimal Genet 14: 125133.
chorea masquerading as Parkinson plus syndrome. J Neu- Silberstein P, Kuhn AA, Kupsch A et al. (2003). Patterning
rol Neurosurg Psychiatry 68: 238241. of globus pallidus local field potentials differs between
Rice ME, Cragg SJ (2004). Nicotine amplifies reward-related Parkinsons disease and dystonia. Brain 126: 25972608.
dopamine signals in striatum. Nat Neurosci 7: 583584. Simonati A, Santorum E, Tessa A et al. (2000). A CLN2 gene
Ridding MC, Sheean G, Rothwell JC et al. (1995). Changes nonsense mutation is associated with severe caudate atro-
in the balance between motor cortical excitation and inhi- phy and dystonia in LINCL. Neuropediatrics 31: 199201.
bition in focal, task specific dystonia. J Neurol Neurosurg Singleton A (2004). What does PINK1 mean for Parkinson
Psychiatry 59: 493498. diseases? Neurology 63: 13501351.
Rinne JO, Daniel SE, Scaravilli F et al. (1994). Nigral degen- Singleton A, Hague S, Hernandez D (2004). X-linked reces-
eration in neuroacanthocytosis. Neurology 44: 16291632. sive dystonia parkinsonism (XDP; Lubag; DYT3). Adv
Rinne JO, Iivanainen M, Metsahonkala L et al. (2004). Stri- Neurol 94: 139142.
atal dopaminergic system in dopa-responsive dystonia: a Smith Y, Bennett BD, Bolam JP et al. (1994). Synaptic rela-
multi-tracer PET study shows increased D2 receptors. J tionships between dopaminergic afferents and cortical or
Neural Transm 111: 5967. thalamic input in the sensorimotor territory of the striatum
Risvoll H, Kerty E (2001). To test or not? The value of diag- in monkey. J Comp Neurol 344: 119.
nostic tests in cervical dystonia. Mov Disord 16: 286289. Sohn YH, Hallett M (2004). Disturbed surround inhibition in
Rodriguez MC, Guridi OJ, Alvarez L et al. (1998). The sub- focal hand dystonia. Ann Neurol 56: 595599.
thalamic nucleus and tremor in Parkinsons disease. Mov Specola N, Vanier MT, Goutieres F et al. (1990). The juve-
Disord 13: 111118. nile and chronic forms of GM2 gangliosidosis: clinical
Rostasy K, Augood SJ, Hewett JW et al. (2003). TorsinA and enzymatic heterogeneity. Neurology 40: 145150.
protein and neuropathology in early onset generalized dys- Stevanin G, Camuzat A, Holmes SE et al. (2002). CAG/CTG
tonia with GAG deletion. Neurobiol Dis 12: 1124. repeat expansions at the Huntingtons disease-like 2 locus
Rovito AD, Pirone FJ (1963). Acanthrocytosis associated are rare in Huntingtons disease patients. Neurology 58:
with schizophrenia. Am J Psychiatry 120: 182185. 965967.
Rubio JP, Levy ER, Dobson-Stone C et al. (1999). Genomic Stevanin G, Fujigasaki H, Lebre AS et al. (2003). Hunting-
organization of the human galpha14 and Galphaq genes tons disease-like phenotype due to trinucleotide repeat
and mutation analysis in chorea-acanthocytosis (CHAC). expansions in the TBP and JPH3 genes. Brain 126:
Genomics 57: 8493. 15991603.
Russo D, Redman C, Lee S (1998). Association of XK and Symmans WA, Shepherd CS, Marsh WL et al. (1979).
Kell blood group proteins. J Biol Chem 273: 1395013956. Hereditary acanthocytosis associated with the McLeod
Russo D, Wu X, Redman CM et al. (2000). Expression of phenotype of the Kell blood group system. Br J Haematol
Kell blood group protein in nonerythroid tissues. Blood 42: 575583.
96: 340346. Takahashi-Fujigasaki J (2003). Neuronal intranuclear hya-
Saiki S, Sakai K, Kitagawa Y et al. (2003). Mutation in the line inclusion disease. Neuropathology 23: 351359.
CHAC gene in a family of autosomal dominant chorea- Takashima H, Sakai T, Iwashita H et al. (1994). A family of
acanthocytosis. Neurology 61: 16141616. McLeod syndrome, masquerading as chorea-acanthocyto-
Saiki S, Hirose G, Sakai K et al. (2004). Chorea-acanthocyto- sis. J Neurol Sci 124: 5660.
sis associated with Tourettism. Mov Disord 19: 833836. Tan EK, Chan LL, Wong MC (2003a). Levodopa-induced
Saka E, Iadarola M, Fitzgerald DJ et al. (2002). Local circuit oromandibular dystonia in progressive supranuclear palsy.
neurons in the striatum regulate neural and behavioral Clin Neurol Neurosurg 105: 132134.
PARKINSONISM AND DYSTONIA 529

Tan LC, Tanner CM, Chen R et al. (2003b). Marked variation polyglutamine-containing neuronal inclusions. Neurology
in clinical presentation and age of onset in a family with a 58: 10311037.
heterozygous parkin mutation. Mov Disord 18: 758763. Walker RH, Jankovic J, OHearn E et al. (2003a). Phenoty-
Taniguchi S, McDonagh AM, Pickering-Brown SM et al. pic features of Huntington disease-like 2. Mov Disord 18:
(2004). The neuropathology of frontotemporal lobar 15271530.
degeneration with respect to the cytological and biochem- Walker RH, Rasmussen A, Rudnicki D et al. (2003b). Hun-
ical characteristics of tau protein. Neuropathol Appl Neu- tingtons disease-like 2 can present as chorea-acanthocyto-
robiol 30: 118. sis. Neurology 61: 10021004.
Tassin J, Durr A, Bonnet AM et al. (2000). Levodopa- Walker RH, Danek A, Jung HH et al. (2005). McLeod syndrome
responsive dystonia. GTP cyclohydrolase I or parkin mut- presenting with hepatic disease. Mov Disord 20: S6S7.
ations? Brain 123: 11121121. Walker RH, Danek A, Uttner I et al. McLeod phenotype
Tayebi N, Walker J, Stubblefield B et al. (2003). Gaucher without the McLeod syndrome. Transfusion (in press).
disease with parkinsonian manifestations: does glucocer- Waters CH, Faust PL, Powers J et al. (1993a). Neuropath-
ebrosidase deficiency contribute to a vulnerability to par- ology of lubag (X-Linked dystonia parkinsonism). Mov
kinsonism? Mol Genet Metab 79: 104109. Disord 8: 387390.
Toda H, Hamani C, Lozano A (2004). Deep brain stimula- Waters CH, Takahashi H, Wilhelmsen KC et al. (1993b).
tion in the treatment of dyskinesia and dystonia. Neuro- Phenotypic expression of X-linked dystonia-parkinsonism
surg Focus 17: E2. (Lubag) in two women. Neurology 43: 15551558.
Trottenberg T, Volkmann J, Deuschl G et al. (2005). Treat- Wichmann T, Bergman H, DeLong MR (1994). The primate
ment of severe tardive dystonia with pallidal deep brain subthalamic nucleus. III. Changes in motor behavior and
stimulation. Neurology 64: 344346. neuronal activity in the internal pallidum induced by sub-
Ueno S, Maruki Y, Nakamura M et al. (2001). The gene thalamic inactivation in the MPTP model of parkinsonism.
encoding a newly discovered protein, chorein, is mutated J Neurophysiol 72: 521530.
in chorea-acanthocytosis. Nat Genet 28: 121122. Wihl G, Volkmann J, Allert N et al. (2001). Deep brain stimula-
Ueyama H, Kumamoto T, Nagao S et al. (2000). A novel tion of the internal pallidum did not improve chorea in a
mutation of the McLeod syndrome gene in a Japanese patient with neuro-acanthocytosis. Mov Disord 16: 572575.
family. J Neurol Sci 176: 151154. Wills AJ, Sawle GV, Guilbert PR et al. (2002). Palatal tre-
Umemura A, Jaggi JL, Dolinskas CA et al. (2004). Pallidal mor and cognitive decline in neuroferritinopathy. J Neurol
deep brain stimulation for longstanding severe generalized Neurosurg Psychiatry 73: 9192.
dystonia in Hallervorden-Spatz syndrome. Case report. J Wohrle JC, Weigel R, Grips E et al. (2003). Risperidone-
Neurosurg 100: 706709. responsive segmental dystonia and pallidal deep brain sti-
Uyama E, Terasaki T, Watanabe S et al. (1992). Type-3 mulation. Neurology 61: 546548.
GM1 gangliosidosis-characteristic MRI findings corre- Xu X, Pin S, Gathinji M et al. (2004). Aceruloplasminemia:
lated with dystonia. Acta Neurol Scand 86: 609615. an inherited neurodegenerative disease with impairment of
Valente EM, Abou-Sleiman PM, Caputo V et al. (2004). Her- iron homeostasis. Ann NY Acad Sci 1012: 299305.
editary early-onset Parkinsons disease caused by muta- Yoshida K, Ikeda S, Kawaguchi K et al. (1994). Adult GM1
tions in PINK1. Science 304: 11581160. gangliosidosis: immunohistochemical and ultrastructural
Velayos-Baeza A, Vettori A, Copley RR et al. (2004). Analy- findings in an autopsy case. Neurology 44: 23762382.
sis of the human VPS13 gene family. Genomics 84: Yoshioka A, Saiki S, Yamaya Y et al. (1999). [A 54-year-old
536549. man with action myoclonus, parkinsonism and epilepsy].
Vidailhet M, Vercueil L, Houeto JL et al. (2005). Bilateral No To Shinkei Brain Nerve 51: 9991007.
deep-brain stimulation of the globus pallidus in primary Younes-Mhenni S, Thobois S, Streichenberger N et al.
generalized dystonia. N Engl J Med 352: 459467. (2002). [Mitochondrial encephalomyopathy, lactic acido-
Vitek JL, Zhang J, Evatt M et al. (1998). GPi pallidotomy for sis and stroke-like episodes (Melas) associated with a Fahr
dystonia: Clinical outcome and neuronal activity. Adv disease and cerebellar calcifications]. Rev Med Interne 23:
Neurol 78: 211219. 10271029.
Vitek JL, Chockkan V, Zhang JY et al. (1999). Neuronal Zhou B, Westaway SK, Levinson B et al. (2001). A novel
activity in the basal ganglia in patients with generalized pantothenate kinase gene (PANK2) is defective in Haller-
dystonia and hemiballismus. Ann Neurol 46: 2235. vorden-Spatz syndrome. Nat Genet 28: 345349.
Walker RH, Danisi FO, Swope DM et al. (2000). Intrathecal Zimprich A, Muller-Myhsok B, Farrer M et al. (2004). The
baclofen for dystonia: Benefits and complications during PARK8 locus in autosomal dominant parkinsonism:
six years experience. Mov Disord 15: 12421247. confirmation of linkage and further delineation of the
Walker RH, Friedman J, Wiener J et al. (2002a). A family disease-containing interval. Am J Hum Genet 74: 1119.
with a tau P301L mutation presenting with parkinsonism. Zuhlke C, Gehlken U, Hellenbroich Y et al. ((2003)). Pheno-
Parkinsonism Relat Disord 9: 121123. typical variability of expanded alleles in the TATA-bind-
Walker RH, Morgello S, Davidoff-Feldman B et al. ing protein gene. Reduced penetrance in SCA17?
(2002b). Autosomal dominant chorea-acanthocytosis with J Neurol 250: 161163.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 60

Dementia with Lewy bodies

IAN MCKEITH*

Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK

60.1. Introduction 75 years in most studies) than for Parkinsons disease


(PD).
Dementia with Lewy bodies (DLB) is the preferred
term (McKeith et al., 1996) for a clinicopathological 60.1.2. Dementia with Lewy bodies and dementia
syndrome that has been variously labeled over the last in Parkinsons disease
40 years as diffuse Lewy body disease (DLBD)
(Kosaka et al., 1984; Dickson et al., 1987; Lennox At autopsy DLB is neuropathologically indistinguish-
et al., 1989a), dementia associated with cortical Lewy able from dementia occurring late in the course of
bodies (DCLB) (Byrne et al., 1991), the Lewy body PD (PDD). This has led to a continuing debate about
variant of Alzheimers disease (LBVAD) (Hansen the relationship between these two clinically defined
et al., 1990; Forstl et al., 1993), senile dementia of syndromes. Most clinicians find it helpful to make a
Lewy body type (SDLT) (Perry et al., 1989a, b, 1990) distinction between DLB and PDD based on the tem-
and Lewy body dementia (LBD) (Gibb et al., 1987). poral sequence in which symptoms appear. This
Initially regarded as uncommon, DLB is now thought approach is (understandably) not generally favored
to account for up to 20% of all elderly cases of dementia by some contemporary neuroscience researchers, who
reaching autopsy (Jellinger, 1996), with a clinical pre- regard the different clinical presentations as simply
sentation primarily characterized by cognitive decline representing different points on a common spectrum
leading to dementia, accompanied in the majority of of LB disease with shared abnormalities in a-synuclein
cases (75%) by extrapyramidal motor features and metabolism. This unified approach to classification is
additional neuropsychiatric features. probably preferable for molecular and genetic studies
and for developing therapeutic agents.
60.1.1. Prevalence and incidence The extent to which PD, PDD and DLB are similar
remains to be investigated in further depth. For exam-
There is very little systematically collected informa- ple, a proposal to stage LB pathology in the brain has
tion about the prevalence, incidence and associated relatively recently been proposed for PD (Braak et al.,
risk factors for DLB (McKeith et al., 2004a). A com- 2003). This postulates a progressive spread from the
munity study of 85-year-olds in Finland found that medulla oblongata and olfactory bulb (presymptomatic
5.0% met clinical diagnostic criteria for DLB, repre- stages 1 and 2) through the substantia nigra and other
senting 22% of all demented cases (Rahkonen et al., midbrain nuclei and basal forebrain (symptomatic
2003). This is similar to other clinical estimates (Shergill stages 3 and 4), eventually encroaching upon the tele-
et al., 1994; Stevens et al., 2002) and consistent with ncephalic cortex in end-stages 5 and 6. It has been sug-
estimates of Lewy body (LB) prevalence (15%) in a gested that in DLB the burden of pathology is
dementia case register followed to autopsy (Holmes differently distributed, with greater emphasis on the
et al., 1999). Little is known about risk factors for cortex (Kosaka et al., 1996). The validity of the Braak
LB disease except for male sex and age of onset, staging scheme and its relevance to DLB remain to be
which is on average 10 years greater for DLB (mean determined. The remainder of the chapter will focus on

*Correspondence to: Professor Ian McKeith, Institute for Ageing and Health, Newcastle General Hospital, Westgate Road,
Newcastle upon Tyne NE4 6BE, UK. E-mail: i.g.mckeith@ncl.ac.uk, Tel: 44-(0)191-256-3018, Fax: 44-(0)191-219-5071.
532 I. MCKEITH
clinical, pathological and management aspects of although reports of a period of increased confusion or
DLB. PDD is covered in detail in Chapter 18. prominent hallucinations may give the impression of a
sudden onset. In the first instance the patient may be
60.1.3. Clinical significance of dementia with Lewy considered to have a delirium rather than a dementia
bodies syndrome. The course is almost invariably progressive,
with cognitive function scores declining about 10% per
Until the last decade DLB was not widely recognized as annum. In one study over 1 year, subjects with DLB,
a common form of dementia in older people and the AD and vascular dementia had similar rates of overall
majority of cases were probably incorrectly diagnosed cognitive decline, approximately 45 points on Mini-
in the clinic as Alzheimers disease (AD) or vascular Mental State Examination (MMSE) and 1214 points
dementia (McKeith et al., 1994). Although case reports on Cambridge Cognitive Examination (CAMCOG)
describing dementia in association with LB pathology (Ballard et al., 2001). Survival times from onset until
had been published intermittently during the previous death are also similar to AD (Walker et al., 2000a),
30 years (Okazaki et al., 1961; Kosaka et al., 1984), it although a minority of DLB patients have a very rapid
was the advent of antiubiquitin immunocytochemical disease course (Armstrong et al., 1991; Lopez et al.,
staining (Lennox et al., 1989a, b) that prompted wide- 2000; Collerton et al., 2003).
spread recognition that LB pathology was common in
patients with dementia. It was only after this that oper- 60.2.1. Cognition
ationalized criteria for DLB started to be developed.
The history of these developments has been reviewed The profile of neuropsychological impairments in
elsewhere (McKeith, 2005a) and a Consortium on patients with DLB differs from that of AD and other
DLB was established in 1985 to set internationally dementia syndromes (Collerton et al., 2003), reflecting
agreed standards for clinical and pathological diagnosis the combined involvement of cortical and subcortical
(McKeith et al., 2005). The importance of recognizing pathways and relative sparing of the hippocampus. Epi-
DLB relates particularly to its pharmacological man- sodic recall and recognition (Calderon et al., 2001; Col-
agement, with reports of good responsiveness to choli- lerton et al., 2003) can be relatively preserved in the
nesterase inhibitors (ChEIs) (McKeith et al., 2000; early stages, in contrast to AD, in which memory failure
Aarsland et al., 2004), extreme sensitivity to the side- is often the presenting complaint. Patients with DLB per-
effects of neuroleptics (McKeith et al., 1992a; Ballard form better than those with AD on tests of verbal mem-
et al., 1998a) and limited responsiveness to levodopa ory (McKeith et al., 1992b) but worse on visuospatial
(Bonelli et al., 2004; Molloy et al., 2005). performance tasks (Walker et al., 1997) and tests of
attention (Sahgal et al., 1992). This profile may be harder
60.2. Clinical features to recognize later in the disease when global cognitive
difficulties obscure the picture. It has been suggested that
The central characteristic of DLB is a progressive this double discrimination can help differentiate DLB
dementia with marked impairments in visuoperceptual, from AD, with relative preservation of confrontation
attentional and executive functions reflecting a combi- naming, short- and medium-term recall as well as recog-
nation of cortical and subcortical damage. The clinical nition, and greater impairment on verbal fluency, visual
picture does not always conform to the classic descrip- perception and performance tasks (Walker et al., 1997;
tion of a dementia syndrome, e.g. as described in Diag- Connor et al., 1998; Ferman et al., 1999; Collerton
nostic and Statistical Manual of Mental Disorders et al., 2003; Mormont et al., 2003). Despite these selec-
(DSM-IVR), which requires a combination of decline tive patterns of impairment, composite global cognitive
in memory and at least one other cognitive function, in assessment tools such as MMSE cannot be relied upon
the context of unimpaired consciousness. In DLB distur- to distinguish DLB from other common dementia syn-
bances in the patients level of alertness and attention are dromes (Ala et al., 2004) and detailed neuropsychologi-
usually apparent and often severe, and the cognitive def- cal assessment is often required. Computer-based test
icits do not always include severe memory loss. Fluctuat- systems may be particularly useful to capture elements
ing cognition, recurrent visual hallucinations (VHs) and of attentional (Wesnes et al., 2002) and visuoperceptual
extrapyramidal motor symptoms are the core features performance (Mosimann et al., 2004) in DLB.
by which DLB is recognized clinically (McKeith et al.,
1996), although these features are now known to be 60.2.2. Cognitive fluctuation
absent in a significant minority of cases, particularly
those with significant additional Alzheimer pathology Fluctuations in cognitive function, which may vary
(Merdes et al., 2003). The onset tends to be insidious, over minutes, hours or days, occur in 5075% of
DEMENTIA WITH LEWY BODIES 533
patients and are associated with shifting levels of having the highest of all (Collerton et al., 2003). A
attention and alertness. Cognitive fluctuations may novel perception and attention deficit model for recur-
contribute to large variability in repeated test scores, rent complex VH suggests that a combination of
e.g. 5 MMSE points difference over the course of a impaired attentional binding and poor sensory activa-
few days or weeks (Mega et al., 1996), making it dif- tion of a correct proto-object, in conjunction with a
ficult to be sure of the severity of cognitive impair- relatively intact scene representation, bias perception
ment by a single examination. The assessment of to allow the intrusion of a hallucinatory proto-object
fluctuating cognitive impairment poses considerable into a scene perception. Incorporation of this image
difficulty to many clinicians and has been repeatedly into a context-specific hallucinatory scene representa-
cited as a reason for low clinical ascertainment of tion accounts for repetitive hallucinations (Collerton
DLB (Mega et al., 1996; Litvan et al., 1998). Newly et al., 2005). These impairments, which are under-
proposed care-giver and observer-rated scales may be pinned by disturbances in a lateral frontal cortex-
particularly helpful in this regard (Walker et al., ventral visual stream system, are consistent with the
2000b). Questions such as are there episodes when known distribution of neurochemical and pathological
his/her thinking seems quite clear and then becomes deficits of DLB, as discussed later in this chapter.
muddled? were originally thought to be useful probes
(Ballard et al., 1993), although two recent studies 60.2.4. Other neuropsychiatric features
found most carers responded positively to such ques-
tions regardless of diagnostic subtype (Bradshaw Neuropsychiatric features other than VH are common
et al., 2004; Ferman et al., 2004). These investigators in DLB (Del-Ser et al., 2000), but none is quite so
also established that more reliable predictors of DLB characteristic and most are also equally prevalent in
diagnosis are objective questions about daytime sleepi- other dementing disorders. Auditory hallucinations
ness, episodes of staring blankly or incoherent speech occur in about 20% of DLB cases and, together with
and qualitative assessment of the range of fluctuation, olfactory and tactile hallucinations, may lead to initial
e.g. best versus worst. Recording variation in atten- diagnoses of late-onset psychosis (Birkett et al., 1992)
tional performance using a computer-based test system or temporal lobe epilepsy (McKeith et al., 1992b), the
offers an independent method of measuring fluctuation latter particularly when they are associated with appar-
which is also sensitive to drug treatment effects ent disturbances of consciousness. Delusions are com-
(Wesnes et al., 2002). mon in DLB, occurring in over half and usually being
based on recollections of hallucinations and perceptual
60.2.3. Recurrent visual hallucinations disturbances. They consequently have a fixed, com-
plex and bizarre content that contrasts with the mun-
Recurrent VHs are the most characteristic neuropsy- dane and often poorly formed persecutory ideas
chiatric feature of DLB, occurring in up to 80% of encountered in AD patients that are usually based
cases, and they are often the symptom which first upon forgetfulness and confabulation. The assessment
alerts the clinician to a DLB diagnosis. Their presence of depression, which is also frequently encountered
early in the course of illness (Ballard et al., 1999) and in DLB (McKeith et al., 1992b), is complicated not
their persistence throughout (McShane et al., 1998) only by apathy and attentional deficits, but by extra-
help to distinguish them from the transient perceptual pyramidal motor dysfunction, including facial and
disturbances that may occasionally occur in dementias body bradykinesia (Klatka et al., 1996). Apathy is
of other etiology or during delirium. Well-formed, another common feature of DLB and may mimic
detailed and animate figures are experienced, provok- depression or excessive daytime somnolence. Anxiety,
ing emotional responses varying through fear, amuse- agitation and behavioral disturbances are frequently
ment or indifference, usually with some insight into secondary to fluctuating confusion and hallucinations
the unreality of the episode once it is over. Why VH and may improve when these are treated.
and other visual symptoms such as illusions and misi-
dentifications are so common in DLB (Ballard et al., 60.2.5. Sleep disorders
1999; Mosimann et al., 2006) is not known but it
seems probable that they are closely related to the pro- Sleep disorders have recently been recognized as com-
nounced visuoperceptual and attentional deficits. mon in DLB, with daytime somnolence and nocturnal
Meta-analysis of neuropschological studies in different restlessness (Boeve et al., 1998; Grace et al., 1998)
neurogenerative disorders reveals a significant and sometimes as a prodromal feature. Rapid-eye move-
positive association between the severity of visuoper- ment (REM) sleepwakefulness dissociations may
ceptual impairments and the frequency of VH, DLB explain several features of DLB that are characteristic
534 I. MCKEITH
of narcolepsy (REM sleep behavior disorder (RBD), repeated falls and syncope and the transient losses of
daytime hypersomnolence, VHs and cataplexy) consciousness that are seen in some DLB patients
(Boeve et al., 2001). RBD is manifested by vivid and (Ballard et al., 1998b).
often frightening dreams during REM sleep, but with-
out the normal muscle atonia typical of REM sleep. 60.3. Clinical diagnostic criteria for dementia
Patients therefore appear to act out their dreams, with Lewy bodies
vocalizing, flailing limbs and moving around the bed,
sometimes violently. Vivid visual images are often Following the recognition of LB pathology in neuro-
reported, although the patient may have little recall pathological autopsy series of elderly patients with
of these episodes. The history is obtained from the dementia, several preliminary attempts were made to
bed-partner who may report many years of this sleep describe the clinical course and characteristics by which
disorder prior to the onset of dementia and parkinson- these patients could be identified antemortem (Okazaki
ism (Boeve et al., 2004). RBD is frequently associated et al., 1961; Kosaka, 1978; Kosaka et al., 1984; Byrne
with LB disease and only rarely with other neurode- et al., 1989; Gibb et al., 1989; Crystal et al., 1990; Han-
generative disorders (Boeve et al., 2003). Sleep disor- sen et al., 1990; Perry et al., 1990; Forstl and Levy,
ders may contribute to the fluctuations typical of DLB 1991; Kuzuhara and Yoshimura, 1993).
and their treatment may improve fluctuations and
quality of life (Boeve et al., 2003). 60.3.1. Early diagnostic criteria

