Sei sulla pagina 1di 10

EUROPEAN NEUROLOGICAL JOURNAL REVIEW ARTICLE

L-dopa-Induced Dyskinesias in Parkinson’s Disease


Iciar Aviles-Olmos*, Raul Martinez-Fernandez*, and Thomas Foltynie
Affiliation: Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London,
United Kingdom

A B S T R A C T

Levodopa (L-dopa) is the most effective treatment for the relief of the motor symptoms of Parkinson’s disease (PD). Its use is limited by
the development of drug-induced involuntary movements known as dyskinesias, which can appear in approximately 40% of the PD patients
after 5 years of treatment. In recent years we have learned a great deal about the processes involved in the appearance of L-dopa-induced
dyskinesia (LID). Non-physiological pulsatile stimulation of dopamine receptors that occurs as a consequence of the use of short-lasting drugs,
dysfunctional dopamine release, and abnormal postsynaptic dopamine receptor internalization together with disrupted synaptic plasticity all
appear to have direct effects on LID development. Avoiding or delaying dyskinesia development and treating patients in this “advanced”
stage of PD is a challenge and requires considerable expertise and individualization of therapy. The aim of this review is to outline the clinical
phenomenology of LID, describe an overview of what is known about their pathophysiology, and provide guidance in the range of therapeutic
approaches that can be tailored to the individual patient.

Keywords: levodopa-induced dyskinesia (LID), Parkinson’s disease (PD), synaptic plasticity, dysfunctional dopamine release, dopamine receptor
supersensitivity

Correspondence: Iciar Aviles-Olmos, Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement
Disorders, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, United Kingdom. Tel: +44-08451555000 Ext. 18739; e-mail:
i.olmos@ion.ucl.ac.uk

INTRODUCTION basis for preventing/delaying their development and current


and future possible options for their treatment.
The introduction of levodopa (L-dopa) for the treatment of
Parkinson’s disease (PD) in the 1960s dramatically changed the
clinical management and prognosis of PD patients.1–2 L-dopa
crosses the blood–brain barrier and is converted to dopamine TYPES OF DYSKINESIA
through the action of aromatic-L-amino-acid decarboxylase The effectiveness of L-dopa treatment decreases as the dis-
in surviving dopaminergic terminals, and is the most potent ease progresses because of the progressive loss of nigrostriatal
symptomatic agent against the motor symptoms of the disor- neurons. Typically the first sign of this is the gradual return of
der. In most patients, L-dopa treatment leads to a “honeymoon” PD symptoms before the next dose of medication is due. This
period during which the motor symptoms are well controlled. is called “wearing off” and it generally necessitates increases in
However, after 5 years of treatment, approximately 40% of L-dopa dosage or frequency. This phase is frequently accompa-
the patients will develop fluctuations in symptom control in nied by the onset of involuntary LID.
response to the drug, as well as involuntary movements known LID are involuntary choreiform or dystonic (hyperkinetic)
as “L-dopa-induced dyskinesia” (LID).3 These complications movements. They can affect any part of the body but the legs
affect as many as 89% of PD patients after 10 years of L-dopa and neck are most frequently involved. There are three main
treatment.4 subtypes of LID:
Many PD clinicians delay using L-dopa following diagnosis,
in view of concerns that the basal ganglia become “primed” • Peak-dose dyskinesias: these are the most common and early
for LID development even following brief exposure to the drug. appearing LID and occur when L-dopa levels are at their high-
Although the use of alternative treatments has certainly reduced est. These usually consist of stereotypic, choreic, or ballistic
the frequency with which severe LID are seen, there has also movements but occasionally dystonia can also be present.
been an evolution of an “L-dopa phobia” among some clini- They tend to be more easily manageable by patients because
cians and patients, resulting in some instances in unnecessary they occur during periods of good motor control, that is, in
disability through the undertreatment of symptoms.5 This arti- the “on” motor state.6
cle will review the clinical subtypes of LID, the current under- • Off-period dyskinesias: these emerge when L-dopa levels are at
standing of their pathogenesis and will outline the evidence their lowest. These often present with painful leg cramps or


These 2 authors contributed equally to this work.