60.2.6. Motor parkinsonism Based upon these observations, attempts were made to
describe the typical course of illness (Burkhardt et al.,
Extrapyramidal signs (EPS) are reported in 2550% of 1988; Crystal et al., 1990; McKeith et al., 1992b) and the
DLB cases at diagnosis and 7580% develop some first formal clinical diagnostic criteria were proposed.
EPS during the natural course. This means that up to The Nottingham (UK) group based their proposed
25% of patients with pathologically confirmed LB dis- system upon the clinical characteristics of 15 personal
ease will have no clinical history of parkinsonism and cases, the largest individual series published at that time
clinicians must be prepared to consider and diagnose (Byrne et al., 1989). Seven were men, the mean age at
DLB in its absence. Originally said to be mild and onset was 72 years and the mean duration of illness
to appear late in the clinical course, the profile of was 5.5 years. A total of 40% presented with symptoms
EPS in DLB is now generally thought to be similar and signs of idiopathic PD, with cognitive impairment
to that in age-matched non-demented PD patients with occurring 14 years later. A further 20% had parkinson-
regard to overall severity (Aarsland et al., 2001b). ism and mild cognitive impairment at presentation and
There is however greater symmetry and axial ten- the remaining 40% showed motor features later in their
dency, postural instability and facial impassivity but illnesses, gait disturbance and postural abnormalities
less tremor (Burn et al., 2003). The rate of motor dete- being most common. These latter cases presented with
rioration on the Unified Parkinsons Disease Rating neuropsychiatric features only, in various combinations
Scale (UPDRS) (Fahn and Elton, 1987) is about 10% of cognitive impairment, paranoid delusions and visual
per annum, similar to PD (Ballard et al., 2000a). Levo- or auditory hallucinations. Fourteen of the 15 were
dopa-responsiveness is reduced in DLB compared with demented before death, the exception presenting with
PD (Bonelli et al., 2004; Molloy et al., 2005), possibly classic PD and later becoming depressed, irritable and
due to additional intrinsic striatal pathology and dys- mildly forgetful with frequent falls. Fluctuating cogni-
function (Duda et al., 2002) and the fact that some tion with episodic confusion, for which no adequate
of the parkinsonian features are non-dopaminergic in underlying cause could be found, was observed in 80%
origin (Burn and McKeith, 2004). of the Nottingham cases. Byrne et al. (1991) also drew
attention to the frequent occurrence of depression
60.2.7. Autonomic dysfunction (20%) and psychosis (33%). This led to the first formal
proposal of operational criteria for DCLB.
Severe autonomic dysfunction may occur early in the The presence of extrapyramidal features was man-
clinical course, producing orthostatic hypotension, datory, although these could be mild and occur late
neurocardiovascular instability, urinary incontinence, in the course of the illness. Since at least 25% of
constipation and impotence as well as eating and swal- DLB cases reported in the whole literature (Kosaka
lowing difficulties (Kuzuhara and Yoshimura, 1993; and Iseki, 1998; Papka et al., 1998) never have motor
Del-Ser et al., 1996; Horimoto et al., 2003; McKeith, parkinsonism, the sensitivity of the Nottingham cri-
2003). Autonomic dysfunction may also contribute to teria was inevitably restricted by this requirement,
DEMENTIA WITH LEWY BODIES 535
which may have been influenced by the inclusion in Recurrent confusional episodes are accompanied
their sample of 5 cases who had motor-only PD diag- by vivid hallucinatory experiences, visual misi-
nosed for at least 12 months before neuropsychiatric dentification syndromes and topographical disor-
features developed. According to current criteria these ientation. Extensive medical screening is usually
cases would be regarded as having PDD and not a negative. Attentional deficits are apparent as
primary diagnosis of DLB. apathy, and daytime somnolence and sleep beha-
The Newcastle upon Tyne (UK) criteria which were viour disorder may be severe. Gait disorder and
formulated at about the same time, based upon a series bradykinesia are often overlooked, particularly
of 21 autopsy cases sampled from a geriatric psychia- in elderly subjects. Frequent falls occur due
try service, stipulated the presence of fluctuating cog- either to postural instability or syncope.
nitive impairment plus two of the following three
The third and final stage often begins with a sud-
items: (1) visual and/or auditory hallucinations, which
den increase in behavioural disturbance leading
are usually accompanied by secondary paranoid delu-
to requests for sedation or hospital admission by
sions; (2) mild spontaneous extrapyramidal features
perplexed and exhausted carers. The natural
or neuroleptic sensitivity syndrome, i.e. exaggerated
course from this point is variable and obscured
adverse responses to standard doses of neuroleptic
by the high incidence of adverse reactions to
medication; and (3) repeated unexplained falls and/or
neuroleptic medication. For patients not receiv-
transient clouding or loss of consciousness.
ing, or tolerating neuroleptics, a progressive
Applied retrospectively to a sample of autopsy-
decline into severe dementia with dysphasia
confirmed cases, these criteria identified 71% of LB
and dyspraxia occurs over months or years, with
pathology cases on initial presentation and 86% between
death usually due to cardiac or pulmonary dis-
presentation and death. Although some of the cases also
ease. During this terminal phase patients show
met clinical criteria for AD, no AD cases met the pro-
continuing behavioural disturbance including
posed LB criteria, indicating that the core features
vocal and motor responses to hallucinatory phe-
selected have good specificity.
nomena. Lucid intervals with some retention of
Applied to another independent sample of DLB,
recent memory function and insight may still be
AD and vascular dementia patients, the Newcastle cri-
apparent. Neurological disability is often pro-
teria yielded a 74% sensitivity rate, averaged across
found with fixed flexion deformities of the neck
four raters, and a specificity of 95%. Inter-rater agree-
and trunk and severe gait impairment.
ment was highest and diagnosis most accurate (90%
versus 55% sensitivity) among more experienced clin-
icians. The most common errors among less experi-
enced clinicians were failure to recognize cognitive 60.3.2. The dementia with Lewy bodies Consortium
fluctuations unique to DLB patients, and a tendency Consensus Criteria
to overvalue comorbid disease as responsible for the
clinical presentation (McKeith et al., 1994). By the early 1990s it was becoming apparent that DLB
The clinical narrative that accompanied these cri- was a relatively common dementia subtype and that
teria described the evolution of the disease in three the several research groups investigating it were adopt-
stages (McKeith et al., 1992b): ing different terminologies for what were essentially
the same patients. The Consortium on DLB therefore
The first stage is often recognised only in retro-
met in October 1995 in Newcastle upon Tyne to agree
spect and may extend back one to three years
common clinical and pathological methods and
pre-presentation with occasional minor episodes
nomenclature. The Consensus Criteria (McKeith
of forgetfulness, sometimes described as lapses
et al., 1996) which resulted were largely based on the
of concentration or switching off. A brief per-
symptom content of the earlier Newcastle and Notting-
iod of delirium is sometimes noted for the first
ham schemes and followed the general structure of
time, often associated with genuine physical ill-
operationalized criteria already in use for AD
ness and/or surgical procedures. Disturbed
(McKhann et al., 1984), assigning levels of possible
sleep, nightmares and daytime drowsiness often
and probable clinical diagnosis.
persist after recovery.
The particular characteristics of the cognitive impair-
Progression to the second stage frequently ments of DLB were described in some detail as differ-
prompts psychiatric or medical referral. A more ing from the dementia syndrome of AD. Probable
sustained cognitive impairment is established, DLB is diagnosed when two of the three key symptoms
albeit with marked fluctuations in severity. in Table 60.1 are present, namely fluctuation, VHs or
536 I. MCKEITH
Table 60.1 DLB cognitive profile, showing pronounced memory
deficits and a clinical presentation more characteristic
Consensus guidelines for the clinical diagnosis of probable
of AD. Fluctuation, VHs and parkinsonism were gener-
and possible dementia with Lewy bodies (DLB)
ally absent in such cases or if present were masked by
1. Central feature the Alzheimer-like clinical picture. The Consensus cri-
Progressive cognitive decline of sufficient magnitude to teria are then able to detect DLB cases with a high posi-
interfere with normal social and occupational function. tive predictive value, but even when used in conjunction
Prominent or persistent memory impairment may not with optimal clinical assessment tools for core features,
necessarily occur in the early stages but is usually will inevitably fail to detect a significant proportion of
evident with progression. Deficits on tests of attention DLB cases in whom those features are absent.
and of frontal subcortical skills and visuospatial ability
may be especially prominent 60.3.3. Revised consensus criteria for the clinical
2. Core features (two core features essential for a diagnosis
diagnosis of dementia with Lewy bodies
of probable, one for possible DLB)
Fluctuating cognition with pronounced variations in
attention and alertness In order to address the perceived shortcomings of the
Recurrent visual hallucinations that are typically well original diagnostic criteria, the DLB Consortium met
formed and detailed again in 2003 to resolve improved methods of identify-
Spontaneous features of parkinsonism ing cases antemortem (McKeith et al., 2005). No major
3. Supportive features amendments to the three core features of DLB were
Repeated falls proposed, but better methods for their clinical assess-
Syncope ment (Table 60.2) were recommended for use in
Transient loss of consciousness diagnosis and for measurement of symptom severity.
Neuroleptic sensitivity A new category of features suggestive of DLB was
Systematized delusions
described, comprising RBD, severe neuroleptic sens-
Hallucinations in other modalities
itivity or abnormal dopamine transporter (DAT) neuroi-
Rapid-eye movement sleep behavior disorder
Depression maging. If one or more of these suggestive features is
4. Features less likely to be present present, in addition to one or more core features, a diag-
History of stroke nosis of probable DLB is made. Possible DLB can be
Any other physical illness or brain disorder sufficient diagnosed if one or more is present in a patient with
enough to interfere with cognitive performance dementia even in the absence of any core features. Sug-
gestive features therefore have a similar diagnostic
Reproduced from McKeith et al. (1996) with permission from Lip- weighting as core clinical features but are not consid-
pincott/Williams & Wilkins.
ered sufficient, even in combination, to warrant a diag-
nosis of probable DLB if all three core features are
spontaneous motor features of parkinsonism and absent.
possible DLB if only one is present. The revised criteria are also more explicit about the
Several retrospective and two prospective studies importance to be attached to clinical and radiological
subsequently examined the predictive accuracy of Con- evidence of cerebrovascular disease (McKeith et al.,
sensus clinical criteria for probable DLB (Litvan et al., 2005) since pathological and imaging studies suggest
2003). These suggested that sensitivity of case detection that white-matter lesions (periventricular and deep
was variable and, although high in one prospective study white matter), microvascular changes and lacunes
(McKeith et al., 2000b), was unacceptably low in several may be present in up to 30% of DLB cases (McKeith,
others. In contrast, specificity was generally found to be et al., 2000b; Jellinger, 2003).
high. The main criticism of the Consensus criteria was
lack of operationalization of the core feature items,
fluctuation in particular (Mega et al., 1996). Modified 60.4. Differential diagnosis
versions of the criteria were proposed which essentially
traded increased sensitivity against reduced diagnostic The main differential diagnoses of DLB are AD, vascular
specificity (Luis et al., 1999; Litvan et al., 2003). How- dementia, PDD, atypical parkinsonian syndromes such as
ever, a later series of neuropathological autopsy studies progressive supranuclear palsy (PSP), MSA, corticobasal
revealed a further reason for the failure of the clinical cri- degeneration (CBD) and also CreutzfeldtJakob
teria to detect demented cases with LB. These studies disease (CJD) (McKeith et al., 1996). The mainstay of
suggested that DLB patients with additional neocortical differential diagnosis remains careful evaluation by a
tangle (Alzheimer) pathology often lacked the typical specialist clinician familiar with these disorders and
DEMENTIA WITH LEWY BODIES 537
Table 60.2
Revised criteria for the clinical diagnosis of dementia with Lewy bodies (DLB)

1. Central feature (essential for a diagnosis of possible or probable DLB)


Dementia defined as progressive cognitive decline of sufficient magnitude to interfere with normal social or occupational
function. Prominent or persistent memory impairment may not necessarily occur in the early stages but is usually evident
with progression. Deficits on tests of attention, executive function and visuospatial ability may be especially prominent
2. Core features (two core features are sufficient for a diagnosis of probable DLB, one for possible DLB)
Fluctuating cognition with pronounced variations in attention and alertness
Recurrent visual hallucinations that are typically well formed and detailed
Spontaneous features of parkinsonism
3. Suggestive features (if one or more of these is present in the presence of one or more core features, a diagnosis of probable
DLB can be made. In the absence of any core features, one or more suggestive features is sufficient for possible DLB.
Probable DLB should not be diagnosed on the basis of suggestive features alone)
Rapid-eye movement sleep behavior disorder
Severe neuroleptic sensitivity
Low dopamine transporter uptake in basal ganglia demonstrated by SPECT or PET imaging
4. Supportive features (commonly present but not proven to have diagnostic specificity)
Repeated falls and syncope
Transient, unexplained loss of consciousness
Severe autonomic dysfunction, e.g. orthostatic hypotension, urinary incontinence
Hallucinations in other modalities
Systematized delusions
Depression
Relative preservation of medial temporal lobe structures on CT/MRI scan
Generalized low uptake on SPECT/PET perfusion scan with reduced occipital activity
Abnormal (low-uptake) MIBG myocardial scintigraphy
Prominent slow-wave activity on EEG with temporal lobe transient sharp waves
5. A diagnosis of DLB is less likely
In the presence of cerebrovascular disease evident as focal neurological signs or on brain imaging
In the presence of any other physical illness or brain disorder sufficient to account in part or in total for the clinical picture
If parkinsonism only appears for the first time at a stage of severe dementia
6. Temporal sequence of symptoms
DLB should be diagnosed when dementia occurs before or concurrently with parkinsonism (if it is present). The term
Parkinsons disease dementia (PDD) should be used to describe dementia that occurs in the context of well-established
Parkinsons disease. In a practice setting the term that is most appropriate to the clinical situation should be used and
generic terms such as Lewy body disease are often helpful. In research studies in which distinction needs to be made
between DLB and PDD, the existing 1-year rule between the onset of dementia and parkinsonism DLB continues to be
recommended. Adoption of other time periods will simply confound data pooling or comparison between studies. In other
research settings that may include clinicopathologic studies and clinical trials, both clinical phenotypes may be considered
collectively under categories such as Lewy body disease or a-synucleinopathy.

Reproduced from McKeith et al. (2005) with permission from Lippincott/Williams & Wilkins.
SPECT, single-photon emission computed tomography; PET, positron emission tomography; CT, computed tomography; MRI, magnetic
resonance imaging; MIBG, 123I-metaiodobenzylguanadine; EEG, electroencephalogram.

incorporating a history from an independent informant. neuroimaging investigations may be helpful. Changes
Enquiry should be made not only about cognitive, psy- associated with DLB include preservation of hippocam-
chiatric and motor features, but also about other symptom pal and medial temporal lobe volume on magnetic reso-
domains such as sleep or autonomic dysfunction. nance imaging (Barber et al., 1999, 2000a) (Fig. 60.1)
and occipital hypoperfusion on single-photon emission
60.4.1. Role of investigations computed tomography (SPECT) (Lobotesis et al., 2001;
Colloby et al., 2002). Other features, such as general-
There are as yet no clinically applicable electrophysio- ized atrophy (Barber et al., 2000a), white-matter
logical, genotypic or cerebrospinal fluid markers to changes (Barber et al., 2000b) and rates of progression
support a DLB diagnosis (McKeith et al., 2004a) but of whole-brain atrophy (OBrien et al., 2001), appear to
538 I. MCKEITH
60.4.2. Distinguishing between dementia with Lewy
bodies and Parkinsons disease dementia

An issue, already alluded to in section 60.1.1 , which


repeatedly causes difficulty in diagnosing DLB, is
uncertainty about its relationship with PDD. PDD is
similar to DLB (Aarsland et al., 2003; Emre, 2003)
with respect to fluctuating neuropsychological func-
tion (Ballard et al., 2002), neuropsychiatric features
(Aarsland et al., 2001a) and extrapyramidal motor fea-
tures (Aarsland et al., 2001b). The findings at autopsy
are also similar in DLB and PDD and the clinical his-
Fig. 60.1. Coronal magnetic resonance imaging (MRI) scan
tory cannot be extrapolated from the neuropathological
of patients with Alzheimers disease (AD) and dementia with
Lewy bodies (DLB) matched for clinical severity of dementia.
findings. The 1996 Consensus statement recommended
Medial temporal lobe (particularly hippocampal) atrophy is less an arbitrary 1-year rule that proposes that the onset
pronounced in DLB, consistent with autopsy findings. Courtesy of dementia within 12 months of parkinsonism quali-
of Dr. Emma Burton. Reproduced from McKeith et al. (2004a), fies as DLB and more than 12 months of parkinsonism
The Lancet Neurology, with permission from Elsevier. before dementia qualifies as PDD. As previously sta-
ted, this distinction between DLB and PDD as two dis-
be unhelpful in differential diagnosis. Functional ima- tinct clinical phenotypes, based solely on the temporal
ging of the DAT defines integrity of the nigrostriatal sequence of appearance of symptoms, has been criti-
dopaminergic system and currently has its main clinical cized by those who regard the different clinical presen-
application in assisting diagnosis of patients with tre- tations as simply representing different points on a
mor of uncertain etiology (Marshall and Grosset, common spectrum of LB disease (Hardy, 2003). This
2003). Imaging with specific SPECT ligands for DAT approach to classification may be preferable for mole-
provides a marker for presynaptic neuronal degenera- cular and genetic studies and for developing therapeu-
tion. DAT imaging is abnormal in idiopathic PD, multi- tics. Clinicians on the other hand prefer diagnostic
ple system atrophy (MSA) and PSP and does not labels that describe the symptoms as patients present
distinguish between these disorders. Low striatal uptake to them and this will often include consideration of
has also been reported in DLB but is normal in AD (Pig- the temporal course. The revised Consensus statement
gott et al., 1999), making DAT scanning particularly use- recommends that, for clinical, operational definitions,
ful in distinguishing between the two disorders (OBrien DLB should be diagnosed when dementia occurs
et al., 2004; Walker et al., 2004) (Fig. 60.2). Scintigraphy before or concurrently with parkinsonism and PDD
with MIBG (Yoshita et al., 2001) enables the quantifica- should be used to describe dementia that occurs in
tion of postganglionic sympathetic cardiac innervation the context of well-established PD. In a practice set-
and has also been suggested to have high sensitivity and ting the term that is most appropriate to the clinical
specificity in the differential diagnosis of DLB from AD situation should be used and generic terms such as
(Taki et al., 2004). LB disease are often more helpful. In research studies
in which distinction is made between DLB and PDD,
the 1-year rule between the onset of dementia and
parkinsonism for DLB continues to be recommended.
Further research efforts which lead to better understand-
ing of the molecular basis for these disorders may lead to
modifications in these temporal considerations.

60.4.3. Pathology and etiology


Fig. 60.2. For full color figure, see plate section. Single-
The original delineation of DLB from other dementing
photon emission computed tomography (SPECT) images of
the dopamine transporter at the level of the striatum using disorders was made on the basis of neuropathological
fluoropropyl (FP)-CIT show marked reduction of activity in findings, i.e. the presence of cortical LB at autopsy.
dementia with Lewy bodies (DLB) compared with normal It is generally accepted that most DLB cases show a-
activity in Alzheimers disease (AD) and normal aging. Cour- synuclein-positive LB and Lewy neurites (LN)
tesy of Professor JT OBrien. Reproduced from McKeith et al. together (Fig. 60.3) with abundant Alzheimer-type
(2004a), The Lancet Neurology, with permission from Elsevier. pathology, predominantly in the form of amyloid
DEMENTIA WITH LEWY BODIES 539
and elements of the ubiquitin-proteasome system, indi-
cating a role of the aggresomal response, but these fea-
tures are not specific for LBs and are found in other
neuronal inclusions (McNaught et al., 2002; Tanaka
et al., 2004).
Cortical LB density is not robustly correlated
with either the severity or duration of dementia
(Gomez-Tortosa et al., 1999; Harding and Halliday,
2001), although associations have been reported with
LB and plaque density in mid frontal cortex (Samuel
et al., 1996). LNs and neurotransmitter deficits are bet-
ter correlates of clinical symptoms (Gomez-Tortosa
et al., 1999; Perry et al., 2003c).
Formal criteria for the pathological diagnosis of
DLB have not yet been established in the way that
they have for other neurodegenerative disorders. The
original 1996 Consensus paper provided guidelines
Fig. 60.3. For full color figure, see plate section. Neuro- for sampling procedures and recommendations about
pathology of dementia with Lewy bodies (DLB). Pathological staining and counting methods (McKeith et al.,
a-synuclein aggregates assume many forms in DLB, including 1996), but these have had two subsequent revisions
typical classical Lewy bodies (LBs) in the pigmented nuclei of
(McKeith et al., 1999, 2005). For the purposes of
the brainstem (A, B), cortical LBs in the neocortex (C) and
assessing Lewy-related pathology, the latest recom-
amygdala, dystrophic or Lewy neurites (LNs) in the CA 2/3
subfield of Ammons horn (D) and neuroaxonal spheroids mendation is to use a-synuclein immunohistochemis-
(E). The burden of a-synuclein aggregates in the neocortex try and a semiquantitative grading of lesion density
(F) can be extreme with cortical LBs in the deeper layers rather than the counting methods previously proposed.
(top left) and LNs throughout the cortical mantle to the pial LB-type pathology is assigned according to the guide-
surface (bottom right). The striatum is also affected (G) with lines in the original Consensus report. Semiquantitative
primarily LNs (H) and dot-like aggregates (I). LNs tend to grading of LB severity is adopted rather than counting
cluster around -amyloid plaques (J), which are also common LB in various brain regions. The pattern of regional
in DLB, and are primarily axonal in location (K). (Antibodies: involvement is more important than total LB count.
a-synuclein in A, CI, and J, K (green); tyrosine hydroxylase
in B; -amyloid in J (red); neurofilaments light chain in K
60.4.5. Alzheimer-type pathology
(red).) Courtesy of Dr. John E Duda. Reproduced from
McKeith et al. (2004a), The Lancet Neurology, with permis-
sion from Elsevier. The guidelines have also been modified to address the
issue of concomitant Alzheimer-type pathology, taking
account of the pathoplastic effect that this has upon the
plaques. Tau-positive inclusions and neocortical neu- clinical presentation. Cases are assigned a likelihood
rofibrillary tangles sufficient to meet Braak stages V that the dementia can be attributed to AD pathology
or VI (sufficient to qualify for a diagnosis of concomi- using the NIAReagan criteria (NIA, National Institute
tant AD) occur in only a minority (1025%) of cases. on Aging), which employs the Consortium to Establish
Alzheimer pathology of any type is not a prerequisite a Registry for Alzheimers Disease (CERAD) method
for the existence of dementia, however, since older for assessing neuritic plaques (Mirra et al., 1991) and
patients with pure LB disease (no plaques or tangles) a topographic staging method for neurofibrillary degen-
may present clinically with cognitive impairment and eration (Harding et al., 2000). This approach allows the
other neuropsychiatric features. reporting pathologist not only to give a detailed semi-
quantitative description of the pathological findings in
60.4.4. Lewy bodies and Lewy-related pathologies an individual case, but also to estimate the probability
(low, intermediate or high) of the patient having had
Immunohistochemical and protein chemical studies the characteristic DLB clinical syndrome.
indicate that LBs and LNs are pathological aggrega-
tions of a-synuclein, DLB being a member of the 60.4.6. Synuclein pathology in Alzheimers disease
family of a-synucleinopathies (Jellinger, 2003) which
also includes PD and MSA. LBs and LNs are asso- a-Synuclein immunoreactive deposits with many of
ciated with intermediate filaments, chaperone proteins the characteristics of LB have also been reported in
540 I. MCKEITH
60% of AD cases, particularly in the amygdalae There are recent reports that triplication of the
(Hamilton, 2000) but seldom involving other brain a-synuclein gene (SNCA) can cause DLB, PD and
regions to any significant extent. This distribution PDD whereas gene duplication is only associated with
probably represents an end-stage phenomenon, with a motor PD, suggesting a gene dose effect (Singleton
secondary accumulation of aggregated synuclein in and Gwinn-Hardy, 2004). However, SCNA multiplica-
severely dysfunctional neurons that are already heavily tion is not found in most LB disease patients (Johnson
burdened by plaque and tangle pathology (Lippa and et al., 2004).
McKeith, 2003). Such cases with amydalae-only LB
are highly unlikely to be associated with the DLB clin- 60.4.8. Neurochemical pathology of dementia with
ical syndrome and their inclusion in a clinicopatholo- Lewy bodies
gical series serves to underestimate the diagnostic
sensitivity of clinical criteria. DLB is a disorder in which functional deficits might
reasonably be postulated to be related to perturbations
60.4.7. Genetics of dementia with Lewy bodies in neurotransmitter function, given the fluctuating
nature of the clinical syndrome and the relative lack
There are few reports to date of autosomal-dominant of major structural disruption of the cortex compared
DLB families. This may partly be due to the relatively with AD. Neurochemical studies of transmitter and
late onset of DLB but also to a failure to recognize the receptor function in frozen autopsy tissue lend some
relationship of DLB with other phenotypes, including support to such a model. Neurotransmitter replacement
PD and autonomic dysfunction. The identification of treatments in DLB which are based on these obser-
mutations in the a-synuclein gene in familial PD vations have produced mixed results, with good
(Polymeropoulos et al., 1997) has not been followed response experienced by some patients receiving choli-
by similar findings in sporadic PD and DLB. Nor have nergic, but a generally poor response to dopaminergic,
associations been found with DLB with polymorph- treatments.
isms in the genes for presenilin 1, presenilin 2, and
a1 antichymotrypsin. These genes are therefore unli- 60.4.8.1. Dopaminergic system
kely to have a major influence on the pathogenesis of
Although neuronal loss within the substantia nigra
DLB.
Because of the association of the 4 allele of the apo- may be less in DLB than in PD, striatal dopamine
concentrations are reduced to an equivalent level
lipoprotein E (APO E) gene on chromosome 19 and
(Piggott et al., 1999). Within the striatum, dopamine
AD, and the presence of b-amyloid in DLB, several
D2-receptor levels are elevated in PD and this compen-
groups have reported genotyping studies. In DLB, the
satory upregulation is maintained for several years. In
e4 allele frequency is elevated in a manner analogous
contrast, D2-receptors are modestly reduced in DLB,
to that found in AD (Benjamin et al., 1994). In PD
particularly in the putamen (Piggott et al., 1999), con-
no association is observed with APO E e4. There are,
tributing to severe neuroleptic sensitivity. Extrastriatal
however, subtle differences in the APO E allele fre-
quencies between AD and DLB, with a higher e2 allele dopaminergic systems are also affected in PD and
DLB. Mesocortical projections to the frontal cortex
frequency and a reduced frequency of the e4/4 geno-
are implicated in working memory performance, and
type in DLB (Morris et al., 1996). Differences in the
dopamine receptors are also expressed in temporal cor-
APO E frequencies may account for some of the dif-
tex (Farde et al., 1997). Investigations in PD have
ferences between the two diseases in terms of clinical
shown no change in early disease in cortical D1-receptors
presentation and pathology, but it is unlikely that one
(Gnanalingham et al., 1993), whereas in advanced PD,
single genetic determinant accounts for the differences
functional imaging studies have indicated D2-receptors
between DLB and AD. Polymorphisms in other puta-
tive risk genes for DLB, which have not been reliably to be reduced by 40% in frontal cortex (Kaasinen et al.,
2000).
replicated, include the butyrylcholinesterase gene K
variant (BCHE K), the cytochrome P-450 gene
CYP2D6 (debrisoquine 4-hydroxylase) and N-acetyl- 60.4.8.2. Cholinergic system
transferase 2 gene locus. Butyrylcholinesterase gene Cholinergic deficits are more marked in PD patients
K/K and atypical variants do however appear to have with dementia compared to those without, as evi-
a role in modifying the level of cognitive impairment denced by greater neuronal loss in the nucleus basalis
and responsiveness to ChEI responsiveness in DLB of Meynert (Jellinger and Bancher, 1995). Further-
(OBrien et al., 2003) and may also influence rate of more, using [123I] iodobenzovasamicol (a marker of
progression of untreated disease (Perry et al., 2003a). vesicular acetylcholine transporter) and SPECT brain
DEMENTIA WITH LEWY BODIES 541
imaging, PDD cases demonstrate extensive cortical 60.5.1. A problem-oriented approach to
binding decreases similar to early-onset (age at onset management
< 64 years) AD (Kuhl et al., 1996). Not only are neo-
cortical presynaptic cholinergic inputs reduced in DLB It is helpful first to draw up a problem list of cognitive,
to a greater extent than in AD (Tiraboschi et al., 2000; psychiatric and motor disabilities and ask the patient
Shinotoh et al., 2001), but there are also losses in and carer to identify the symptoms that they find most
the striatum and pedunculopontine pathway projecting disabling or distressing and which carry highest prior-
to areas including the thalamus. Cortical choline acet- ity for treatment (McKeith, 2005b). The clinician
yltransferase reductions in DLB correlate with cogni- should explain, before any drugs are prescribed, that
tive impairment and VH. The cortical cholinergic treatment gains in a target symptom may be associated
deficit in DLB is, however, independent of the extent with worsening of symptoms in other domains. The
of Alzheimer-type pathology, although this pathology specific risks of neuroleptic sensitivity reactions
is associated with more severe cognitive impairment should be mentioned in all cases and it is prudent to
(Samuel et al., 1997). Nicotinic receptor density, mea- mark patient case notes and records with an alert to
sured using a-bungarotoxin binding (a7 subunit) is reduce the possibility of inadvertent neuroleptic pre-
reported to be lower in hallucinating compared with scribing, particularly in primary care or emergency
non-hallucinating DLB patients (Court et al., 2001). room settings.
In PDD and DLB, muscarinic M1-receptor binding Non-pharmacologic strategies for cognitive symp-
is elevated. In DLB patients at least, this elevation is toms, including explanation, education, reassurance,
greater in individuals with delusions than those with- orientation and memory prompts, attentional cues and
out (Ballard et al., 2000b), suggesting that this psycho- targeted behavioral interventions are an integral part
tic feature may be associated with a greater loss of of the management of DLB and pharmacological treat-
presynaptic cholinergic activity and a consequent ment is most successful when prescribed as part of a
upregulation in the M1-receptor subtype. It is not yet comprehensive management approach (Cohen-Mans-
established whether loss of cholinergic activity in the field, 2006). Similarly, if a patient with DLB has
thalamic reticular formation, reflecting loss of peduncu- become acutely confused and psychotic, intercurrent
lopontine neurons, correlates with fluctuating attention infection and subdural hematoma, in particular, should
and cognition. be actively excluded (McKeith et al., 1996). It cannot
In summary, in both DLB and PDD, extrastriatal always be assumed that worsening of symptoms is
dopaminergic and particularly cholinergic deficits play simply part of the natural fluctuating history of DLB.
a central role in mediating dementia. Non-essential medications capable of causing confu-
sion should be discontinued.
60.5. Clinical management
60.5.2. Antiparkinsonian agents
The pharmacological management of DLB can
be one of the most challenging issues facing neurolo- Levodopa monotherapy is generally accepted as the
gists, psychiatrists, geriatricians, primary care preferred option in DLB. Medication should generally
physicians or others caring for older people. Poly- be introduced at low doses and slowly increased to the
pharmacy is the norm, with multiple pharmacological least dose required to minimize disability. Levodopa-
treatment targets including motor parkinsonism, cog- responsiveness is of the order of 50% in DLB (Bonelli
nitive failure, psychiatric symptoms and autonomic et al., 2004; Molloy et al., 2005), considerably less
dysfunction. The positive effects of ChEIs seen in than in uncomplicated PD but reminiscent of the loss
many DLB patients contrast with the severe, some- in treatment sensitivity which comes later in the course
times fatal, neuroleptic sensitivity reactions that are of PD, especially as cognitive decline and dementia
seen in up to 50% of patients exposed to such agents intervene. Although the reasons for this reduced
(McKeith et al., 1992a; Aarsland et al., 2005). There response to treatment are unclear, it may be partially
is an intermediate responsiveness to antiparkinsonian related to the development of synuclein-positive striatal
agents (Bonelli et al., 2004; Molloy et al., 2005). pathology and associated neuronal dysfunction (Duda
Since there are no treatments currently licenced for et al., 2002) which is no longer amenable to neurotrans-
DLB, all prescribing to this group of patients is mitter replacement. Patients and carers will usually
essentially off-license. In addition to the medicole- indicate when they feel that the lower acceptable limit
gal and liability issues that this can pose for prescri- of antiparkinsonian treatment has been reached. It is
bers, health care providers may be reluctant to not uncommon to find that confusional and psychotic
reimburse drug costs for DLB patients. symptoms are not significantly ameliorated by dose
542 I. MCKEITH
reductions in antiparkinsonians, nor is motor disability ChEI, is additionally an allosteric modulator of nicoti-
substantially aggravated. This suggests that non-dopa- nic receptors. The outcomes of ChEI treatment of DLB
minergic mechanisms may be playing a major role in and PDD have been comprehensively reviewed (Aars-
symptom formation. land et al., 2004). Placebo-controlled randomized con-
Other antiparkinsonian medications, including sele- trolled trials of rivastigmine have shown benefits in
geline, amantadine, catechol-O-methyltransferase inhi- DLB (McKeith et al., 2000a) and PDD (Emre et al.,
bitors and dopamine agonists are contraindicated in 2004), in addition to which there is some short-term
DLB in view of concerns about inducing confusion (Maclean et al., 2001; Reading et al., 2001) and long-
and psychosis (visual illusions, hallucinations and term open-label data (Grace et al., 2001). In the latter
delusions). Anticholinergics should also be avoided study MMSE and Neuropsychiatric Inventory (NPI)
in both DLB and PDD, because they may cause mental scores remained stable over the first 12 months of treat-
clouding, impaired cognition and induce or aggravate ment, then gradually worsened, although not statisti-
hallucinations. There is evidence that their long-term cally significantly so even 2 years after baseline.
use in PD may be associated with dementia (Pondal Motor UPDRS scores tended to improve, probably
et al., 1996) and the accumulation of a higher density because antiparkinsonian treatment was initiated over
of plaque and tangle pathology (Perry et al., 2003b). that time. With donepezil there is a double-blind
cross-over study in PDD (Aarsland et al., 2002) and a
60.5.3. Cholinesterase inhibitors series of open-label studies in DLB (Kaufer et al.,
1998; Shea et al., 1998), including one reporting a
ChEIs appear to be effective and relatively safe in the rebound worsening of neuropsychiatric symptoms,
treatment of neuropsychiatric and cognitive symptoms when treatment was stopped abruptly (Minett et al.,
in DLB (Aarsland et al., 2004), but the number of 2003). Although reinstatement of treatment may reverse
patients studied is relatively small and larger trials such deterioration, it is recommended that DLB patients
are still needed. Apathy, anxiety, impaired attention, who are assessed as responding to ChEIs are maintained
hallucinations, delusions, sleep disturbance and cogni- on treatment long-term. Attempts at switching from one
tive changes are the most frequently cited treatment- ChEI to another were similarly associated with clini-
responsive symptoms. Improvements are generally cally significant withdrawal effects and the authors did
reported as greater than those achieved in AD (Samuel not recommended this treatment strategy (Bhanji and
et al., 2000). Gauthier, 2003). With galantamine there are as yet only
The first report of benefit with ChEIs was with preliminary open-label data (Aarsland and Hutchinson,
tacrine, which was administered in an open-label trial 2003; Edwards et al., 2004).
to 7 PDD patients. Mean age at the time of treatment Taken overall, the effects of the three available
was 74 years and the duration of PD before the onset ChEIs appear similar, with doses in the same range
of dementia was 7.6 years. In all cases there was a as used in AD. Parkinsonian signs do not generally
greatly reduced frequency of VH (Hutchinson and worsen on treatment. Predominant adverse effects are
Fazzini, 1996). Both MMSE scores and UPDRS motor cholinergic in nature (nausea, vomiting, anorexia and
scores improved significantly after treatment, suggest- somnolence) and are generally rated as mild or moder-
ing that not only did cognitive and neuropsychiatric ate. Hypersalivation, rhinorrhea and lacrimation were
features improve with tacrine, but so did extrapyrami- recorded in approximately 15% of DLB and PDD
dal motor function. This latter finding was unexpected patients treated with donepezil (Thomas et al., 2005)
since theoretical predictions would anticipate motor and postural hypotension, falls and syncope are possi-
worsening. Subsequent studies in PDD and DLB have bly also increased. This latter side-effect profile is con-
found no evidence for significant motor deterioration, sistent with the known pre-existing autonomic
although a minority of patients do experience dose- dysfunction in DLB and such symptoms are likely to
dependent worsening of their tremor. If troublesome, occur with all procholinergic agents.
this can be managed in most cases by ChEI dose The presence of VH in DLB predicted a good
reduction or increase in levodopa. response to rivastigmine in DLB assessed using a com-
The ChEI generally available at present are rivas- puter-based system measuring attentional speed and
tigmine, donepezil and galantamine; all three prevent accuracy (McKeith et al., 2004b). Cognitive reaction
the inactivation of acetylcholine after its release from time, calculated by subtracting simple from choice reac-
the neurone. Rivastigmine is a dual inhibitor of acetyl- tion times, was however not improved by rivastigmine.
cholinesterase and butyrylcholinesterase, whereas This suggests that this cognitive component may be
donepezil and galantamine are acetylcholinesterase- dopaminergically mediated, and the marked reductions
selective. Galantamine, the most recently licenced that are seen in cognitive reaction time in DLB and
DEMENTIA WITH LEWY BODIES 543
PDD but not in AD (Ballard et al., 2002; Wesnes et al., frequently seen in LB disease and may be an early feature.
2002) are related to a loss of mesocortical dopaminergic RBD can be treated with clonazepam 0.25 mg at bedtime,
projections. melatonin 3 mg at bedtime or quetiapine 12.5 mg at bed-
time and titrated slowly, monitoring for both efficacy
60.5.4. Antipsychotic agents and side-effects (McKeith et al., 2005). ChEIs may also
be helpful for disturbed sleep (Reading et al., 2001).
Since hallucinations, delusions and associated beha- The frequent occurrence of electroencephalographic
vioral disturbances are so frequent in DLB, a very (EEG) abnormalities in DLB, incuding transient tem-
common clinical scenario is for the clinician to con- poral slow waves, has prompted the use of carbamaze-
sider using antipsychotic medication. Traditional pine and sodium valproate, usually for agitation or
agents with potent D2-receptor antagonism are asso- insomnia. No systematic reports of efficacy or side-
ciated with severe neuroleptic sensitivity reactions in effects are available. The 5-HT3 antagonist ondansetron
up to 50% of DLB patients, leading to substantial mor- was reported as having antipsychotic effects in halluci-
bidity and a two- to threefold increased mortality risk nated PD patients but this has not been independently
(McKeith et al., 1992a; Aarsland et al., 2005). These replicated and the high doses required make the cost
reactions are generally evident within the first few prohibitive for routine practice (Harrison and McKeith,
doses or after increase from a previously tolerated 1995).
dose. Sensitivity reactions have also been reported
for second-generation antipsychotic agents such as ris-
60.5.6. Disease-modifying agents
peridone and olanzapine (McKeith et al., 1995; Walker
et al., 1999). Newer atypicals with potentially more
There are no disease-modifying pharmacological
favorable pharmacological properties, such as quetia-
therapies for DLB. Better understanding of the neuro-
pine, clozapine and aripiprazole, have theoretical
biology of synuclein may lead to the development of
advantages in LB disease (Fernandez et al., 2003; Terao
novel therapeutic interventions applicable to a diverse
et al., 2003) but controlled clinical trial data are lacking
group of neurodegenerative disorders, including DLB,
and clinicians should remain vigilant to the possibility
PD and MSA, but until then the clinician has only
of adverse side-effects. If acute deterioration occurs
symptomatic treatments available. Clinical trial data
in a confused elderly patient following neuroleptic
suggest that ChEIs may potentially slow disease pro-
administration, DLB should always be considered as
gression in AD, and similar effects could be antici-
part of the differential diagnosis. However, since up to
pated in DLB.
50% of DLB patients receiving typical or atypical anti-
psychotic agents do not react adversely, a history of
neuroleptic tolerance does not therefore rule out a diag- 60.6. Summary
nosis of DLB and deliberate neuroleptic challenge is
neither a safe nor a reliable diagnostic procedure. DLB is now established as a relatively common form
of dementia in older people and is part of a larger
60.5.5. Other pharmacological agents for spectrum of LB disease which has diverse clinical pre-
symptomatic treatment sentations, including PD, primary autonomic failure
and RBD. Recent modifications to clinical and patho-
Depression is common in DLB but there have been no logical guidelines for the assessment and diagnosis of
systematic studies of its management. At the present DLB have been made (McKeith et al., 2005) and these
time selective serotonin reuptake inhibitors and sero- will require independent validation in a variety of set-
tonin-norepinephrine reuptake inhibitors (SNRIs) are tings. Suspicion of a DLB diagnosis should be raised
probably the preferred pharmacological treatments. in the presence of any one of the proposed core or sug-
Tricylic antidepressants and those with anticholinergic gestive features and may be supported by the use of
properties should be avoided. Apathy is a common specifically designed assessment scales and appropri-
feature which may mimic depression or excessive day- ate neuroimaging techniques. ChEIs offer the greatest
time somnolence and which may improve with ChEIs. hope for symptomatic improvement in both psychotic
Anxiety is frequently secondary to fluctuating confu- and cognitive features of DLB and seem to offer these
sion and hallucinations and improves when these are benefits without significant compromise of motor
treated. Small doses of benzodiazepines may offer function. Ongoing multicenter, double-blind, placebo-
short-term symptomatic relief but at the expense of wor- controlled studies will hopefully clarify these issues,
sening amnesia, alertness and motor function, with whereas long-term studies are required to assess the
increased risk of falls. Sleep disorders are also duration of benefit. Antipsychotic drugs need to be
544 I. MCKEITH
used sparingly and only when other pharmacological Ballard C, Holmes C, McKeith I et al. (1999). Psychiatric
and non-pharmacological approaches fail to prevent morbidity in dementia with Lewy bodies: a prospective
severe distress or injurious behavior to self or others. clinical and neuropathological comparative study with
Clinicians should be vigilant for severe neuroleptic Alzheimers disease. Am J Psychiatry 156 (7): 10391045.
Ballard C, OBrien J, Swann A et al. (2000a). One year fol-
sensitivity reactions. Clonazepam may have a particu-
low-up of parkinsonism in dementia with Lewy bodies.
lar role in the management of sleep disorders but the
Dement Geriatr Cogn Disord 11: 219222.
role of other sedatives and anticonvulsants is unpro- Ballard C, Piggott M, Johnson M et al. (2000b). Delusions
ven. New-generation antidepressants may be helpful associated with elevated muscarinic binding in dementia
for persistent depressive symptoms. Disease-modifying with Lewy bodies. Ann Neurol 48: 868876.
treatments are urgently required. Better understanding Ballard C, OBrien J, Morris CM et al. (2001). The progres-
of the pathobiological processing of synuclein proteins sion of cognitive impairment in dementia with Lewy
could ultimately lead to the development of novel bodies, vascular dementia and Alzheimers disease. Int
therapeutic interventions for DLB. J Geriatr Psychiatry 16 (5): 499503.
Ballard CG, Aarsland D, McKeith IG et al. (2002). Fluctua-
tions in attentionPD dementia vs DLB with parkinson-
References ism. Neurology 59 (11): 17141720.
Barber R, Gholkar A, Scheltens P et al. (1999). Medial tem-
Aarsland D, Hutchinson M (2003). Galantamine for Parkin- poral lobe atrophy on MRI in dementia with Lewy bodies.
sons disease with dementia. Int Psychogeriatr 15: 243. Neurology 52: 11531158.
Aarsland D, Ballard C, Larsen JP et al. (2001a). A compara- Barber R, Ballard C, McKeith IG et al. (2000a). MRI volu-
tive study of psychiatric symptoms in dementia with Lewy metric study of dementia with Lewy bodies. A comparison
bodies and Parkinsons disease with and without dementia. with AD and vascular dementia. Neurology 54: 13041309.
Int J Geriatr Psychiatry 16: 528536. Barber R, Gholkar A, Scheltens P et al. (2000b). MRI
Aarsland D, Ballard C, McKeith I et al. (2001b). Comparison volumetric correlates of white matter lesions in dementia
of extrapyramidal signs in dementia with Lewy bodies and with Lewy bodies and Alzheimers disease. Int J Geriatr
Parkinsons disease. J Neuropsychiatry Clin Neurosci 13: Psychiatry 15: 911916.
374379. Benjamin R, Leake A, Edwardson JA et al. (1994). Apolipo-
Aarsland D, Laake K, Larsen JP et al. (2002). Donepezil for cog- protein E genes in Lewy body and Parkinsons disease.
nitive impairment in Parkinsons disease: a randomised con- Lancet 343: 1565.
trolled study. J Neurol Neurosurg Psychiatry 72 (6): 708712. Bhanji NH, Gauthier S (2003). Dementia with Lewy bodies:
Aarsland D, Andersen K, Larsen JP et al. (2003). Prevalence preliminary observations on cholinesterase inhibitor
and characteristics of dementia in Parkinson disease: an switching. Int Psychogeriatr 15: 179.
8-year prospective study. Arch Neurol 60 (3): 387392. Birkett DP, Desouky A, Han L et al. (1992). Lewy bodies in
Aarsland D, Mosimann UP, McKeith IG (2004). Role of psychiatric patients. Int J Geriatr Psychiatry 7: 235240.
cholinesterase inhibitors in Parkinsons disease and Boeve BF, Silber MH, Ferman TJ et al. (1998). REM sleep
dementia with Lewy bodies. J Geriatr Psychiatry Neurol behaviour disorder and degenerative dementia. An associa-
17 (3): 164171. tion likely reflecting Lewy body disease. Neurology 51:
Aarsland D, Ballard C, Larsen JP et al. (2005). Marked 363370.
neuroleptic sensitivity in dementia with Lewy bodies and Boeve BF, Silber MH, Ferman TJ et al. (2001). Association of
Parkinsons disease. J Clin Psychiatry 66 (5): 633637. REM sleep behavior disorder and neurodegenerative disease
Ala T, Hughes LF, Kyrouac GA et al. (2004). Use of MMSE may reflect an underlying synucleinopathy. Mov Disord 16:
to differentiate Alzheimers disease from dementia with 622630.
Lewy bodiesresponse to Larner The Mini-Mental State Boeve BF, Silber MH, Parisi JE et al. (2003). Synucleinopa-
exam may help in the differentiation of dementia with thy pathology and REM sleep behavior disorder plus
Lewy bodies and Alzheimers disease. Int J Geriatr dementia or parkinsonism. Neurology 61: 4045.
Psychiatry 19 (12): 1209; author reply 12091210. Boeve BF, Silber MH, Ferman TJ (2004). REM sleep
Armstrong TP, Hansen LA, Salmon DP et al. (1991). Rapidly behavior disorder in Parkinsons disease and dementia with
progressive dementia in a patient with the Lewy body Lewy bodies. J Geriatr Psychiatry Neurol 17 (3): 146157.
variant of Alzheimers disease. Neurology 41: 11781180. Bonelli SB, Ransmayr G, Steffelbauer M et al. (2004). L-dopa
Ballard CG, Mohan RNC, Patel A et al. (1993). Idiopathic responsiveness in dementia with Lewy bodies, Parkinson dis-
clouding of consciousnessdo the patients have cortical ease with and without dementia. Neurology 63 (2): 376378.
Lewy body disease? Int J Geriatr Psychiatry 8: 571576. Braak H, Del Tredici K, Rub U et al. (2003). Staging of brain
Ballard C, Grace J, McKeith I et al. (1998a). Neuroleptic pathology related to sporadic Parkinsons disease. Neuro-
sensitivity in dementia with Lewy bodies and Alzheimers biol Aging 24 (2): 197211.
disease. Lancet 351: 10321033. Bradshaw J, Saling M, Hopwood M et al. (2004). Fluctuating
Ballard C, Shaw F, McKeith I et al. (1998b). High prevalence cognition in dementia with Lewy bodies and Alzheimers
of neurovascular instability in neurodegenerative demen- disease is qualitatively distinct. J Neurol Neurosurg
tias. Neurology 51: 17601762. Psychiatry 75: 382387.
DEMENTIA WITH LEWY BODIES 545

Burkhardt CR, Filley CM, Kleinschmidt-DeMasters BK et al. Edwards KR, Hershey L, Wray L et al. (2004). Efficacy and
(1988). Diffuse Lewy body disease and progressive demen- safety of galantamine in patients with dementia with Lewy
tia. Neurology 38: 15201528. bodies: a 12-week interim analysis. Dement Geriatr Cogn
Burn DJ, McKeith IG (2004). Update on treatment strategies for Disord S1: 4048.
dementia with Lewy bodies. World J Biol Psychiatry 5 (1): 78. Emre M (2003). Dementia associated with Parkinsons dis-
Burn DJ, Rowan EN, Minett T et al. (2003). Extrapyramidal ease. Lancet Neurol 2 (4): 229237.
features in Parkinsons disease with and without dementia Emre M, Aarsland D, Albanese A et al. (2004). Rivastigmine
and dementia with Lewy bodies: a cross-sectional com- for dementia associated with Parkinsons disease. N Engl J
parative study. Mov Disord 18 (8): 884889. Med 351 (24): 25092518.
Byrne EJ, Lennox G, Lowe J et al. (1989). Diffuse Lewy Fahn S, Elton RL (1987). Unified Parkinsons disease rating
body disease: clinical features in 15 cases. J Neurol Neu- scale. In: S Fahn, CD Marsden, M Goldstein, DB Calne
rosurg Psychiatry 52: 709717. (Eds.), Recent Developments in Parkinsons Disease.
Byrne EJ, Lennox G, Godwin-Austen RB et al. McMillan, New York, pp. 153163.
(1991). Dementia associated with cortical Lewy bodies. Farde L, Suhara T, Nyberg S et al. (1997). A PET study of
Proposed diagnostic criteria. Dementia 2: 283284. [11C]FLB 457 binding to extrastriatal D2-dopamine recep-
Calderon J, Perry RJ, Erzinclioglu SW et al. (2001). Perception, tors in healthy subjects and antipsychotic drug-treated
attention, and working memory are disproportionately patients. Psychopharmacology (Berl) 133: 396404.
impaired in dementia with Lewy bodies compared with Alzhei- Ferman TJ, Boeve BF, Smith GE et al. (1999). REM sleep
mers disease. J Neurol Neurosurg Psychiatry 70 (2): 157164. behaviour disorder and dementia. Cognitive differences
Cohen-Mansfield J (2006). Non-pharmacological manage- when compared with AD. Neurology 52: 951957.
ment of DLB. In: JT OBrien, I McKeith, D Ames, E Ferman TJ, Smith GE, Boeve BF et al. (2004). DLB fluctua-
Chiu (Eds.), Dementia with Lewy Bodies. Dunitz, New tions: specific features that reliably differentiate from AD
York, pp. 193206. and normal aging. Neurology 62 (2): 181187.
Collerton D, Burn D, McKeith I et al. (2003). Systematic Fernandez HH, Wu CK, Ott BR (2003). Pharmacotherapy of
review and meta-analysis show that dementia with Lewy dementia with Lewy bodies. Expert Opin Pharmacother
bodies is a visual-perceptual and attentional-executive 4 (11): 20272037.
dementia. Dement Geriatr Cogn Disord 16 (4): 229237. Forstl H, Levy R (1991). FH Lewy on Lewy bodies, par-
Collerton D, Perry E, McKeith I (2005). Why people see kinsonism and dementia. Int J Geriatr Psychiatry 6:
things that are not there: a novel Perception and Attention 757766.
Deficit model for recurrent complex visual hallucinations. Forstl H, Burns A, Luthert P et al. (1993). The Lewy-body
Behav Brain Sci 28: 737757. variant of Alzheimers disease. Clinical and pathological
Colloby SJ, Fenwick JD, Williams ED et al. (2002). A com- findings. Br J Psychiatry 162: 385392.
parison of 99mTc-HMPAO SPECT changes in dementia Gibb WRG, Esiri MM, Lees AJ (1987). Clinical and patho-
with Lewy bodies and Alzheimers disease using statisti- logical features of diffuse cortical Lewy body disease
cal parametric mapping. Eur J Nucl Med 29 (5): 615622. (Lewy body dementia). Brain 110: 11311153.
Connor DJ, Salmon DP, Sandy TJ et al. (1998). Cognitive Gibb WRG, Luthert PJ, Janota I et al. (1989). Cortical Lewy
profiles of autopsy-confirmed Lewy body variant vs pure body dementia: clinical features and classification. J Neu-
Alzheimer disease. Arch Neurol 55 (7): 9941000. rol Neurosurg Psychiatry 52: 185192.
Court JA, Ballard CG, Piggott MA et al. (2001). Visual Gnanalingham KK, Smith LA, Hunter AJ et al. (1993). Altera-
hallucinations are associated with lower a-bungarotoxin tions in striatal and extrastriatal D-1 and D-2 dopamine
binding in dementia with Lewy bodies. Pharmacol Biochem receptors in the MPTP-treated common marmoset: an
Behav 70: 571579. autoradiographic study. Synapse 14: 184194.
Crystal HA, Dickson DW, Lizardi JE et al. (1990). Antemor- Gomez-Tortosa E, Newell K, Irizarry MC et al. (1999). Clini-
tem diagnosis of diffuse Lewy body disease. Neurology 40: cal and quantitative pathological correlates of dementia
15231528. with Lewy bodies. Neurology 53: 12841291.
Del-Ser T, Munoz DG, Hachinski V (1996). Temporal pat- Grace J, Daniel S, Stevens T et al. (2001). Long-term use of
tern of cognitive decline and incontinence is different in rivastigmine in patients with dementia with Lewy bodies:
Alzheimers disease and diffuse Lewy body disease. Neu- an open-label trial. Int Psychogeriatr 13 (2): 199205.
rology 46: 682686. Hamilton RL (2000). Lewy bodies in Alzheimers disease:
Del-Ser T, McKeith I, Anand R et al. (2000). Dementia with a neuropathological review of 145 cases using alpha-
Lewy bodies: findings from an international multicentre synuclein immunohistochemistry. Brain Pathol 10:
study. Int J Geriatr Psychiatry 15: 10341045. 378384.
Dickson DW, Davies P, Mayeux R et al. (1987). Diffuse Hansen L, Salmon D, Galasko D et al. (1990). The Lewy
Lewy body disease. Neuropathological and biochemical body variant of Alzheimers disease: a clinical and patho-
studies of six patients. Acta Neuropathol (Berl) 75: 815. logic entity. Neurology 40: 18.
Duda JE, Giasson BI, Mabon ME et al. (2002). Novel anti- Harding AJ, Halliday GM (2001). Cortical Lewy body
bodies to synuclein show abundant striatal pathology in pathology in the diagnosis of dementia. Acta Neuropathol
Lewy body diseases. Ann Neurol 52 (2): 205210. (Berl) 102: 355363.
546 I. MCKEITH

Harding AJ, Kril JJ, Halliday GM (2000). Practical mea- Lennox G, Lowe J, Landon M et al. (1989b). Diffuse Lewy
sures to simplify the Braak tangle staging method for body disease: correlative neuropathology using anti-ubi-
routine pathological screening. Acta Neuropathol (Berl) quitin immunocytochemistry. J Neurol Neurosurg Psy-
99 (2): 199208. chiatry 52: 12361247.
Hardy J (2003). The relationship between Lewy body dis- Lippa CF, McKeith I (2003). Dementia with Lewy bodies
ease, Parkinsons disease, and Alzheimers disease. Ann improving diagnostic criteria. Neurology 60: 15711572.
NY Acad Sci 991: 167170. Litvan I, MacIntyre A, Goetz CG et al. (1998). Accuracy
Harrison RH, McKeith IG (1995). Senile dementia of Lewy of the clinical diagnoses of Lewy body disease, Parkin-
body typea review of clinical and pathological features: sons disease, and dementia with Lewy bodies. Arch
implications for treatment. Int J Geriatr Psychiatry 10: Neurol 55: 969978.
919926. Litvan I, Bhatia KP, Burn DJ et al. (2003). SIC Task Force
Holmes C, Cairns N, Lantos P et al. (1999). Validity of current appraisal of clinical diagnostic criteria for parkinsonian
clinical criteria for Alzheimers disease, vascular dementia disorders. Mov Disord 18 (5): 467486.
and dementia with Lewy bodies. Br J Psychiatry 174: 4550. Lobotesis K, Fenwick JD, Phipps A et al. (2001). Occipital
Horimoto Y, Matsumoto M, Akatsu H et al. (2003). Auto- hypoperfusion on SPECT in dementia with Lewy bodies
nomic dysfunctions in dementia with Lewy bodies. J Neu- but not AD. Neurology 56: 643649.
rol 250 (5): 530533. Lopez OL, Wisniewski S, Hamilton RL et al. (2000). Predic-
Hutchinson M, Fazzini E (1996). Cholinesterase inhibitors in tors of progression in patients with AD and Lewy bodies.
Parkinsons disease. J Neurol Neurosurg Psychiatry 61: Neurology 54 (9): 17741779.
324325. Luis CA, Barker WW, Gajaraj K et al. (1999). Sensitivity
Jellinger KA (1996). Structural basis of dementia in neurode- and specificity of three clinical criteria for dementia with
generative disorders. J Neural Transm 47: 129. Lewy bodies in an autopsy-verified sample. Int J Geriatr
Jellinger KA (2003). Neuropathological spectrum of synu- Psychiatry 14: 526533.
cleinopathies. Mov Disord 18 (Suppl 6): S2S12. Maclean LE, Collins CC, Byrne EJ (2001). Dementia with
Jellinger KA, Bancher CH (1995). Structural basis of mental Lewy bodies treated with rivastigmine: effects on cogni-
impairment in Parkinsons disease. Neuropsychiatrie 9: tion, neuropsychiatric symptoms, and sleep. Int Psycho-
914. geriatr 13: 277288.
Johnson J, Hague SM, Hanson M et al. (2004). SNCA multi- Marshall V, Grosset D (2003). Role of dopamine transporter
plication is not a common cause of Parkinson disease or imaging in routine clinical practice. Mov Disord 18 (12):
dementia with Lewy bodies. Neurology 63 (3): 554556. 14151423.
Kaasinen V, Nagren K, Hietala J et al. (2000). Extrastriatal McKeith I (2005a). Dementia with Lewy bodies: a clinical
dopamine D2 and D3 receptors in early and advanced Par- overview. In: A Burns, JT OBrien, D Ames (Eds.),
kinsons disease. Neurology 54: 14821487. Dementia. Arnold, London.
Kaufer DI, Catt KE, Lopez OL et al. (1998). Dementia with McKeith I (2005b). Pharmacological management of DLB.
Lewy bodies: response of delirium-like features to done- In: JT OBrien, I McKeith, D Ames, E Chiu (Eds.),
pezil. Neurology 51: 1512. Dementia with Lewy Bodies. Dunitz, New York, pp.
Klatka LA, Louis ED, Schiffer RB (1996). Psychiatric fea- 183192.
tures in diffuse Lewy body disease: findings in 28 patholo- McKeith I, Fairbairn A, Perry R et al. (1992a). Neuroleptic
gically diagnosed cases. Neurology 46: A180. sensitivity in patients with senile dementia of Lewy body
Kosaka K (1978). Lewy bodies in cerebral cortex, report of type. BMJ 305: 673678.
three cases. Acta Neuropathol (Berl) 42: 127134. McKeith I, Del-Ser T, Spano PF et al. (2000a). Efficacy of
Kosaka K, Iseki E (1998). Recent advances in dementia rivastigmine in dementia with Lewy bodies: a randomised,
research in Japan: non-Alzheimer type degenerative double-blind, placebo-controlled international study.
dementias. Psychiatry Clin Neurosci 52: 367373. Lancet 356: 20312036.
Kosaka K, Yoshimura M, Ikeda K et al. (1984). Diffuse type McKeith I, Mintzer J, Aarsland D et al. (2004a). Dementia
of Lewy body disease: progressive dementia with abun- with Lewy bodies. Lancet Neurol 3: 1928.
dant cortical Lewy bodies and senile changes of varying McKeith I, Dickson D, Emre M et al. (2005). Dementia with
degreea new disease? Clin Neuropathol 3 (5): 185192. Lewy bodies: diagnosis and management: Third Report of
Kosaka K, Iseki E, Odawara T et al. (1996). Cerebral type of the DLB Consortium. Neurology 65 (12): 18631872.
Lewy body disease. Neuropathology 16: 3235. McKeith IG (2003). The clinical spectrum of Lewy body dis-
Kuhl DE, Minoshima S et al. (1996). In vivo mapping of ease. The Third Kuopio Alzheimer Symposium, Vol. 3,
cholinergic terminals in normal aging, Alzheimers dis- Kuopio, Finland. p. 25.
ease, and Parkinsons disease. Ann Neurol 40: 399410. McKeith IG, Perry RH, Fairbairn AF, et al. (1992b). Opera-
Kuzuhara S, Yoshimura M (1993). Clinical and neuropatho- tional criteria for senile dementia of Lewy body type
logical aspects of diffuse Lewy body disease in the (SDLT). Psychol Med 22: 911922.
elderly. Adv Neurol 60: 464469. McKeith IG, Fairbairn AF, Perry RH et al. (1994). The
Lennox G, Lowe J, Byrne EJ et al. (1989a). Diffuse Lewy clinical diagnosis and misdiagnosis of senile dementia of
body disease. Lancet 1 (8633): 323324. Lewy body type (SDLT). Br J Psychiatry 165: 324332.
DEMENTIA WITH LEWY BODIES 547

McKeith IG, Ballard CG, Harrison RWS (1995). Neuroleptic disease dementia and dementia with Lewy bodies. Am J
sensitivity to risperidone in Lewy body dementia. Lancet Geriatr Psychiatry 14: 153160.
346: 699. OBrien JT, Paling S, Barber R et al. (2001). Progressive
McKeith IG, Galasko D, Kosaka K et al. (1996). Consensus brain atrophy on serial MRI in dementia with Lewy
guidelines for the clinical and pathologic diagnosis of demen- bodies, AD, and vascular dementia. Neurology 56 (10):
tia with Lewy bodies (DLB): report of the consortium on 13861388.
DLB international workshop. Neurology 47: 11131124. OBrien JT, Colloby SJ, Fenwick J et al. (2004). Dopamine
McKeith IG, Perry EK, Perry RH (1999). Report of the sec- transporter loss visualised with FP-CIT SPECT in demen-
ond dementia with Lewy body international workshop: tia with Lewy bodies. Arch Neurol 61 (6): 919925.
diagnosis and treatment. Consortium on dementia with OBrien KK, Saxby BK, Ballard CG et al. (2003). Regulation
Lewy bodies. Neurology 53 (5): 902905. of attention and response to therapy in dementia by
McKeith IG, Ballard CG, Perry RH et al. (2000b). Pros- butyrylcholinesterase. Pharmacogenetics 13: 231239.
pective validation of consensus criteria for the diagnosis Okazaki H, Lipkin LE, Aronson SM (1961). Diffuse intracyto-
of dementia with Lewy bodies. Neurology 54: 10501058. plasmic ganglionic inclusions (Lewy type) associated with
McKeith IG, Wesnes KA, Perry E et al. (2004b). Hallucina- progressive dementia and quadriparesis in flexion. J Neuro-
tions predict attentional improvements with rivastigmine pathol Exp Neurol 20: 237244.
in dementia with Lewy bodies. Dement Geriatr Cogn Disord Papka M, Rubio A, Schiffer RB (1998). A review of Lewy
18: 94100. body disease, an emerging concept of cortical dementia.
McKhann G, Drachman D, Folstein M et al. (1984). Clinical J Neuropsychiatry Clin Neurosci 10: 267279.
diagnosis of Alzheimers diseasereport of the NINCDS- Perry E, McKeith I, Ballard C (2003a). Butyrylcholinesterase
ADRDA Work Group under the auspices of Department and progression of cognitive deficits in dementia with
of Health and Human Services Task Force on Alzheimers Lewy bodies. Neurology 60 (11): 18521853.
disease. Neurology 34 (7): 939944. Perry EK, Kilford L, Lees AJ et al. (2003b). Increased
McNaught KS, Shashidharan P, Perl DP et al. (2002). Aggre- Alzheimer pathology in Parkinsons disease related to
some-related biogenesis of Lewy bodies. Eur J Neurosci antimuscarinic drugs. Ann Neurol 54 (2): 235238.
16 (11): 21362148. Perry EK, Piggott MA, Johnson M et al. (2003c). Neuro-
McShane RH, Esiri MM, Joachim C et al. (1998). Prospective transmitter correlates of neuropsychiatric symptoms in
evaluation of diagnostic criteria for dementia with Lewy dementia with Lewy bodies. In: MA Bedard, Y Agid, S
bodies. Neurobiol Aging 19 (4S): S204. Chouinard et al. (Eds.), Mental and Behavioral Dysfunc-
Mega MS, Masterman DL, Benson F et al. (1996). Dementia tion in Movement Disorders. Humana Press Inc, Totowa,
with Lewy bodies: reliability and validity of clinical and NJ, pp. 285294.
pathologic criteria. Neurology 47: 14031409. Perry RH, Irving D, Blessed G et al. (1989a). Clinically and
Merdes AR, Hansen LA, Jeste DV et al. (2003). Influence of neuropathologically distinct form of dementia in the
Alzheimer pathology on clinical diagnostic accuracy in elderly. Lancet 1 (8630): 166.
dementia with Lewy bodies. Neurology 60 (10): 15861590. Perry RH, Irving D, Blessed G et al. (1989b). Senile demen-
Minett TSC, Thomas A, Wilkinson LM et al. (2003). What tia of Lewy body type and spectrum of Lewy body
happens when donepezil is suddenly withdrawn? An open disease. Lancet 1 (8646): 1088.
label trial in dementia with Lewy bodies and Parkinsons Perry RH, Irving D, Blessed G et al. (1990). Senile dementia
disease with dementia. Int J Geriatr Psychiatry 18: 988993. of Lewy body type. A clinically and neuropathologically
Mirra SS, Heyman A, McKeel D et al. (1991). The Consortium distinct form of Lewy body dementia in the elderly.
to Establish a Registry for Alzheimers Disease (CERAD) II. J Neurol Sci 95: 119139.
Standardisation of the neuropathological assessment of Piggott MA, Marshall EF, Thomas N et al. (1999). Striatal
Alzheimers disease. Neurology 41: 479486. dopaminergic markers in dementia with Lewy bodies,
Molloy S, McKeith I, OBrien JT et al. (2005). The role of Alzheimers and Parkinsons diseases: rostrocaudal distri-
levodopa in the management of dementia with Lewy bution. Brain 122: 14491468.
bodies. J Neurol Neurosurg Psychiatry 76 (9): 12001203. Polymeropoulos MH, Lavedan C, Leroy E et al. (1997). Muta-
Mormont E, Grymonprez LL, Baisset-Mouly C et al. (2003). tion in the alpha-synuclein gene identified in families with
The profile of memory disturbance in early Lewy body Parkinsons disease. Science 276 (5321): 20452047.
dementia differs from that in Alzheimers disease. Rev Pondal M, DelSer T, Bermejo F (1996). Anticholinergic ther-
Neurol (Paris) 159 (89): 762766. apy and dementia in patients with Parkinsons disease.
Morris CM, Massey HM, Benjamin R et al. (1996). Molecular J Neurol 243 (7): 543546.
biology of APO E alleles in Alzheimers and non-Alzheimers Rahkonen T, Eloniemi-Sulkava U, Rissanen S et al. (2003).
dementias. J Neural Transm 47: 205218. Dementia with Lewy bodies according to the consensus
Mosimann UP, Mather G, Wesnes K et al. (2004). Visual criteria in a general population aged 75 years or older.
perception in Parkinson disease dementia and dementia J Neurol Neurosurg Psychiatry 74 (6): 720724.
with Lewy bodies. Neurology 63 (11): 20912096. Reading PJ, Luce AK, McKeith IG (2001). Rivastigmine in
Mosimann UP, Rowan EN, Partington CE, et al. (2006). the treatment of parkinsonian psychosis and cognitive
Characteristics of visual hallucinations in Parkinsons impairment. Mov Disord 16 (6): 11711174.
548 I. MCKEITH

Sahgal A, Galloway PH, McKeith IG et al. (1992). A com- Terao T, Shimomura T, Izumi Y et al. (2003). Two cases of
parative study of attentional deficits in senile dementias quetiapine augmentation for donepezil-refractory visual
of Alzheimer and Lewy body types. Dementia 3: 350354. hallucinations in dementia with Lewy bodies. J Clin
Samuel W, Galasko D, Masliah E et al. (1996). Neocortical Psychiatry 64 (12): 15201521.
Lewy body counts correlate with dementia in the Lewy Thomas AJ, Burn DJ, Rowan EN et al. (2005). A comparison
body variant of Alzheimers disease. J Neuropathol Exp of the efficacy of donepezil in Parkinsons disease with
Neurol 55 (1): 4452. dementia and dementia with Lewy bodies. Int J Geriatr
Samuel W, Alford M, Hofstetter CR et al. (1997). Dementia Psychiatry 20: 938944.
with Lewy bodies versus pure Alzheimer disease: differ- Tiraboschi P, Hansen LA, Alford M et al. (2000). Cholinergic
ences in cognition, neuropathology, cholinergic dysfunc- dysfunction in diseases with Lewy bodies. Neurology 54:
tion, and synapse density. J Neuropathol Exp Neurol 56: 407411.
499508. Walker MP, Ayre GA, Cummings JL et al. (2000a). The
Samuel W, Caligiuri M, Galasko D et al. (2000). Better cogni- Clinician Assessment of Fluctuation and the One Day
tive and psychopathologic response to donepezil in patients Fluctuation Assessment Scale. Two methods to assess
prospectively diagnosed as dementia with Lewy bodies: a fluctuating confusion in dementia. Br J Psychiatry 177:
preliminary study. Int J Geriatr Psychiatry 15 (9): 794802. 252256.
Shea C, MacKnight C, Rockwood K (1998). Donepezil for Walker Z, Allan RL, Shergill S et al. (1997). Neuropsycholo-
treatment of dementia with Lewy bodies: a case series of gical performance in Lewy body dementia and Alzhei-
nine patients. Int Psychogeriatr 10 (3): 229238. mers disease. Br J Psychiatry 170: 156158.
Shergill S, Mullan E, Dath P et al. (1994). What is the clin- Walker Z, Grace J, Overshot R et al. (1999). Olanzapine in
ical prevalence of Lewy body dementia? Int J Geriatr Psy- dementia with Lewy bodies: a clinical study. Int J Geriatr
chiatry 9: 907912. Psychiatry 14 (6): 459466.
Shinotoh H, Aotsuka A, Ota T et al. (2001). Brain cholinergic Walker Z, Allen RL, Shergill S et al. (2000b). Three years
function in dementia with Lewy bodies and Alzheimers survival in patients with a clinical diagnosis of dementia
disease measured by PET. In: Abstracts. Proceedings of with Lewy bodies. Int J Geriatr Psychiatry 15: 267273.
5th International Conference on Progress in PD and AD: 74. Walker Z, Costa DC, Walker RWH et al. (2004). Striatal
Singleton A, Gwinn-Hardy K (2004). Parkinsons disease dopamine transporter in dementia with Lewy bodies and
and dementia with Lewy bodies: a difference in dose? Parkinson disease: a comparison. Neurology 62 (9):
Lancet 364 (9440): 11051107. 15681572.
Stevens T, Livingston G, Kitchen G et al. (2002). Islington Wesnes KA, McKeith IG, Ferrara R et al. (2002). Effects of
study of dementia subtypes in the community. Br J Psy- rivastigmine on cognitive function in dementia with Lewy
chiatry 180: 270276. bodies: a randomised placebo-controlled international study
Taki J, Yoshita M, Yamada M et al. (2004). Significance of using the Cognitive Drug Research computerised assess-
I-123-MIBG scintigraphy as a pathophysiological indica- ment system. Dement Geriatr Cogn Disord 13 (3):
tor in the assessment of Parkinsons disease and related 183192.
disorders: it can be a specific marker for Lewy body dis- Yoshita M, Taki J, Yamada M (2001). A clinical role for
ease. Ann Nucl Med 18 (6): 453461. I-123 MIBG myocardial scintigraphy in the distinction
Tanaka M, Kim YM, Lee G et al. (2004). Aggresomes between dementia of the Alzheimers-type and dementia
formed by alpha-synuclein and synphilin-1 are cytoprotec- with Lewy bodies. J Neurol Neurosurg Psychiatry 71 (5):
tive. J Biol Chem 279 (6): 46254631. 583588.
Handbook of Clinical Neurology, Vol. 84 (3rd series)
Parkinsons disease and related disorders, Part II
W. C. Koller, E. Melamed, Editors
# 2007 Elsevier B. V. All rights reserved

Chapter 61

Myoclonus and parkinsonism

DANIEL D. TRUONG1* AND ROONGROJ BHIDAYASIRI1,2,3

1
The Parkinsons and Movement Disorder Institute, Fountain Valley, CA, USA
2
Movement Disorders Group, Division of Neurology, Chulalongkorn University Hospital, Bangkok, Thailand
3
Department of Neurology, UCLA Medical Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA

61.1. Introduction mental status abnormalities) guide the physicians to


the appropriate diagnosis and relevant investigations.
Myoclonus is defined as sudden, brief, shock-like, Often, there is clinical and pathological evidence of
involuntary movements caused by muscular contrac- diffuse nervous system involvement. Within the symp-
tions (positive myoclonus) or inhibitions (negative tomatic group, posthypoxic syndrome and neurodegen-
myoclonus) (Fahn et al., 1986). Myoclonus is not a erative disorders (particularly Alzheimers disease
diagnosis but rather a descriptive term denoting symp- (AD) and CreutzfeldtJakob disease (CJD)), account
toms or signs that are non-specific in relation to their for most of the cases (Caviness, 2002). Most myoclo-
neuroanatomical source and pathogenesis. Despite its nus in neurodegenerative disease is classified as corti-
non-specific nature, myoclonus has been described in cal, consistent with the well-known cortical pathology
various conditions and has strong implications for the in AD and CJD. In AD, myoclonus is more likely to
diagnosis, prognosis and treatment of the underlying occur at a late stage when dementia is severe and tends
disorder. to be multifocal, affecting distal muscles, both sponta-
Myoclonus has been classified according to clinical neously and in response to various stimuli (Hauser
presentation, etiology, pathophysiology, pharmacologi- et al., 1986). On the other hand, myoclonus is much
cal and biochemical pathology, with an attempt to more frequent and more likely to be generalized in
establish the relationship between semiology, physiol- CJD, although focal jerking in the face and limb can
ogy and etiology (Marsden et al., 1981). Of these, the be seen initially (Van Everbroeck et al., 2004). The
etiological classification is considered the most helpful rapidly progressive dementia, associated neurological
and appropriate in clinical practice and includes signs, including cortical blindness, cerebellar, parkin-
physiological myoclonus, a normal manifestation in sonism and typical electroencephalographic (EEG)
healthy people; essential myoclonus, where no other discharges, help distinguish CJD from the more com-
neurological deficit is present and can be sporadic or mon AD (Wojcieszek et al., 1994; Tschampa et al.,
inherited; epileptic myoclonus, where epilepsy domi- 2001). When the disease is of longer duration, asso-
nates the clinical picture and myoclonus is considered ciated with myoclonus and rigidity, AD should be
as an epileptic phenomenon; and symptomatic myoclo- considered (Tschampa et al., 2001).
nus, in the setting of an identifiable underlying disorder Myoclonus is a recognized feature of many basal
(Fahn et al., 1986). In a study conducted by Caviness ganglia disorders where parkinsonian signs predomi-
(2002), symptomatic myoclonus was found to be the nate. Although tremor is frequently observed in
most common (72%), followed by epileptic myoclonus patients with idiopathic Parkinsons disease (PD),
(17%) and essential myoclonus (11%). myoclonus is much less common than tremor in par-
In symptomatic myoclonus, the features of myoclo- kinsonian syndromes and the presence of myoclonus
nus may not be distinctive. Instead, the history and usually suggests a less common cause of parkinsonism
associated clinical signs (such as seizures, ataxia and (Caviness et al., 2002a). Klawans et al. (1986) were

*Correspondence to: Daniel D. Truong, The Parkinsons and Movement Disorder Institute, 9940 Talbert Ave, Fountain Valley,
CA 92708, USA. E-mail: dtruong@pmdi.org, Tel: 714-378-5062, Fax: 714-378-5061.
550 D. D. TRUONG AND R. BHIDAYASIRI
Table 61.1 but are not limited to, multichannel surface electro-
myography (EMG) recording with testing for long-
Common causes of myoclonus and parkinsonism
latency EMG responses to nerve stimulation, EEG,
Idiopathic Parkinsons disease EEG-EMG polygraphy with back-averaging and
Related to medications, including levodopa, dopamine somatosensory evoked potentials (SEPs). Since the
agonists, amantadine majority of myoclonic jerks are caused by hyperexcit-
Unrelated to medications ability of groups of neurons in certain brain structures,
Diffuse Lewy body disease the relationship of myoclonic jerks to EEG activities is
Corticobasal degeneration of primary importance in the study of myoclonus
Multiple system atrophy
(Shibasaki, 2000). Jerk-locked back-averaging is an
Encephalitis lethargica
extension of the EEG-EMG polygraph and its princi-
Juvenile-onset Huntingtons disease
Dentatorubral-pallidoluysian atrophy ple is to back-average the simultaneously recorded
Frontotemporal dementia with parkinsonism EEGs with respect to myoclonus. SEPs record EEG
Chronic manganese poisoning potentials time-locked to EMG events such as a
myoclonus EMG discharge.
A good example of the utility of electrophysiologi-
cal studies is the differentiation between myoclonus
in MSA and CBD (Table 61.2). Myoclonus in MSA
the first to report that myoclonus and parkinsonism has been reported to be typical of cortical reflex myo-
occurred simultaneously in three conditions: (1) PD clonus, with enlarged SEPs, exaggerated long-latency
where patients are treated chronically with levodopa; EMG responses to median nerve stimulation and
(2) von Economos encephalitis; and (3) acutely in 1- back-averaged EEG transients (Obeso et al., 1985). In
methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) contrast to MSA, there is no preceding cortical dis-
intoxication. Since then, the association between myo- charge time-locked to action myoclonus in CBD, SEPs
clonus and parkinsonism has been described in other are not enlarged and latency on stimulation of median
parkinsonian disorders, including multiple system nerve at the wrist is shorter (40 versus 50 ms)
atrophy (MSA), corticobasal degeneration (CBD), (Thompson et al., 1994b).
diffuse Lewy body disease (DLB), postencephalitic
parkinsonism and frontotemporal dementia with 61.2. Myoclonus in idiopathic Parkinsons
parkinsonism (FTD) (Table 61.1) (Howard and Lees, disease
1987; Chen et al., 1992; Thompson et al., 1994b; Wen-
ning et al., 1994; Louis et al., 1997; Caviness and Unilateral resting tremor in the distal limb is usually
Wszolek, 2002). In addition, myoclonus has been the first motor manifestation in the majority of patients
recognized in other neurological conditions in which with PD. While tremor, either at rest or during muscle
parkinsonism may occur but is less prominent, such activation, is common in PD, myoclonus is much less
as cerebellar degenerations, Huntingtons disease prevalent and was originally described as a conse-
(HD), dentatorubral-pallidoluysian atrophy (DRPLA) quence of levodopa therapy. In a study of 12 PD
or even chronic manganese poisoning (Rodriguez patients who developed myoclonus after taking levo-
et al., 1994; Thompson et al., 1994a; Thompson and dopa for 12 months, abnormal movements consisted
Shibasaki, 2000; Ono et al., 2002). of single, abrupt jerks of the extremities, which were
Although myoclonus has been reported in various bilateral and symmetrical or occasionally in an arm
parkinsonian syndromes, particularly cortical myoclo- or leg on the same side (Klawans et al., 1975). In gen-
nus in all Lewy body disorders, the mechanism of eral, the trunk and limbs are most commonly affected,
myoclonus is not identical across these different par- whereas facial muscle involvement is unusual. Myo-
kinsonian syndromes and different types of myoclonus clonus can occur during sleep, fatigue, dozing or even
may coexist in the same syndrome (Caviness et al., wakefulness, but abates with levodopa cessation.
2002a, b). Hence, electrophysiological studies play a Luquin et al. (1992) reported spontaneous and action
crucial role in the evaluation of myoclonus in these myoclonus with multifocal distribution in only 6 out
parkinsonian disorders, not only for differential of 168 patients with levodopa-induced dyskinesias,
diagnosis, but also for better understanding of the phy- whereas Marconi et al. (1994) observed sporadic myo-
siological mechanism underlying each type of myoclo- clonic jerks in most patients (10 out of 15 patients)
nus. Indeed, certain patterns have some etiological during the 1020 minutes following absorption of
specificity. The specific methods used in the neuro- levodopa. Methylsergide, a serotonin-specific antago-
physiological study of myoclonus usually include, nist, has been reported to improve levodopa-induced
MYOCLONUS AND PARKINSONISM 551
Table 61.2
Electrophysiological findings in various subtypes of myoclonus

Back-averaged
Types EEG findings EMG EEG transients SEPs Reflex responses

Cortical Variable, may show Bursts typically Variable but focal Enlarged  C reflex at rest
epileptiform less than 75 ms sharp-wave 1040 cortical
discharges and ms before jerks component
slow waves
Cortical Generalized spike Bursts < 100 ms Time-locked Enlarged  C reflex at rest
subcortical and wave association cortical
component
possible
Subcortical No consistent Variable burst No correlate Normal Sometimes reflex
supraspinal abnormalities properties response to
sound
Spinal Normal Bursts > 100 ms No correlate Normal Variable but can
and  rhythmic be very short in
latency
Peripheral Normal Variable burst, No correlate Normal Normal
may be
denervation

EEG, electroencephalogram; EMG, electromyogram; SEPs, somatosensory evoked potentials.


Reproduced from Caviness and Brown (2004), The Lancet Neurology, with permission from Elsevier.

myoclonus, without worsening levodopa-induced tertiary care center (Caviness et al., 1998, 2002b). The
dyskinesias (Klawans et al., 1975). myoclonus in PD in this category is typically of small
In addition to levodopa, other medications have amplitude, occurring during muscle activation, and
been shown to induce myoclonus in PD. The dopa- may seem repetitive and rhythmic enough to resemble
mine agonist bromocriptine was reported to induce action tremor. However, the movement is irregular in
nocturnal myoclonus in 6 PD patients after the levo- both timing and amplitude and thus deserves the term
dopa was discontinued (Vardi et al., 1978). Another myoclonus. In a few cases, frequent (> 6 Hz) repetitive
patient with generalized dystonia from anoxic brain rhythmic trains of EMG discharges coincided with a
injury also developed myoclonus affecting the proxi- movement that could overlap with a tremor phenotype.
mal arms and axial muscles following the increasing The myoclonus is rarely present at rest and reflex
doses of bromocriptine (Buchman et al., 1987). When myoclonus cannot be demonstrated to touch, stretch or
bromocriptine was discontinued, the myoclonus tendon reflexes. The amplitude is often greater on one
improved but the dystonia worsened. Amantadine, a side, but not always the same side as the more prominent
dopamine-reuptake inhibitor, a dopamine agonist and parkinsonian signs.
an N-methyl-D-aspartate (NMDA) receptor agonist, This myoclonus is very different from the levodopa-
can also cause myoclonus and delirium. It was impli- induced myoclonus reported by Klawans et al. (1975)
cated as a cause of vocal myoclonus affecting the because the latter usually occurs at rest, often during
larynx, pharynx and face in a PD patient who was on sleep or drowsiness, and is sensitive to levodopa dosage.
concomitant levodopa (Pfeiffer, 1996). The myoclonus In addition, this small-amplitude myoclonus was present
resolved after discontinuation of amantadine. In addi- even before levodopa therapy in 1 of the 2 patients with
tion, amantadine has also been reported to induce levodopa-responsive parkinsonism (Caviness et al.,
delirium and multifocal myoclonic jerks in all extremi- 1998). Because this type of myoclonus can be observed
ties, neck, tongue and face in another 2 PD patients in early and moderate stages of non-demented PD
(Matsunaga et al., 2001). One patient also had renal patients, advanced parkinsonism or dementia is gener-
impairment with toxic levels of amantadine. ally not considered as a requirement. This is in contrast
Recently, the occurrence of myoclonus unrelated to to asterixis observed in PD, which is associated with
medication has been recognized, with the incidence of signs of chronic drug toxicity, dementia and its reversal
approximately 5% in non-demented PD patients in a following drug withdrawal (Glantz et al., 1982).
552 D. D. TRUONG AND R. BHIDAYASIRI
Electrophysiological studies of myoclonus in PD 1997). The source of myoclonus in DLB is cortical
reported by Caviness et al. (2002b) are quite different and previously published series of myoclonus in PD
from the cortical reflex myoclonus physiology. EMG showed electrophysiological characteristics that are
recording during muscle activation showed irregular, indistinguishable from the DLB series, even though
multifocal, brief (< 50 ms) myoclonic EMG dis- myoclonus in DLB is clinically more severe (Caviness
charges. Back-averaging consistently showed a focal, et al., 2002b, 2003a). In addition, myoclonus seen in
short-latency, EEG transient prior to the myoclonic PD and DLB also showed many properties in common
EMG discharges. Furthermore, cortical SEPs were with the myoclonus that was previously reported in
not enlarged and long-latency EMG responses at rest individuals with hereditary DLB (Caviness et al.,
were not present. Corticomuscular coherence is also 2000). However, the electrophysiological properties
increased in PD patients with small-amplitude myoclo- of myoclonus in Lewy body disorders differ from
nus (Caviness et al., 2003b). Indeed, these electrophy- those of myoclonus in AD.
siological findings distinguish myoclonus in PD from The fact that cortical myoclonus shares similar phy-
the myoclonus seen in MSA, CBD, AD, minipolymyo- siological properties across a spectrum of Lewy body
clonus and cortical tremor. The mechanisms for the disorders suggests that a unifying mechanism may be
cortical myoclonus in PD are probably secondary to responsible for its occurrence. It is at present unclear
the lack of inhibitory influences or excessive excita- if the abnormal basal ganglia output to the cortex, cor-
tion of the sensorimotor cortex produced by the neuro- tical pathology itself (either within the sensorimotor
degeneration locally around the sensorimotor cortical region or more diffusely in the motor circuits in the
region (Caviness et al., 2002b). frontal and parietal lobes) or both in some way facili-
tate cortical myoclonus. Compared to the pathology
61.3. Myoclonus in diffuse Lewy body disease in PD, there is more widespread cortical pathology in
DLB, which may explain why myoclonus in DLB is
Myoclonus in DLB is similar to myoclonus in PD, more severe. However, the neuropathological examina-
although the former is more clinically impressive, with tion of previously reported cases only showed occa-
larger amplitude, and is much more likely to occur at sional Lewy bodies in the pre- and postcentral gyri
rest (Fig. 61.1) (Caviness, 2002, 2003). Myoclonus as well as parietal, cingulate and entorhinal cortex
in DLB is also more common, occurring in about (Caviness et al., 1998). Furthermore, there is no correla-
1520% of cases (Burkhardt et al., 1988; Louis et al., tion between cortical myoclonus and the severity of

LEFT DELT

20.00 mV

LEFT BICEP


0.00 0.25 0.50 0.75 1.00 1.25 1.50 1.75 2.00
S
Fig. 61.1. Rectified surface electromyogram (EMG) of left deltoid and left biceps during the finger-to-nose maneuver in a
patient with dementia with Lewy bodies. The EMG burst in the center of the figure produced a myoclonic jerk. Courtesy of
Dr. John Caviness, Mayo Clinic, Scottsdale, AZ.
MYOCLONUS AND PARKINSONISM 553

RFCU

850 mV

100 ms

RFDI

Fig. 61.2. Polygram in a patient with corticobasal degeneration demonstrating spontaneous myoclonus. Two bipolar surface
electromyogram channels are shown. RFCU, right flexor carpi ulnaris; RFDI, right first dorsal interosseous. Courtesy of
Dr. Zoltan Mari, Human Motor Control Section, NINDS.

parkinsonism in DLB. It is unclear if cortical myoclonus cutaneous stimuli (Fig. 61.2). However, it may be
is a marker for cortical dysfunction in DLB and is related masked by increased muscle tone. Action and reflex
to the presence of cognitive dysfunction. It is possible myoclonus are characteristic (Thompson and Shibasaki,
that cortical pathology in DLB exerts some influence 2000). The presence of spontaneous myoclonus is diffi-
on the abnormal basal ganglia output to the cortex. cult to judge because of the action myoclonus in the
setting of rigidity and alien-limb movements, which
61.4. Myoclonus in corticobasal degeneration interfere with the ability to complete relaxation.
Attempts to move the limb voluntarily are interrupted
The most frequent clinical presentation in CBD is a pro- by trains of repetitive-action myoclonus.
gressive levodopa-unresponsive akinetic-rigid syn- The myoclonus in CBD is focal in distribution and
drome associated with both cortical and basal ganglia predominantly distal. They are stimulus-sensitive and
dysfunction (Kompoliti et al., 1998). A clinical diagnos- closely resemble those of cortical reflex myoclonus
tic criterion has been proposed to include manifestations (Fig. 61.3) (Riley et al., 1990; Chen et al., 1992;
reflecting dysfunction of the cortex (corticosensory loss, Thompson et al., 1994b; Carella et al., 1997). Thompson
apraxia, frontal lobe reflexes, hyperreflexia, Babinskis et al. (1994b) studied the clinical and physiological
sign) and basal ganglia (akinesia, rigidity, limb dysto- characteristics of myoclonus in 14 patients with
nia, postural instability) (Riley and Lang, 2000). When CBD, compared to 13 patients with epilepsia partialis
specific signs are absent, the evolution of asymmetric continua and progressive myoclonic ataxia. Although
rigid dystonia and reflex myoclonus is highly sugges- hypersynchronous short-duration bursts of muscle
tive of CBD. At the time of presentation, myoclonus activity are common to both (often comprising runs
of one limb has been reported in one-third of cases of 24 discharges with an interburst interval of
and usually evolves in a further 20% over the following 6080 ms), major differences between myoclonus
2 years as the disease progresses (Rinne et al., 1994). in CBD and typical cortical reflex myoclonus
Overall, approximately 50% of patients with CBD exist (Thompson and Shibasaki, 2000). First of all,
develop myoclonus during the course of illness (Rinne myoclonus in CBD is not generally associated with
et al., 1994). The tremor in CBD is usually of 68 Hz enlarged or giant SEPs, in contrast to typical cor-
action postural and action tremor, more irregular and tical reflex myoclonus. Secondly, cortical potentials
also jerky (Stover et al., 2004). A jerky tremor may pre- of myoclonus in CBD do not precede each myoclonic
cede the appearance of myoclonus in the limb early in jerk on back-averaged EEG. In addition, the latency
the condition (Brunt et al., 1995). of reflex myoclonic jerks is shorter, about 40 ms in
Myoclonus is best demonstrated during action or CBD, in contrast to 5060 ms in typical cortical
maintenance of a posture in distal muscles; it is also reflex myoclonus. Lastly, the estimates of the cortical
stimulus-sensitive in most cases, easily elicited by delay for generation of myoclonic activity in typical
554 D. D. TRUONG AND R. BHIDAYASIRI

Lt biceps br.

triceps br.

rhomboid

Rt biceps br.

triceps br.

rhomboid

500 mV
1 sec
Fig. 61.3. Electromyogram polygraph obtained from a patient with clinical diagnosis of corticobasal ganglionic degeneration.
Repetitive spontaneous discharges are seen in the right biceps and triceps muscles almost synchronously at an approximate rate
of 78 Hz. Courtesy of Dr. Hiroshi Shibasaki, Kyoto University, Kyoto, Japan, reprinted with permission from Elsevier.

cortical reflex myoclonus associated with giant SEPs dramatic inflated EMG-EMG coherence in the absence
(6.9  3.7 ms) are significantly longer than those of myo- of any evidence of a pathological corticospinal drive
clonus in CBD (1.4  0.8 ms) (Thompson et al., 1994b). as determined by EEG-EMG coherence, raising the
Different patterns of abnormality in cortical reflex myo- possibility of involvement of subcortical motor sys-
clonus may be explained by the differences in the proces- tems in the myoclonus of CBD. Since the latency of
sing and relay of sensory information in thalamocortical reflex myoclonus in CBD is only 12 ms longer than
pathways. In addition, other factors, such as fatigue, if the sum of afferent and efferent times to and from
present, may influence the occurrence of periodic the cortex, it is therefore possible that myoclonus in
discharges in the cortex and muscles in patients with CBD is due to the direct sensory input to motor corti-
CBD (Manto et al., 2000). cal areas that activate corticospinal tract output
The combination of focal, predominantly distal, (Thompson et al., 1994b). Alternatively, prominent
hypersynchronous jerks, evidence of enhanced cortical parietal involvement in CBD could result in a loss of
excitability, together with the known pathology in inhibitory input from the somatosensory area to the
CBD suggests that the myoclonus in these patients motor cortex or thalamic pathology, possibly resulting
may be cortical in origin, despite the fact that back- in a loss of thalamocortical recurrent inhibition (Shafiq
averaging fails to detect a cortical correlate to sponta- and Lang, 2002).
neous or action-induced jerks and giant SEPs are Shafiq and Lang (2002) provided their personal
seldom found (Thompson et al., 1994b; Monza et al., experience of evaluating 1 patient with CBD whose
2003). Furthermore, the study involving magnetoence- reflex latencies were consistent with typical cortical
phalogram also supported the role of precentral cortex reflex myoclonus and other patients with a CBD phe-
in generating spontaneous myoclonus (Mima et al., notype whose reflex myoclonus also did not have
1998). Grosse et al. (2003) evaluated EEG-EMG and classical electrophysiological features of CBD,
EMG-EMG coherence and phase in 5 patients with described by Thompson et al. (1994b). Whether there
clinically probable CBD and unilateral, action-induced are electrophysiological features that are specific and
stimulus-sensitive myoclonus. All patients exhibited sensitive for the pathology of CBD in order to confirm
MYOCLONUS AND PARKINSONISM 555
or exclude the diagnosis remains uncertain and more and silent periods of variable duration and amplitude
studies are needed to address this issue. and electrical stimuli elicited synchronized reflex
response of the type observed in reflex myoclonus.
61.5. Myoclonus in multiple system atrophy Despite the direct neurophysiological evidence of
reflex myoclonus in these patients, authors cannot
MSA encompasses a wide spectrum of clinical presen- firmly conclude the cortical origin of the myoclonus,
tations with various combinations of extrapyramidal, since no EEG spikes and no cortical activity time-
pyramidal, cerebellar and autonomic signs and symp- locked to the EMG bursts were obtained (Salazar
toms (Quinn, 1989). Since a statement on the diagnosis et al., 2000). In a separate study, typical cortical reflex
of MSA was published in 1998 (Gilman et al., 1998a, b), myoclonus in the olivopontocerebellar form of MSA is
definite MSA is restricted to patients with postmor- well recognized and characterized by stimulus-
tem pathological confirmation. A possible diagnosis sensitive myoclonus with enlargement of the P25-N33
may be conferred in patients with either parkinsonism component of the cortical SEPs (Rodriguez et al.,
with poor levodopa-responsiveness or cerebellar par- 1994). Photic cortical reflex myoclonus with general-
kinsonism. A probable diagnosis applies to patients ized muscle jerks may also occur in both olivopontocer-
presenting with autonomic failure, urinary dysfunc- ebellar and striatonigral forms of MSA (MSA-C and
tion and poor levodopa-responsive parkinsonism or MSA-P, respectively) (Artieda and Obeso, 1993).
cerebellar dysfunction. Although myoclonus is not Electrophysiological studies recorded a large (36.9
recognized as a feature in the diagnostic criteria, it mV) positive/negative wave maximum in the mid
is increasingly recognized as a common manifestation frontal region following visual stimuli (3.8 ms later)
in MSA. Indeed, involuntary movements of the hands preceding the myoclonus in a time-locked fashion.
and fingers during posture and action were described in The myoclonus markedly improved following levodopa
1 of the 3 original cases by Adams et al. (1964). Such and piracetam.
movements are usually referred to as jerky tremor In addition to a cortical origin on various forms of
by some authors and at least two series have reported myoclonus in patients with MSA, Clouston et al.
an upper-extremity small-amplitude jerky postural tre- (1996) evaluated 1 patient with non-dopa-responsive
mor in 20% and 55% of cases (Wenning et al., 1994; parkinsonism who exhibited early autonomic symp-
Colosimo et al., 1995; Gouider-Khouja et al., 1995). toms, a marked sleep disturbance and myoclonus with
Myoclonus predominantly occurs in the parkinsonian- a brainstem origin that was both spontaneous and
type MSA (MSA-P), although stimulus-sensitive induced by somatosensory stimuli. No giant SEPs
myoclonus can also occur in cerebellar-type MSA and no preceding EEG activity were recorded. In addi-
(MSA-C) and the literature is split as to whether this tion, reflex latencies were compatible with a brainstem
stimulus-sensitive myoclonus is more prevalent in myoclonus and there was no evidence of photically
MSA-P or MSA-C (Obeso et al., 1985; Rodriguez induced myoclonus following discontinuation of levo-
et al., 1994; Wenning et al., 1994; Salazar et al., dopa. The mechanism of brainstem myoclonus is
2000). Wenning et al. (1994) reported that myoclonus thought to be disruption of the lower brainstem reticu-
was present in 37% of those with MSA-P and 6% of lar formation, which may also explain abnormal sleep
those in MSA-C, whereas Obeso et al. (1985) found that architecture in this patient (Hallett et al., 1977).
stimulus-sensitive myoclonus occurred more frequently Furthermore, hyperexcitability of the cortex and brain-
in patients with MSA-C. However, 92% of their stem has been reported in a patient with MSA-P, who
patients exhibited bradykinesia and 50% had rigidity. exhibited postural and action myoclonus (Kofler et al.,
Salazar et al. (2000) proposed the term minipolymyo- 1998). Interestingly, facial and whole-body jerks in
clonus to describe jerky tremor in 82% of patients response to tapping the nose, consistent with exagger-
with MSA-P. ated startle response, indicating an underlying brainstem
Clinical observation showed that patients with dysfunction, were also observed. Although isolated
MSA-P had abnormal involuntary movements predo- cortical reflex myoclonus is frequently encountered in
minantly in the hands and fingers with maintenance patients with MSA, the authors emphasize that exagger-
of a posture or at the beginning of an action. Clini- ated startle response secondary to brainstem hyperexcit-
cally, these movements were jerky, irregular and not ability can also occur in a case of definite MSA (Kofler
entirely rhythmic. Neurophysiological evidence also et al., 2000). The authors further hypothesized that
favored these movements as a form of postural and brainstem atrophy with loss of pontine nuclei and glial
reflex myoclonus (Salazar et al., 2000). The EMG cytoplasmic inclusion deposition in the reticular forma-
activity recorded in the forearm and hand muscles tion may be responsible for startle responses in their
was not usually regular but made of synchronous burst case (Kofler et al., 2000).
556 D. D. TRUONG AND R. BHIDAYASIRI
61.6. Myoclonus in encephalitis lethargica following peripheral nerve stimulation had a latency of
approximately 40 ms, similar to CBD, and were unlike
Encephalitis lethargica (von Economos encephalitis) typical cortical reflex myoclonus.
appeared suddenly in 19171918 and was pandemic Caviness and Kurth (1997) reported the electrophy-
for the next 10 years (von Economo, 1931). Since siological findings of myoclonus in a patient with HD
then, no further epidemics have occurred and the exact who was studied postoperatively following a bilateral
etiology has never been identified. However, rare fetal cell transplant in the striatum. Incomplete transi-
cases resembling von Economos encephalitis contin- ent improvement was seen in the myoclonus, followed
ued to be reported (Howard and Lees, 1987; Blunt by gradual deterioration. The myoclonus itself was
et al., 1997). Although the manifestations were pro- consistent with typical cortical reflex myoclonus with
tean, three main clinical presentations have been enlarged SEPs, unlike those reported by Thompson
described (Howard and Lees, 1987). The commonest et al. (1994a). The reason for the transient improvement
variety, the somnolent-ophthalmoplegic form, started of the myoclonus is unclear, but this study raised the
with an influenza-like illness, which was followed by possible role of basal ganglia in the modulation of
increasing drowsiness and confusion, progression to cortical myoclonic activity.
continuous sleep, stupor and finally coma. In addition
to external ophthalmoplegia, oculogyric crisis and nys- 61.8. Myoclonus in dentatorubral-
tagmus, a smaller number of patients presented with pallidoluysian atrophy
bradykinesia and mutism, whereas a third hyperkinetic
group developed extreme motor restlessness, dyskine- DRPLA is a rare autosomal-dominant neurodegenera-
sia and myoclonus. In fact, Klawans et al. (1986) tive disorder, characterized by various combinations
included von Economos encephalitis as one of the of cerebellar ataxia, chorea, myoclonus, epilepsy, par-
three conditions in which myolonus and parkinsonism kinsonism, dementia and psychiatric symptoms (Naito
occurred. They reported that adrenocorticotropic hor- and Oyanagi, 1982). After the gene was identified in
mone relieved all symptoms the first time it was given 1994, DRPLA became known as one of the CAG
and twice more brought about significant amelioration repeat expansion diseases, in which the responsible
of all symptoms, especially the myoclonus. In general, gene is located on chromosome 12p, and its product
myoclonic movements tended to occur at onset and is called atrophin-1 with prominent anticipation
often subsided as the patient recovered (McAlpine, (Koide et al., 1994; Nagafuchi et al., 1994). The condition
1920). However, myoclonus persisted indefinitely in has mainly been described in Japan (Kanazawa, 1998).
some patients (Rail et al., 1981). Myoclonus is a distinctive feature of some families with
Although characterized at the time as a separate myoclonus epilepsy phenotype of this condition,
entity, epidemic hiccup may have been another form although uncommon (Thompson, 2002). Although a
of myoclonus in encephalitis lethargica. Hiccups cortical source is likely for the myoclonus because of
occurred sporadically in small epidemics during the the epileptiform activity on the EEG, a detailed electro-
1920s and were sometimes one of the first symptoms physiological examination of the myoclonus has not been
hailing the onset of encephalitis lethargica (Loewe reported.
and Strauss, 1919).
61.9. Myoclonus in frontotemporal dementia
61.7. Myoclonus in Huntingtons disease with parkinsonism

Myoclonus is uncommon in HD, but recognized in a FTD, linked to chromosome 17 (FTD-17), is asso-
young-onset akinetic-rigid form when it is often accom- ciated with tau protein mutations (Foster et al.,
panied by seizures (Gambardella et al., 2001; Thompson, 1997). Pallidopontonigral degeneration (PPND) is
2002). Myoclonus and seizures may occur in the early one example of a FTD-17 syndrome (Wszolek et al.,
stages of juvenile HD and dominate its clinical picture. 1992; Caviness and Wszolek, 2002). The clinical
The myoclonus is usually stimulus-sensitive and induced manifestations include frontal lobe behavior, psychia-
by action (Thompson et al., 1994a). Thompson et al. tric problems and memory loss. Motor symptoms
(1994a) studied 3 patients with young-onset HD who associated with FTD-17 consist of akinetic-rigid par-
were the offspring of an affected father. Neurophysiolo- kinsonian features without resting tremor, dystonia,
gical studies documented generalized and multifocal spasticity and occasional amyotrophy. Most patients
action myoclonus of cortical origin that was strikingly harboring exon-10 missense and intronic mutations
stimulus-sensitive, without enlargement of the cortical in the tau gene develop a parkinsonian phenotype.
SEPs. Furthermore, reflex myoclonus in hand muscles Myoclonus is rarely seen in FTD-17 kindreds but
MYOCLONUS AND PARKINSONISM 557
has been reported with the N279K, P301S and manganese poisoning (Pal et al., 1999; Ono et al.,
V337M tau mutations (Caviness and Wszolek, 2002; 2002). Ono et al. (2002) reported a 17-year-old welder
Tsuboi et al., 2002). with myoclonic involuntary movements involving the
Not all affected individuals with PPND develop right upper and lower extremities who showed ele-
myoclonus. Myoclonus is invariably present in the vated levels of manganese in the blood and hair and
upper extremities of rather small, subtle and action- characteristic MRI abnormalities. Electrophysiological
induced, without detectable activation with rest or studies indicated absence of giant SEPs or paroxys-
stimuli (Caviness and Wszolek, 2002). Caviness and mal discharge in jerk-locked EEG averaging. Chelation
Wszolek (2002) observed that clinical progression is therapy with Ca-EDTA (EDTA, ethylene diamine tetra
more advanced for the myoclonus cases than for those acetic acid) improved the myoclonus and MRI abnor-
without myoclonus, although the explanation is malities. Although the exact mechanism of myoclonus
unclear. Previous EMG studies reported muscle in chronic manganese poisoning is uncertain, authors
activation tremor-like discharges in 3 subjects and proposed that subcortical neural circuits via globus
definite myoclonic discharges in 1 (Wszolek et al., pallidus might have been involved (Ono et al., 2002).
1998). Further study demonstrated at least two dif- Since Klawans et al. (1986) first reported 3
ferent myoclonus physiological patterns in PPND, patients in whom the onset of parkinsonian signs
associated with the N279K tau mutation (Caviness was clinically associated with myoclonic movements,
et al., 2003c). The absence of a back-averaged more conditions have been recognized to encompass
EEG transient characterized the myoclonus physiol- a variety of atypical parkinsonian syndromes as well
ogy associated with disease progression, whereas as recently identified neurodegenerative disorders.
pre-myoclonus EEG transient was present in the In addition to clinical descriptions of myoclonus,
myoclonus that occurred in one of the individuals more advances have been made in electrophysiologi-
with stage 0 (presymptomatic but gene-positive) cal and molecular studies to understand better the
(Caviness et al., 2003c). However, the physiology pathophysiology, etiology, neuroanatomy and possi-
of myoclonus may change over the course of the ble therapeutic options of various subtypes of myo-
disorder, reflecting progressive cortical pathology clonus. In the setting of patients with predominant
and other brain regions. So far, the exact physiology parkinsonian features, the occurrence of myoclonus
of myoclonus in this disorder is still unclear, although should alert physicians to the possibility of many dif-
cortical origin has been proposed, based on the find- ferent neurodegenerative disorders, not limited to PD.
ings of the cortical transient back-averaged to the Although the use of clinical neurophysiology helps in
myoclonus in one of the at-risk individuals. defining the nature of the EMG discharges in such
disorders, the current ability of these techniques to
61.10. Myoclonus in chronic manganese provide absolute specificity and sensitivity is still
poisoning lacking. Therefore, further studies in various parkin-
sonian disorders are needed to clarify neurochemical,
Manganese is a paramagnetic heavy metal that is electrophysiology, molecular pathophysiology and
widely distributed in the environment, in air, water neural networks of myoclonus, which will hopefully
and food. It has long been appreciated that manganese lead to the development of therapeutic targets in
exposure can cause neurotoxicity and certain brain these progressive disorders.
areas, including globus pallidus and substantia nigra
pars reticulata, are preferentially affected (Yamada Acknowledgments
et al., 1986). Substantia nigra pars compacta and stria-
tal dopamine are relatively spared. The most common Roongroj Bhidayasiri (UK) is supported by the Lilian
neurological manifestations of chronic manganese Schorr Postdoctoral Fellowship of the Parkinsons Dis-
poisoning are extrapyramidal syndromes, especially ease Foundation (PDF).
parkinsonism, comprising bradykinesia, rigidity and Daniel D. Truong is supported by the Parkinsons
postural instability (Olanow, 2004). Magnetic reso- and Movement Disorder Foundation and the Long
nance imaging (MRI) studies in manganese-intoxi- Beach Memorial Foundation.
cated patients demonstrated a characteristic signal
abnormality on T1-weighted images that are not seen References
in normal individuals or PD patients or other forms
of parkinsonism (Olanow, 2004). Adams RD, Vanbogaert L, Vandereecken H (1964). Strao-
In addition to parkinsonism, chorea, tremor and nigral degeneration. J Neuropathol Exp Neurol 23:
myclonus have been described in patients with chronic 584608.
558 D. D. TRUONG AND R. BHIDAYASIRI

Artieda J, Obeso JA (1993). The pathophysiology and phar- Clouston PD, Lim CL, Fung V et al. (1996). Brainstem
macology of photic cortical reflex myoclonus. Ann Neurol myoclonus in a patient with non-dopa-responsive parkin-
34: 175184. sonism. Mov Disord 11: 404410.
Blunt SB, Lane RJ, Turjanski N et al. (1997). Clinical fea- Colosimo C, Albanese A, Hughes AJ et al. (1995). Some
tures and management of two cases of encephalitis specific clinical features differentiate multiple system
lethargica. Mov Disord 12: 354359. atrophy (striatonigral variety) from Parkinsons disease.
Brunt ER, van Weerden TW, Pruim J et al. (1995). Unique Arch Neurol 52: 294298.
myoclonic pattern in corticobasal degeneration. Mov Disord Fahn S, Marsden CD, Van Woert MH (1986). Definition and
10: 132142. classification of myoclonus. Adv Neurol 43: 15.
Buchman AS, Bennett DA, Goetz CG (1987). Bromocrip- Foster NL, Wilhelmsen K, Sima AA et al. (1997). Frontotem-
tine-induced myoclonus. Neurology 37: 885. poral dementia and parkinsonism linked to chromosome
Burkhardt CR, Filley CM, Kleinschmidt-DeMasters BK et al. 17: a consensus conference. Conference Participants.
(1988). Diffuse Lewy body disease and progressive demen- Ann Neurol 41: 706715.
tia. Neurology 38: 15201528. Gambardella A, Muglia M, Labate A et al. (2001). Juvenile
Carella F, Ciano C, Panzica F et al. (1997). Myoclonus in Huntingtons disease presenting as progressive myoclonic
corticobasal degeneration. Mov Disord 12: 598603. epilepsy. Neurology 57: 708711.
Caviness JN (2002). Epidemiology of myoclonus. Adv Neurol Gilman S, Low P, Quinn N et al. (1998a). Consensus state-
89: 1922. ment on the diagnosis of multiple system atrophy. Ameri-
Caviness JN (2003). Myoclonus and neurodegenerative can Autonomic Society and American Academy of
diseasewhats in a name? Parkinsonism Relat Disord Neurology. Clin Auton Res 8: 359362.
9: 185192. Gilman S, Low PA, Quinn N et al. (1998b). Consensus state-
Caviness JN, Brown P (2004). Myoclonus: current concepts ment on the diagnosis of multiple system atrophy. J Auton
and recent advances. Lancet Neurol 3: 598607. Nerv Syst 74: 189192.
Caviness JN, Kurth M (1997). Cortical myoclonus in Glantz R, Weiner WJ, Goetz CG et al. (1982). Drug-induced
Huntingtons disease associated with an enlarged asterixis in Parkinson disease. Neurology 32: 553555.
somatosensory evoked potential. Mov Disord 12: Gouider-Khouja N, Vidailhet M, Bonnet AM et al. (1995).
10461051. Pure striatonigral degeneration and Parkinsons disease:
Caviness JN, Wszolek ZK (2002). Myoclonus in pallido- a comparative clinical study. Mov Disord 10: 288294.
ponto-nigral degeneration. Adv Neurol 89: 3539. Grosse P, Kuhn A, Cordivari C et al. (2003). Coherence
Caviness JN, Adler CH, Newman S et al. (1998). Cortical analysis in the myoclonus of corticobasal degeneration.
myoclonus in levodopa-responsive parkinsonism. Mov Mov Disord 18: 13451350.
Disord 13: 540544. Hallett M, Chadwick D, Adam J et al. (1977). Reticular
Caviness JN, Gwinn-Hardy K, Adler CH et al. (2000). Elec- reflex myoclonus: a physiological type of human post-
trophysiological observations in hereditary parkinsonism- hypoxic myoclonus. J Neurol Neurosurg Psychiatry 40:
dementia with Lewy body pathology. Mov Disord 15: 253264.
140145. Hauser WA, Morris ML, Heston LL et al. (1986). Seizures
Caviness JN, Adler CH, Beach TG et al. (2002a). Myoclonus and myoclonus in patients with Alzheimers disease.
in Lewy body disorders. Adv Neurol 89: 2330. Neurology 36: 12261230.
Caviness JN, Adler CH, Beach TG et al. (2002b). Small- Howard RS, Lees AJ (1987). Encephalitis lethargica.
amplitude cortical myoclonus in Parkinsons disease: A report of four recent cases. Brain 110 (1): 1933.
physiology and clinical observations. Mov Disord 17: Kanazawa I (1998). Dentatorubral-pallidoluysian atrophy or
657662. Naito-Oyanagi disease. Neurogenetics 2: 117.
Caviness JN, Adler CH, Caselli RJ et al. (2003a). Electro- Klawans HL, Goetz C, Bergen D (1975). Levodopa-induced
physiology of the myoclonus in dementia with Lewy myoclonus. Arch Neurol 32: 330334.
bodies. Neurology 60: 523524. Klawans HL, Tanner CM, McDermott J (1986). Myoclonus
Caviness JN, Adler CH, Sabbagh MN et al. (2003b). Abnor- and parkinsonism. Clin Neuropharmacol 9: 202205.
mal corticomuscular coherence is associated with the Kofler M, Wenning GK, Poewe W (1998). Cortical and brain
small amplitude cortical myoclonus in Parkinsons stem hyperexcitability in striatonigral degeneration. Mov
disease. Mov Disord 18: 11571162. Disord 13: 602607.
Caviness JN, Tsuboi Y, Wszolek ZK (2003c). Clinical- Kofler M, Wenning GK, Poewe W et al. (2000). Cortical
electrophysiological correlation of tremor and myoclonus and brain stem hyperexcitability in a pathologically con-
in a kindred with the N279K tau mutation. Parkinsonism firmed case of multiple system atrophy. Mov Disord 15:
Relat Disord 9: 151157. 362363.
Chen R, Ashby P, Lang AE (1992). Stimulus-sensitive Koide R, Ikeuchi T, Onodera O et al. (1994). Unstable
myoclonus in akinetic-rigid syndromes. Brain 115 (6): expansion of CAG repeat in hereditary dentatorubral-
18751888. pallidoluysian atrophy (DRPLA). Nat Genet 6: 913.
MYOCLONUS AND PARKINSONISM 559

Kompoliti K, Goetz CG, Boeve BF et al. (1998). Clinical Quinn N (1989). Multiple system atrophythe nature of the
presentation and pharmacological therapy in corticobasal beast. J Neurol Neurosurg Psychiatry (Suppl): 7889.
degeneration. Arch Neurol 55: 957961. Rail D, Scholtz C, Swash M (1981). Post-encephalitic
Loewe L, Strauss I (1919). Etiology of epidemic (lethargic) Parkinsonism: Current experience. J Neurol Neurosurg
encephalitis: Preliminary note. JAMA 73: 10561057. Psychiatry 44: 670676.
Louis ED, Klatka LA, Liu Y et al. (1997). Comparison of Riley DE, Lang AE (2000). Clinical diagnostic criteria. Adv
extrapyramidal features in 31 pathologically confirmed Neurol 82: 2934.
cases of diffuse Lewy body disease and 34 pathologically Riley DE, Lang AE, Lewis A et al. (1990). Cortical-basal
confirmed cases of Parkinsons disease. Neurology 48: ganglionic degeneration. Neurology 40: 12031212.
376380. Rinne JO, Lee MS, Thompson PD et al. (1994). Corticobasal
Luquin MR, Scipioni O, Vaamonde J et al. (1992). Levodopa- degeneration. A clinical study of 36 cases. Brain 117 (5):
induced dyskinesias in Parkinsons disease: clinical and 11831196.
pharmacological classification. Mov Disord 7: 117124. Rodriguez ME, Artieda J, Zubieta JL et al. (1994). Reflex
Manto MU, Jacquy J, Van Bogaert P et al. (2000). Rhythmic myoclonus in olivopontocerebellar atrophy. J Neurol Neu-
cortical and muscle discharges induced by fatigue in cor- rosurg Psychiatry 57: 316319.
ticobasal degeneration. Clin Neurophysiol 111: 496503. Salazar G, Valls-Sole J, Marti MJ et al. (2000). Postural and
Marconi R, Lefebvre-Caparros D, Bonnet AM et al. (1994). action myoclonus in patients with parkinsonian type multi-
Levodopa-induced dyskinesias in Parkinsons disease phe- ple system atrophy. Mov Disord 15: 7783.
nomenology and pathophysiology. Mov Disord 9: 212. Shafiq M, Lang AE (2002). Myoclonus in parkinsonian dis-
Marsden CD, Hallett M, Fahn S (1981). The nosology and orders. Adv Neurol 89: 7783.
pathophysiology of myoclonus. In: CD Marsden, S Fahn Shibasaki H (2000). Electrophysiological studies of myoclo-
(Eds.), Movement Disorders. Butterworth Scientific, nus. Muscle Nerve 23: 321335.
London, pp. 196248. Stover NP, Wainer BH, Watts RL (2004). Corticobasal
degeneration. In: RL Watts, WC Koller (Eds.), Movement
Matsunaga K, Uozumi T, Qingrui L et al. (2001). Amanta-
Disorders. McGraw-Hill Medical Publishing, New York,
dine-induced cortical myoclonus. Neurology 56: 279280.
pp. 763778.
McAlpine D (1920). A case of myoclonic form of acute
Thompson PD (2002). Neurodegenerative causes of myoclo-
epidemic encephalitis with recovery. Lancet 2: 353354.
nus. Adv Neurol 89: 3134.
Mima T, Nagamine T, Ikeda A et al. (1998). Pathogenesis of
Thompson PD, Shibasaki H (2000). Myoclonus in corticoba-
cortical myoclonus studied by magnetoencephalography.
sal degeneration and other neurodegenerations. Adv Neu-
Ann Neurol 43: 598607.
rol 82: 6981.
Monza D, Ciano C, Scaioli V et al. (2003). Neurophysiolo-
Thompson PD, Bhatia KP, Brown P et al. (1994a). Cortical
gical features in relation to clinical signs in clinically
myoclonus in Huntingtons disease. Mov Disord 9:
diagnosed corticobasal degeneration. Neurol Sci 24:
633641.
1623.
Thompson PD, Day BL, Rothwell JC et al. (1994b). The
Nagafuchi S, Yanagisawa H, Sato K et al. (1994). Dentatoru- myoclonus in corticobasal degeneration. Evidence for
bral and pallidoluysian atrophy expansion of an unstable two forms of cortical reflex myoclonus. Brain 117 (5):
CAG trinucleotide on chromosome 12p. Nat Genet 6: 11971207.
1418.
Tschampa HJ, Neumann M, Zerr I et al. (2001). Patients with
Naito H, Oyanagi S (1982). Familial myoclonus epilepsy and Alzheimers disease and dementia with Lewy bodies mis-
choreoathetosis: hereditary dentatorubral-pallidoluysian taken for Creutzfeldt-Jakob disease. J Neurol Neurosurg
atrophy. Neurology 32: 798807. Psychiatry 71: 3339.
Obeso JA, Rothwell JC, Marsden CD (1985). The spectrum Tsuboi Y, Baker M, Hutton ML et al. (2002). Clinical and
of cortical myoclonus. From focal reflex jerks to sponta- genetic studies of families with the tau N279K mutation
neous motor epilepsy. Brain 108 (1): 193224. (FTDP-17). Neurology 59: 17911793.
Olanow CW (2004). Manganese-induced parkinsonism and Van Everbroeck B, Dobbeleir I, De Waele M et al. (2004).
Parkinsons disease. Ann N Y Acad Sci 1012: 209223. Differential diagnosis of 201 possible Creutzfeldt-Jakob
Ono K, Komai K, Yamada M (2002). Myoclonic involuntary disease patients. J Neurol 251: 298304.
movement associated with chronic manganese poisoning. Vardi J, Glaubman H, Rabey JM et al. (1978). Myoclonic
J Neurol Sci 199: 9396. attacks induced by L-dopa and bromocryptin in Parkinson
Pal PK, Samii A, Calne DB (1999). Manganese neurotoxi- patients: a sleep EEG study. J Neurol 218: 3542.
city: a review of clinical features, imaging and pathology. von Economo C (1931). Encephalitis Lethargica: Its Seque-
Neurotoxicology 20: 227238. lae and Treatment. Oxford University Press, London.
Pfeiffer RF (1996). Amantadine-induced vocal myoclonus. Wenning GK, Ben Shlomo Y, Magalhaes M et al. (1994).
Mov Disord 11: 104106. Clinical features and natural history of multiple system
560 D. D. TRUONG AND R. BHIDAYASIRI

atrophy. An analysis of 100 cases. Brain 117 (4): pallido-ponto-nigral degeneration. Electroencephalogr
835845. Clin Neurophysiol 107: 213222.
Wojcieszek J, Lang AE, Jankovic J et al. (1994). What is Wszolek ZK, Pfeiffer RF, Bhatt MH et al. (1992). Rapidly
it? Case 1, 1994: rapidly progressive aphasia, apraxia, progressive autosomal dominant parkinsonism and demen-
dementia, myoclonus, and parkinsonism. Mov Disord 9: tia with pallido-ponto-nigral degeneration. Ann Neurol 32:
358366. 312320.
Wszolek ZK, Lagerlund TD, Steg RE et al. (1998). Clinical Yamada M, Ohno S, Okayasu I et al. (1986). Chronic manga-
neurophysiologic findings in patients with rapidly nese poisoning: a neuropathological study with determina-
progressive familial parkinsonism and dementia with tion of manganese distribution in the brain. Acta
Neuropathol (Berl) 70: 273278.
Index
Page numbers in italic, e.g. 96, refer to figures. Page numbers in bold, e.g. 435, denote tables.

acetylcholine, 1023, 121, 128, 142, antiglutamatergic drugs (Continued ) bilateral striopallidodentate calcinosis,
335, 540, 542 amatadine (Continued ) 479, 480, 481, 4825, 483, 485
acetylcholine esterase, 102, 141, 335, for prevention of disease boxers encephalopathy, see
522, 542 progression, 1301 parkinsonism: posttraumatic:
adenosine, 21, 142, 146, 1901 side-effects, 131 repetitive head trauma
aging for symptoms of parkinsonism, 129 bradykinesia
eye movement, 434 budipine, 128, 1313 in Alzheimers disease, 364, 406, 439
gait abnormality, 432 dextromethorphan, 128, 132, 143 in corticobasal disease, 353, 356, 357
motor memantine, 1278, 1312, 202 essential tremor and, 436
abnormalities, 435 remacemide, 21, 128, 133 in Japanese B encephalitis, 379
slowing, 429 riluzole, 21, 128, 1334, 202 MPTP-induced, 390
Parkinsons disease and, 4379 antioxidants for neuroprotection in in mixed movement disorders, 515
posture, 4312 Parkinsons disease, 1922 myoclonus and, 5557
tone change, 4301 apomorphine, see dopamine agonist(s): in old age with parkinsonism, 4289,
tremor disorders, 4357 apomorphine 429, 432
akathisia, 45, 169, 1734, 224, 226, apoptosis, 18, 22, 44, 103, 104, 105, in pallidonigroluysian degeneration,
399400, 401, 402, 406, 107, 127, 1389, 297, 382, 450
40911 3901, 393, 409, 491 in Parkinsons disease and
Alzheimers disease, 358 apraxia, 357 parkinsonism, 17, 77, 159,
and Parkinsons disease, 439 equilibrium, 462 243, 292, 310, 315, 330, 364,
dystonia and, 516, 522 gait, 4334, 434, 438, 4612 385, 390, 393, 399, 401, 417,
Lewy bodies and, 404, 446 ideomotor, 3556, 461 4278, 430, 434, 462, 5013,
mixed movement disorders kinetic, 356, 461, 474, 476 503
in, 522 armantadine, 143 in progressive supranuclear palsy,
a-synuclein and, 53940 332, 343
with parkinsonism, 364, 439, 516 measurement, 162, 404
aminoindane, 96, 107 ballooned neuron (BN), 336, 352, mechanisms, 2434, 263, 422, 438
amphetamine 1920, 96, 105, 107, 110, 358, 358, 360, 361, 380, 445, in posttraumatic parkinsonism, 493,
139, 165, 228, 2801, 2959, 460, 465 495
3889, 410, see also basal ganglia in Rett syndrome, 453
methamphetamine calcification therapy, 7
amyloid precursor protein, 106 epidemiology, 47981 treatment
amyotrophic lateral sclerosis- etiology, 480 deep brain stimulation, 267, 269
parkinsonism dementia complex history, 479 drug, 51, 101, 111, 112, 129,
of Guam, 336, 3923 hypoparathyroidism and, 480, 1445
antiapoptotic proteins, 105 480 surgical, 249, 2502, 254
anticholinergic drugs pathophysiology, 4845 transplantation, 282, 287
in clinical practice, 1213, 142, 320, treatment, 485, 485 in tuberculosis, 376
401, 411, 419 circuitry, 1923, 5089 bradyphenia, 327
effectiveness, 31819 neurophysiology, 1967 brofaromine, 94
side effects, 406 Bcl-2 family proteins burden of diseases, 1512
tremor and, 123 antiapoptotic (Bcl-2), 105 buspirone, 143
mechanism, 1212 proapoptotic (Bad, Bax), 105 BW 137OU87, 94
antiglutamatergic drugs, 1278 befloxatone, 94
amatadine, 12831 beta-methylaminoalanine (BMAA),
for motor complications, 1301 3923, 392 cabergoline, 378, 48, 735, 74, 78,
pharmacology, 1289 BIA-3-202, 141 7884, 80, 86, 137, 164, 191, 200
562 INDEX
carbon monoxide, 3934, 419, 480 deep brain stimulation (DBS), 205 depression (Continued )
caroxazone, 94 adverse effects, 26970 measurement scales for, 7, 111
catecholamines, 31, 34, 103, 315, 422, clinical efficacy, 266 in multiple system atrophy, 314
437 comparison, 2689 in Parkinsons disease, 10, 40, 45,
catechol-O-methyltransferase inhibitors pallidal, 266, 2667 81, 86, 99103, 108, 114, 129,
(COMT) subthalamic, 267, 267 169, 16970, 226, 26970,
clinical effects, 55, 56 thalamic, 266 331, 4034, 429, 502, 505,
inhibition, 54 magnetic resonance imaging, 263 511, 534
entacapone, 548, 567, 75, 83, methodology in progressive supranuclear palsy,
98, 102, 112, 113, 1646, 437 safety issues, 265 330, 340, 3412
tolcapone, 548, 567, 801, 102, surgical preparation, 2634 non-parkinsonian, 81
1645, 437 target location, 264, 2645 therapy resistant, 100
and MAO inhibitors, 1023 patient selection, 2702 as treatment side-effect, 229, 2312,
side-effects, 56, 568 advanced Parkinsons disease, 2701
Charcot, Jean-Martin, 31, 121, 327, 2712 treatment, 94, 945, 170
351, 487, 495 tremor, 271 dextromethorphan, 143
chlorpromazine, 399, 408, 521 physiological mechanisms, 2613 diffuse Lewy body disease (DLBD), see
choline acetyltransferase, 541 systemic, 2623 dementia: with Lewy bodies;
chorea acanthocytosis, 447, 44950, thalamic, 262 Lewy body disease
51718 dementia DJ-1, 336, 512, 512
clorgyline, 94, 96 aging and, 434 DL-dopa, 31, 35
clozapine, 143 in corticobasal degeneration, 357 donepezil, 141, 341, 542
Cockaynes syndrome, 480 frontotemporal, 153, 364, 508, 512, dopamine
coenzyme A, 446, 520 5567 agonists, see dopamine agonist(s)
corticobasal degeneration chromosome 17 and, 332, 3389, antagonists, 2001, 400
history, 3513 351, 353, 3567, 3601, 363, biosynthesis, 32, 323
diagnosis, 3534 381, 4456, 512, 513, 550, 556 degradation, 32, 323
cognitive and neuropsychiatric, parkinsonism and, 513, 550, 556 -enhancing agents, see catechol-O-
3567, 357 with Lewy bodies (DLB), 364 methyltransferase inhibitors
cortical symptoms, 3556, 357 antiparkinsonian agents for, 541 loss, 422, 430, 435, 439
differential, 364 antipsychotic agents for, 543 monoamine oxidase-B inhibitors,
motor symptoms, 3545, 357 cholinergic system and, 540 534
electrophysiological studies, 3623 cholinesterase inhibitors for, 542 pharmacology, 313
epidemiology, 353 clinical features, 5324 metabolization, 323
imaging studies, 3612 clinical significance of, 532 synthesis and storage, 312
laboratory studies, 363 cognition and, 5323 receptors, 24, 34, 42, 735, 82, 98,
myoclonus and, 550 diagnostic features, 5346, 128, 1613, 167, 188, 191,
parkinsonism and, 512 5367 203, 280, 400, 409, 422, 511,
pathology differential diagnosis, 53641 522, 540
ballooned neurons in, 358, 360 dopaminergic system and, 540 release, 3, 76, 146, 280, 294, 394
macroscopic, 358, 361 etiology, 5389 reuptake, 32, 121, 140, 341, 551
microscopic, 358, 3589, 361 genetics of, 540 signaling, 382
tau, 35960 hallucinations in, 533 structure, 34
tau mutations and ultrastructure, magnetic resonance imaging transporters, 34, 24, 32, 50, 77, 98,
3601, 361 of, 538 140, 298, 309, 340, 362, 390,
therapy management, 541 404, 420, 473, 492, 504, 510,
for cognitive and motor parkinsonism and, 534 536, 537, 538
neuropsychiatric symptoms, myoclonus and, 550 working memory and, 540
364 pathology, 53840, 53940 dopamine agonist(s), 37
for gastrointestinal symptoms, prevalence and incidence, 531 apomorphine, 39, 47, 73, 74, 78, 80,
365 relationship to dementia in 81, 83, 856, 134, 137,
for motor symptoms, 363 Parkinsons disease, 531, 538 1445, 162, 1678, 172,
for orthostatic hypotension, 365 sleep disorders, 5334 174, 191, 198, 201, 2023,
for urinary symptoms, 365 deprenyl, 94 280, 2923, 298, 320, 3412,
Creutzfeldt-Jakob disease, 286, 332, depression 392, 474
355, 380, 364, 535, 549 in corticobasal degeneration, 357, 357 bromocriptine, 74, 78, 80
cystercercosis, 480 in dementia with Lewy bodies cabergoline, see cabergoline
cytokines, 140, 374, 387, 491 536, 540 clinical trials with, 7682
INDEX 563

dopamine agonist(s) (Continued ) dyskinesia (Continued ) dystonia (Continued )


clinical trials with (Continued ) antidyskinetic drugs (Continued ) neuronal intermediate filament
levodopa-nave Parkinsons glutamatergic, 1424, see also inclusion disease and, 513
patients, 769 glutamatergic: antagonists Parkinsons disease and, 512, 51315
motor complication prevention, biochemistry and molecular biology, inherited, 512
7980 18894 postencephalitic, 51213
non-motor complication dopaminergic receptors, 188 sporadic, 51112
prevention, 81 early genes, 191 pathophysiology, 50810
dihydroergocryptine, 74, 78, 80 GABA receptors, 191, 193 primary pallidal degeneration and,
efficacy, 7380, 80, 856 glutamatergic receptors, 188200 513
as levodopa adjunct, 79 neuropeptides and adenosine progressive supranuclear palsy and,
lisuride, 74, 78, 80 receptors, 1903 513
for neuroprotection in Parkinsons serotonergic receptors, 1934 rapid-onset (DYT12), 515
disease, 256, 76 choreic and dystonic movements, 186
Parkinsons disease management deep brain stimulation (DBS), 205
with, 846 diphasia, 45 encephalitis lethargica, 327, 373,
pergolide, see pergolide diphasic, 186, 1867 376, 37980, 399, 513, 550,
piribedil, 76, 78, 80 dopamine agonists and, 81 556
pharmacology, 73 entacapone, 57 Epstein-Barr virus (EBV), 373, 381
pharmacodynamics, 756 incidence in Parkinsons disease, 48 excitotoxicity, 17, 20, 24, 43, 76, 127,
pharmacokinetic properties, 73, levodopa-induced (LID), 445, 143, 1389, 385, 484
74 160, 186, 205
receptor sensitivity, 756 clinical presentation, 1858
pramipexole, 74, 78, 80 symptomatic drugs for, 143 Fahrs disease, see basal ganglia:
receptor D2 agonists, 98 surgery for, 2078 calcification
ropinirole, see ropinirole morphological basis, 1979 fibroblast growth factors 107, 287, 294,
safety issues, 82 neurophysiology, 196, 1967 see also growth factors
synthesis, 98 nocturnal myoclonus, 187 fludrocortisones, 39, 320, 365
type A adverse reactions non-dopaminergic drugs for, 1457 fluoxetine, 143
central, 823 noradrenergic drugs, 1445 FTDP-17, see dementia:
peripheral, 82 adenosine receptors and, 146 frontotemporal: chromosome 17
type B adverse reactions, 834 serotonergic, 146 and
dopaminergic synaptic vesicular proteins and, frontotemporal dementia, see dementia:
drugs, 40, 42, 59, 140, 145, 168, 1467 frontotemporal
1701, 187, 191, 196, 197, off-period, 186, 1867
198, 2034, 2056, 222, 228, pathophysiology
269, 399, 406, 420, 433, see classic model, 194 GABA, see gamma-aminobutyric acid
also dopamine agonist(s); synaptic plasticity, 1956 gait apraxia, 4334, 434, 438, 4612
dopamine: antagonists surgery, 2048, 244 gama-aminobutyric acid (GABA)
neurons, 21, 23, 26, 32, 40, 489, 96, treatment cells, 107, 130, 132, 18991, 194,
103, 140, 146, 159, 285, 291, pharmacological, 199204 205, 243, 453, 509, 51011,
2956, 338, 374, 379, 385, surgery, 2048, 244 513
38792, 394, 417, 421, 491, without benefit, 187 receptors, 48, 163, 191, 193, 514
513, 515 dysphagia, 340, 357, 365 genes immediate early (IEG), 409, 496
systems, 3, 19, 36, 42, 74, 96, 127, dystonia gene therapy for Parkinsons disease
161, 174, 2001, 335, 386, ablative surgery for, 244 drug development strategy, 292
388, 394, 4034, 422, 448, in cerebral toxoplasmosis, 378 glial-derived neurotrophic factor
494, 507, 538, 540 chlorpromazine and, 399 (GDNF) delivery in, 2958,
Downs syndrome, 480 classification, 5078 299300
drug-induced movement disorders in corticobasal degeneration, 357 neurorestoration in
(DIMD), 399400, 403, 4056, dopamine and, 507, 508, 511 6-hydroxydopamine lesioning
40911 dopa-responsive (Segawa disease), and, 2978
DU-127090, 141 515 MPTP and, 298
dysarthria, 35, 24952, 269, 307, 313, genetically defined, 514 neuroturin, 299
315, 319, 333, 340, 3526, 357, in HIV/AIDS, 378 therapeutic enzyme delivery in, 2924
364, 447, 451, 482, 495, 515, lubag (DYT3), 51315 trophic molecule delivery in, 2945
51718, 519, 520 mixed movement disorder and 516, viral vectors
dyskinesia see also mixed movement adeno-associated virus, 293
antidyskinetic drugs, 1425 disorders adenovirus, 293
564 INDEX
gene therapy for Parkinsons disease hydrocephalus (Continued ) lesions (Continued )
(Continued ) normal-pressure (NPH) (Continued ) hemorrhagic, 418, 487
viral vectors (Continued ) clinical features, 4601, 461 nigrostriatal, 61, 294, 37981, 406
herpes simplex virus, 293 cognitive changes in, 4623 striatum, 474
lentivirus, 293, 299300 drugs, 4701 structural, in parkinsonism, 459, 471,
glial cell line-derived neurotrophic history, 459 4746, 475, 504, 5078, 510
factor (GDNF), 4, 234, 106, imaging, 4634, 4635, 467, 476 thalamic, 229, 251, 262, 265, 474,
107, 292, 293, 2949, 300, 301 incontinence and, 462 476
glial cells, 4, 33, 49, 96, 287, 300, 337, mechanisms, 4657 white-matter, 394, 41920, 422, 464,
374, 382, 446 shunting, 46970 536
globus pallidus, 21, 48, 99, 127, 142, treatment, 467, 4689 levodopa
163, 196, 229, 229, 243, 247, hydroxyaminoindane, 96 cheese reaction and, 95
2623, 296, 298, 315, 3346, hyperhomocysteinemia, 434, 422 dopamine synthesis and, 98
353, 360, 394, 418, 4467, hyperreflexia, 31213, 315, 330, 353, aging and, 434
44951, 474, 476, 47980, 508, 356, 357, 376, 4523, 4601, dopa-cecarboxylase inhibition and,
509, 511, 513, 520, 557 466, 494, 517, 533 345
glutamate hypoparathyroidism, 479, 480, 480 dyskinesias and, 448, 48, 186, 197
antagonists, see antiglutamatergic hypotension, see orthostatic early versus late L-dopa
drugs hypotension (ELLDOPA) studies, 35, 35
neurotransmitter function, 21, 127 hypothyroidism, 480 history of use, 31
neuroprotection by, 21 motor complications, 45
receptors, 128, 163, 18891 clinical presentation, 468
release, 99, 133, 142, 294, 511, see idazoxan, 143 effects, 37
also antiglutamatergic drugs influenza A virus, 374, 375, 379 fluctuations, 448
toxicity, 21, 103, 127, 484 insulin-like growth factor, 287, 309, see mechanisms, 468
glutamatergic also growth factors prevalence, 47
antagonists, 142, 143, 202, see also iproniazid, 94 onset, 46
antiglutamatergic drugs iron in neuroprotection for Parkinsons
neurons, 21, 127, 196, 509 in Parkinsons disease, 18, 389 disease, 25
pathways, 127, 188, 194, 208, 509, deposition, 31516, 409, 421, 4467, side-effects, 36, 38, 3944
511 484, 51718, 520 hallucinations, 38, 3941
receptors, 1278, 18890, 196 metabolism, 447, 520 hyperhomocysteinemia, 43
see also glutamate neurodegeneration and, 44, 4468 impulse control deficits, 412
growth factors oxidative stress and, 103
melanoma, 423
fibroblast, 107, 287, 294 transport, 409 nausea, 36, 38
insulin-like, 287, 309 isocarboxazid, 94 orthostatic hypotension, 389
nerve, 26, 105, 107, 138, 134, 280, istradefylline, 141, 146 sleep disturbances, 38
391 somnolence, 38
Guam, amyotrophic lateral sclerosis- withdrawal rate for adverse
Japanese B encephalitis virus, 374, 379
parkinsonism dementia complex events, 38
of, 336, 3923 therapy strategies
KF-17837, 141 infusion, 512
oral formulations, 501
Hallervorden-Spatz syndrome, 446, slow-release preparations, 523
480 lazabemide, 94 tolerance, 3644
Helicobacter pylori, 374, 376, L-dopa, see levodopa treatment of Parkinsons disease,
3801 lead intoxication, 480 356, 445, 141
hemiparkinson-hemiatrophy (HPHA) learning, 7, 265, 379 Lewy bodies
syndrome, 4489 long-term potentiation and, in Alzheimers disease, 404, 446
HIV-AIDS, 2923, 3734, 3756, 163, 195 degeneration, 3940
3778, 407, 480 memory and, 234 dementia with, see dementia: with
hippocampus, 1023, 163, 195, 352, motor, 195 Lewy bodies
3578, 419, 463, 491, 532 spatial, 102 in Parkinsons disease, 427
Huntingtons disease, 21, 23, 355, 362, testing, 232, 233, 252 Lewy body disease, 81, 102, 114, 363,
405, 447, 450, 482, 504, 508, leprosy, 381 427, 436, 4389, 531, 537, 550,
515, 516, 550, 556 lesions 550, 552
hydrocephalus basal ganglia, 419, 420 locus ceruleus, 39, 96, 99, 101, 103,
neurological features, 466, 4667 brainstem, 41819 307, 315, 352, 35860
normal-pressure (NPH), 459 globus, 474 lymphocytes, 373, 391, 447
INDEX 565
manganese mitochondria(l) (Continued ) monoamine oxidase-AB inhibitors
parkinsonism induction by, 38990, see also oxidative stress (Continued )
504, 550 mixed movement disorders reversible, 94, 96, 142
poisoning, 550, 550, 557 in Alzheimers disease, 522, see also structure, 96
melanoma, 423, 105, 112 Alzheimers disease substrates, 97
melevodopa, 140, 141 autosomal-dominant see also specific drugs
mental changes from Parkinsons Huntingtons disease, 515, 516 monoamine oxidase-B inhibitors
disease treatment Huntingtons disease-like 2, for depression, 94
drug-induced psychosis, 219, 220 51516, 516 discovery of, 935
with clear sensorium, 220 autosomal-recessive, 519 distribution, 95, 97
hallucinations, 2202, 221 aceruloplasminemia, 51819 irreversible, 94
with impaired sensorium/ chorea acanthocytosis, 518 for Parkinsons disease, 1922,
dementia, 220 Gaucher disease type III, 5201 989, 10812
long-term outcome, 225 glutaric aciduria, 521 reversible, 94
treatment, 2225 GM1 gangliosidosis, 520 structure, 94, 96
mood and anxiety fluctuations, 2256 Niemann-Pick type C, 520 substrates, 97
repetitive/compulsive disorders, pantothenate kinase-associated tyramine metabolism inhibition
2267 neurodegeneration, 520 by, 95
dopamine dysregulation Wilsons disease, 518, 519 tyrosine metabolism and, 98
syndrome, 227 biotin-responsive basal ganglia see also specific drugs
gambling, pathological, 228 disease, 521 motor fluctuations in Parkinsons
punding, 228 dentatorubropallidoluysian atrophy, disease
surgery-induced, 229 516, 517, 550, 556 epidemiology, 161
adverse cognitive effects, 2324 drug-induced, 521 levodopa-induced, 15960, 160
anxiety, 232 dystonia and, 516 dose-related, 160
apathy, 232 Fahrs disease, 516, 517 dyskinesia, 160
depression, 2312 homocystinuria, 521 motor blocks, 160
executive function changes, 234 neuroferritinopathy, 517 on-off fluctuations, 160
learning and memory disorders, neuronal intranuclear hyaline tachykinetic problems, 160
234 inclusion disease, 521 treatment, 1648
mania, 2301 parkinsonism and, 516 unpredictable sudden offs, 15960
neuropsychological tests for, 233 spinocerebellar ataxia, 516, 51617 pathophysiology, 1613
psychosis, 229 therapy, 522 pharmacodynamics, 1623
visuospatial function changes, X-linked recessive, 517 pharmacokinetics, 1612
2345 McLeod syndrome, 51718 spontaneous, 161
methamphetamine, 96, 98, 1067, 110, moclobemide, 94, 95, 97 see also non-motor fluctuations in
280, 410 monoamine oxidase, 94 parkinsonism
methanol, parkinsonism induction monoamine oxidase inhibitors, see MPTP (1-methyl-4-phenyl-1,2,3,6-
by, 393 monoamine oxidase-A inhibitors; tetrahydropyrodine)
methemoglobinopathies, 480 monoamine oxidase-AB -induced parkinsonism, 18, 391
methotrexate, 480 inhibitors; monoamine oxidase-B lesions, 4, 23, 142, 144, 163, 173,
midodrine, 39, 320, 365 inhibitors 190, 294, 298, 300, 3901
milacemide, 94 monoamine oxidase-A inhibitors toxicity, 18
mitochondria(l) for depression, 94 -treated animals, 21, 48, 56, 102,
abnormalities, 17, 104 discovery of, 935 144, 188, 293, 391
bioenergetics, 21 distribution, 95, 97 multiple system atrophy
cell survival/death and, 105 irreversible, 94, 96 categories, 314
complex in Parkinsons disease, 99101 clinical features, 31014, 315
I, 48, 139, 338, 387, 3912, 394, reversible, 94, 97 behavioral disorders, 314
410 structure, 96 cardiovascular dysfunction, 312
II, 392, 410 substrates, 97 cerebellar dysfunction, 313
dysfunction, 21, 44, 127, 327, 337, tyramine metabolism inhibition disordered sleep, 31314
385 by, 95 dystonia, 310, 402, 4334, 508,
function, 212, 1045, 107, 139, tyrosine metabolism and, 98 512, 513
3857 see also specific drugs gastrointestinal dysfunction, 312
genes and inheritance, 337, 517 monoamine oxidase-AB inhibitors genital dysfunction, 311
membrane, 212, 1045, 107, 480 for depression, 94 parkinsonism, 310
myopathies, 480 irreversible, 94, 96, 102 pyramidal dysfunction, 313
566 INDEX
multiple system atrophy (Continued ) neurofibromatosis, 480 neurotransplantation for Parkinsons
clinical features (Continued ) neuronal death, 105, 139, 373, 484 disease (Continued )
thermoregulatory dysfunction, 313 mechanism, 104 alternative tissue sources for, 286
urinary dysfunction, 31112 models, 767 donor neurons for, 2856
consensus criteria, 314, 315 prevention, 104 dopamine neurons and, 286
diagnosis propargylamines and, 104 effects of, 283
autonomic function tests, 317 see also neuroprotection in experimental basis for, 27981
differential, 364, 380, 438, 451 Parkinsons disease fluorodopa positron emission
functional neuroimaging, 31617 neuroprotection in Parkinsons disease tomography evaluation of, 283
neurophysiological testing, 31718 amyloid precursor protein and, 106 human fetal dopamine cell, 2825
structural neuroimaging, 316 drugs nialamide, 94
epidemiology antiapoptotic agents, 212 nitric oxide, 338, 374, 491, 511
analytical, 3089 antiexcitotoxic agents, 201 Nocardia asteroides, 374, 380, 382
descriptive, 308 antioxidants, 1822 non-motor fluctuations in parkinsonism
history, 3078 CEP-1347, 138 autonomic symptoms, 1713
glial cytoplasmic inclusions, 316 development status of, 138 dysphagia, 172
myoclonus and, 550, 555 dopamine agonists, 256, 1402 dyspnea, 172
neuropathology E-2007, 138 management, 173
macroscopic, 31415, 421 GPI-1485, 138 orthostatic hypotension, 171
microscopic, 315 leteprinim, 138 sphincter function, 172
parkinsonism and, 512 levodopa 25, 140, see also thermoregulation, 171
sonographic brain images, 421 levodopa classification, 168, 169
treatment liatermine, 138 epidemiology, 169
of cardiovascular dysfunction, MITO-4509, 138 neuropsychiatric, 16971
31920 monoamine oxidase-B inhibitors, mood, 16970
of gastrointestinal dysfunction, 1822 psychotic symptoms, 1701
320 ONO-2506, 138 sensory, 173
of genital dysfunction, 319 PAN-408, 138 akathisia, 173
of motor disorders, 31819 PYM-50028, 138 dysthesia, 173
of urinary dysfunction, 319 propargylamine, 106 management, 174
myoclonus, 357 rasagiline (Azilect, Agilect), 103, pain, 173
causes, 550 106, 11012 noradrenaline, 95
chronic manganese poisoning and, selegiline, 103, 106, 109 NS-2330, 141
557 sonic hedgehog protein agonist,
classification, 549 138
corticobasal degeneration and, 553, TCH-346, 138 olanzapine, 143
5535 V-10367, 138 1-methyl-4phenyl-1,2,3,6-
dentatorubropallidoluysian atrophy zydis selegine, 110, 139, 165 tetrahydropyridine, see MPTP
and, 556 mitochondrial modulation, 13940 orthostatic hypotension, 34, 36, 38, 39,
in diffuse Lewy body disease, antiapoptotic kinase inhibitors, 812, 98, 169, 1712, 220, 225,
5523, 552 13940 308, 31113, 315, 31720, 336,
electrophysiological findings, 551 minocycline, 140 365, 534, 537, 542
encephalitis lethargica and, 556 neuroimmunophilin, 139 oxidative stress, 18, 44, 489, 76, 103,
frontotemporal dementia with neurorescuing agents, 1038 107, 327, 3378, 342, 385, 387,
parkinsonism and, 5567 physical activity and, 3 390, 392, 409
idiopathic Parkinsons disease and, primate model, 297
550 restorative therapies and, 224
in multiple system atrophy, 555 therapies, 13840 pallidonigroluysian degeneration,
monoamine oxidase inhibitors, 4501
1389, see also monoamine pallidopyramidal disease, 4512
nabilone, 143 oxidase-AB inhibitors: palsy
naloxone, 143 irreversible; rasagiline; zydis progressive supranuclear, see
nerve growth factor 26, 105, 107, 138, selegiline progressive supranuclear palsy
139, 280, 391, see also growth therapeutic design, 17 pseudobulbar, 341, 371, 393, 417,
factors neurotransplantation for Parkinsons 419, 423
neurodegeneration with brain iron disease pargyline, 94
accumulation (NBIA), 4468, adrenal medulla, 2812 paralysis agitans, see Parkinsons
517, 520 age and, 285 disease
INDEX 567
parkin gene, 161, 250, 340, 448, 512, Parkinsons disease (Continued ) Parkinsons disease (Continued )
512 GDNF proteins receptors and MPTP (Continued )
Parkinson, James, 31, 351, 436, 487 actions, 4, 23, 105, 139, 281, lesions, 4, 23, 142, 144, 163, 173,
Parkinsons disease 287, 292, 293, 2945, 299 190, 294, 298, 300, 3901
aging and gene therapy, see gene therapy for toxicity, 18
bradykinesia and, 42830 Parkinsons disease -treated animals, 21, 48, 56, 102,
cogwheeling phenomenon, hereditary factors, 385 144, 188, 293, 391
4301, 431, 436, 503 history, see Charcot, Jean-Martin; multiple system atrophy and, see
eye movement, 434 Parkinson, James multiple system atrophy
gait abnormality, 4324, 434 hypersexuality and, 42, 82, 167, 170, myoclonus and, 550
motor slowing, 429 227, 270 neuroprotection in, see
normal, 428 idiopathic, 251, 3801, 406, 488, neuroprotection in Parkinsons
paratonia, 429, 430, 431 4956, 549, 550 disease
primitive reflexes, 4345 imaging, 421 oxidative stress and, see oxidative
tone change, 430, 431 incidence, 385 stress
agricultural chemicals and infectious basis patient advocacy groups (PAGs)
atrazine, 387, 388 animal studies, 3812 advocacy by, 154
maneb, 387, 388 Epstein-Barr virus (EBV), 373, collaboration and partnerships, 152
organochlorine compounds, 388, 381 future, 155
388 Helicobacter pylori, 374, 376, policy and, 1534
paraquat, 387, 388 3801 research issues, 1525
rotenone, 3867, 387 HIV, 3746, 3756, 377, 407, 480 responsible advocacy, 152
Alzheimers disease and, 439 immunology, 3734 pesticide models
animal models, 13, 21, 133, 13940, influenza A virus, 374, 375, 379 maneb, 386, 387, 388
146, 1623, 185, 1889, 194, Japanese B encephalitis virus, paraquat, 386, 387, 388
1989, 262, 279, 293, 2945, 374, 379 rotenone, 21, 44, 3867, 387, 393
297, 386, 391, 394, 509 leprosy, 381 physical exercise and, 38
annonacin and, 393, 393 Nocardia asteroides, 374, 3802 physical therapy for, see physical
anticholinergic drugs, see prion protein, 153, 337, 363, 374, therapy
anticholinergic drugs 381 postural reflexes, 431
antiglutamatergics, see Toxoplasma gondii, 374, 376, progressive supranuclear palsy and,
antiglutamatergic drugs 3778, 378, 407, 480 see progressive supranuclear
atypical, 270, 30910, 318, 328, transmissible spongiform palsy
3346, 351, 353, 355, 363, encephalopathy, 3812 risk factors, 408
421, 501, 536, 557 tuberculosis, 376, 377, 379, 480 alcohol, 2278, 309, 319, 408,
carbon monoxide poisoning and, isoquinoline derivatives and, 103, 4367
3934, 419, 480 338, 3912 coffee and caffeine, 17
cycad toxin and, 392, 3923 iron and, see iron gender, 83, 131, 401, 4067
dementia and, 364, see also metals and, 389 head trauma, 461, 48891, 4967,
dementia copper, 363, 38990, 518 508
drug-induced iron, see iron metals, see Parkinsons disease:
6-hydroxydopamine, 3, 49, 75, manganese, 38990, 504, 550 metals and
103, 185, 280, 292, 297, 386, methanol and, 393 obesity, 1
390, 392 mood disorders, 170, 227, 230, 270 pesticide exposure, see
methanol, 393 anxiety, 45, 108, 131, 153, 160, Parkinsons disease: pesticide
MPTP, see MPTP 169, 16970, 2257, 229, 232, models
6-OHDA, 34, 49, 103, 185, 270, 357, 450, 505, 533, 5423 smoking, 7, 17, 43, 108, 309,
18893, 1956, 1989, 201, apathy, 10, 169, 229, 2312, 270, 386, 406, 488
208, 2801, 285, 2929, 392 314, 330, 333, 334, 357, 399, sensory symptom(s), 169, 1734
reserpine, 31, 75, 201, 399, 408, 411 450, 462, 533, 5423 selegiline monotherapy, 108
see also parkinsonism: drug- depression, see depression: in sleep disorders, 40, 222, 357, 5334,
induced Parkinsons disease 5434
environmental risk factors, 3856 mania, 45, 2278, 229, 2301, 270 speech disorders, 138, 354
excitoxicity and, see excitoxicity motor fluctuations, see motor surgery, see also surgery: ablative
freezing, 5, 17, 19, 45, 109, 160, fluctuations in Parkinsons pallidotomy, 24850, 254
168, 401, 401, 41719, 420, disease radio-, 253
433, 437, 462, 520 MPTP subthalamotomy, 2524
strategies, 1013 -induced parkinsonism, 18, 391 thalamotomy, 2502, 254
568 INDEX
Parkinsons disease (Continued ) parkinsonism (Continued ) physical therapy (Continued )
symptoms, 17, 417, 427 drug-induced (DIP) (Continued ) long-term effects, 78
transplantation for, see treatment, 41011 resistance training, 4
neurotransplantation for frontotemporal dementia and, see sensory cueing and, 56
Parkinsons disease dementia: frontotemporal training techniques, 4
trauma and history, 417, 459 treadmill training, 67
as risk factor, 48891 lesions, see lesions see also physical exercise
epidemiology, 489 mechanism for motor symptoms postencephalitic parkinsonism (PEP),
posttraumatic parkinsonism, see bradykinesia, 2434 3278, 336, 382
parkinsonism: posttraumatic dyskinesia, 244 clinical features, 380
treatment dystonia, 244 epidemiology, 376, 379
gene therapy, see gene therapy tremor, 244 pramipexole (Mirapex), 24, 378,
for Parkinsons disease myoclonus and, see myoclonus 48, 49, 73, 74, 75, 7786,
with levodopa, 356, see also postencephalitic, 512 78, 80, 164, 200, 222, 228,
levodopa posttraumatic, 491, 4934 341, 363
mental changes from, 21928, central lesions, 491 prion protein/disease, 153, 337, 363,
see also mental changes from peripheral lesions, 4956 374, 381
Parkinsons disease treatment repetitive head trauma, 4945, progressive supranuclear palsy (PSP)
parkinsonism 497 brain parenchyma sonography
amytrophic lateral sclerosis- single head trauma, 4912, 494 (BPS), 335
parkinsonism dementia worsening of parkinsonism, 496 cerebral fluid analysis, 334
complex of Guam, 336, 3923 psychogenic clinical
Alzheimers and, 439 clinical manifestations, 5023 features, 3301, 340
arteriovenous malformation and, description, 501 heterogeneity, 3323
471 diagnosis, 503, 5035 diagnosis
intracerebral hemorrhage, 471 epidemiology, 5012 criteria, 3312, 3323
midbrain cavernoma, 471, 472 natural history, 501 differential, 364
brain imaging, 4201 prognosis, 505 epidemiology
dystonia and, see also dystonia treatment, 505 age of onset, 329, 329
dopa-responsive (Segawa stroke and, 439 incidence, 328, 328
disease), 515 substantia nigra and, 459 prevalence, 3289, 329
lubag (DYT3), 51315 symptomatic agents, 141 sex ratio, 329, 329
rapid-onset (DYT12), 515 symptoms, 399, 417 familial, 338
drug-induced (DIP) tumors and, 4734 functional imaging, 3356
age/dementia and, 4056 vascular history, 3278
cigarette smoking and, 406 and Parkinsons disease, 4212 magnetic resonance imaging (MRI),
cinnarizine, 4068, 408, 40910, imaging methods, 421 3345
522 history, 417 management
diagnosis, 4035 pergolide, 73, 74, 75, 7781, 78, 80, adrenergic agents, 341
drugs causing/aggravating, 408 834, 86, 164, 222, 228, 341, 363 cholinergic agents, 341
flunarizine, 40910 phenelzine, 94 dopaminergic agents, 341
gender and, 406 physical exercise neurotransmitter replacement
genetics and, 406 metabolic effects, 37 and, 340
history, 399400 neuroprotective effects, 37 non-pharmacological, 342
HIV and, 407 and Parkinsons disease, 810, 89 palliative treatments, 340
magnetic resonance spectroscopy, see also physical therapy natural history, 331
405 physical therapy neuropsychological assessment,
neuroleptics and, 409 for advanced Parkinsons disease 3334, 336
pathology, 4034 discipline and, 10 nortriptyline and, 341
pre-existing movement disorders freezing episodes, 1013 pathology
and, 406 appendicular and axial stretch, 6, molecular, 33840
prevalence, 4001 89 neuro-, 336, 337
progression of, 4023 attentional strategies, 56 tau aggregation and cell death,
schizophrenia subtype and, 407 compensation strategies, 1013 3378
serotonin reuptake inhibitors ambulation assistive devices, 13 parkinsonism and, 512
and, 410 freezing episodes, 1013 propargylamine, 1056
symptoms, 401, 4012 home environment modification, 13 propranolol, 143
ventricular/brain ratio, 407 sensory cues, 11 proteosome inhibition, 104
INDEX 569
protein SKF-39392, 201 surgery
kinase, 107, 390 sleep disorders, 40, 222, 357, 5334, ablative
synthesis, 1045 5434 microelectrode localization, 246
translation, 22, 342 SLV-308, 141, 142 physiological localization, 245
pseudohypoparathyroidism, 480 somatostatin (SOM), 320, 363, 484 radiological localization, 245
psychogenic movement disorders speech stereotactic lesioning techniques,
(PMD), 501, 503 in bilateral striopallidodentate 246
calcinosis, 4823 stereotactic surgical techniques,
in chorea acanthocytosis, 449 245
quinpirole, 201 in corticobasal degeneration, 352, for dyskinesia, 2048, 244
3546, 365 deep brain stimulation (DBS), 205
in dementia with Lewy body disease, of globus pallidum, 204
rasagiline, 20, 53, 94, 98, 1046 533 subthalamic nucleus, 2047
adverse effects, 111 in GM2 gangliosidosis, 520 for dystonia, 244
developmental status, 141 in pantothenate kinase-associated mental changes following, 229, 2325
effects neurodegeneration, 520 for Parkinsons disease, see
longterm, 112 in Parkinsons disease, 3, 138, 229, Parkinsons disease: surgery
shortterm, 94, 98104, 1046, 401, 429, 447 radio-, 253
1078, 111, 113, 1389 in progressive supranuclear palsy, speech impairment from, 204, 224,
efficacy, 113, 98, 165 331, 340, 340 246, 2503, 269
structure, 96 surgery and, 204, 224, 246, 2503, a-synuclein
see also monamine oxidase-B 269 abnormalities and mutations, 297,
inhibitors SPD-473, 141 340, 351, 512, 531, 540
reactive oxygen species (ROS), 385 SR-57667, 141 Alzheimers disease and, 53940
Rett syndrome, 4524 Steele-Richardson-Olszewski degradation, 316
reversible monoamine oxidase (MOA)- syndrome, see progressive expression, 315
A inhibitors supranuclear palsy in glial cytoplasmic inclusions and
RIMA, 97, 99, 101, 114 substantia nigra pars compacta (SNc or Lewy bodies, 307, 315, 316,
moclobemide, 94, 945, 978, SNpc) 359, 374, 381, 385, 388,
99102, 108, 112, 114 in corticobasal degeneration, 361 5389, 539
ritanserin, 143 deep brain stimulation and, 2634, metals and, 38990
ropinirole (Requip) 270 neuroprotection and, 138
dyskinesias, 48 degeneration, 17, 103, 107, 159, 280, and synucleinopathies, 307, 313,
metabolism, 74 287, 307, 315, 352, 361, 390, 330, 340, 351, 357, 359, 363,
pharmacological properties, 74 417, 427, 435, 445, 450, 374, 491, 539
treatment, 245, 734, 7783, 78, 4523, 492, 493, 495, 508, systemic lupus erythematosus, 418, 480
80, 856, 222, 228 531, 540
effectiveness, 3738, 49, 75 dopamine synthesis and, 98
rotenone, 21, 44, 3867, 387, 393 neurons, 96, 190, 192, 197, 243, talipexole, 141
rotigotine, 73, 86, 141, 164 285, 291, 373, 385, 3889, tau protein, 3334, 3389, 343, 360,
391 445, 513, 556
oxidative stress and, 48, 76 tolcapone, 57
safanamide, 94, 96, 141, 142 in Parkinsons disease, 18, 21, 99, toloxatone, 94
sarizotan, 143 389, 421, 509, 531, see also toxoplasmosis and Toxoplasma
selegiline substantia nigra pars gondii, 374, 376, 3778, 378,
as levodopa adjunct, 109 compacta: degeneration 407, 480
effects, 94, 1034, 1046, 109, 223, in progressive supranuclear palsy, transcranial ultrasonography, 318, 336,
433 336, 337 342, 405, 4201, 448
neuroprotection, 109 subthalamic nucleus (STN) transmissible spongiform
in Parkinsons disease treatment, deep brain stimulation and, 168, encephalopathy, 3812
1819, 94, 98103, 105, 2056, 229, 261, 2634, 267, transplantation for Parkinsons disease,
1078, 1089, 165, 2678, 285, 449 see neurotransplantation for
341, 438 Parkinson disease and, 20, 229, 243, Parkinsons disease
structure, 96 294 tranylcypromine, 94
serotonin (5-HT), 32, 93, 97, 100, 142, progressive supranuclear palsy and, tremor
143, 146, 170, 227, 232, 364, 336, 337 ablative surgery, 244
382, 391, 400, 410, 453, 543, 550 surgery, 205 aging and, 434
SKF-82958, 191, 201 sumanirole, 141 in corticobasal degeneration, 357
570 INDEX
tremor (Continued ) tyrosine (Continued ) VR-2006, 141
essential, 250, 253, 4046, 4356, hydroxylase, 31, 32, 98, 101, 103,
496, 504 107, 286, 292, 293, 382, 388,
tone testing, 4301 514, 539 working memory, 540
tuberculosis, 376, 377, 379, potentiation of cardiovascular World Health Organization (WHO),
480 effects, 95 151, 155, 271
tuberous sclerosis, 480
tyrosine, 98
cheese reaction and, 95 UCHL-1, 309 zydis selegiline, 110, 139, 165

Potrebbero piacerti anche