ENJ 2010; 2:(2). August 2010 91 www.slm-neurology.com


European Neurological Journal

abnormal postures (dystonia) typically in the foot, and most direct and indirect pathways, resulting in abnormal GPi activ-
commonly in the early morning. ity. It was originally thought that the clinical features of PD
• Diphasic dyskinesias: these are the least common and occur were representative of simple increases in the “rate” of activ-
with the rise and fall of L-dopa concentration, disappear- ity of the GPi, which consequently acted as a brake to activity
ing when drug levels achieve a certain threshold, and then in the supplementary motor area by inhibiting the motor tha-
recurring as the drug level falls, before resolving again as lamus (Figure 1B). Using the same logic, it was suggested
parkinsonism returns. These usually present as short-lasting therefore that excessive L-dopa stimulation might induce dysk-
(5–10 minutes) choreiform movements or akathisia. inesia by reduction of the inhibition of thalamocortical neurons
and overactivity of motor cortical areas.20
Risk Factors for LID Development This model however is inconsistent with several clinical and
experimental findings. For example, in the nonhuman pri-
Several risk factors have been established for the earlier onset
mate model of PD, metabolic activity in the ventral anterior
of LID:
and ventrolateral thalamic nuclei, regions of the thalamus that
receive input from the GPi, is significantly decreased rather
Age of the patient: Young-onset PD patients develop LID earlier than increased in LID.21 Furthermore among patients with PD
than older-onset PD patients.7,8 However, far greater num- and LID, creation of a lesion within the GPi (pallidotomy) is
bers of young-onset patients have genetic factors contribut- associated with a reduction in LID rather than a deterioration
ing to the development of PD, and therefore distinguishing in LID that would have been predicted by the rate model.22
the effect of age at the onset of PD from the direct effects
The pathophysiological changes that underlie the develop-
of genetic influences on LID risk is difficult even among
ment of LID are evidently far more complex. The striatal
patients with genetically defined PD.9,10
dopaminergic inputs are closely associated with glutamater-
Dose of L-dopa: In the ELLDOPA study, it was shown that patients
gic corticostriatal projections that synapse onto medium spiny
treated with L-dopa over a 40-week period were signifi-
neurons that form the direct striatal output pathway. Further
cantly less impaired than patients treated with placebo even
modulation of the dopaminergic input onto these cells is medi-
after a 2- to 3-week washout period.11 However, dyskinesias
ated through adenosine A2a inputs and metabotropic gluta-
were seen in 16.5% of patients receiving the highest dose of
mate receptors.23 These medium spiny striatal neurons appear
L-dopa even over this short follow-up period compared with
to be central to LID development. Overlapping inter-related
2–3% of patients on lower doses or placebo, demonstrat-
mechanisms appear to trigger abnormal functioning of these
ing that the dose of L-dopa is critical in terms of risk of
neurons, namely, dysfunctional dopamine release, postsynap-
dyskinesia development.
tic dopamine receptor internalization, and disrupted synaptic
Pattern of L-dopa administration: Intermittent administration of
plasticity.
L-dopa has been shown to increase the risk of LID com-
pared with continuous dopaminergic stimulation (CDS)
in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)
primates12 and in advanced PD patients.13 LID eventually can DYSFUNCTIONAL DOPAMINE RELEASE
even occur in normal primates if administered in pulsatile
As the striatum is progressively denervated in PD, the surviv-
doses of L-dopa.12 It is now widely accepted that strategies
ing dopaminergic neurons sprout branches that successfully
to reduce the pulsatility of L-dopa stimulation will reduce
compensate for the neurodegenerative process until approxi-
the risk of triggering the mechanisms that underlie the
mately 60% of neurons are lost. Until this point, the admin-
development of LID.
istration of L-dopa does not alter the concentration of stri-
Stage of disease: The onset of clinical symptoms of PD likely
atal dopamine, but beyond the 60% deficit, the administration
denotes the point of failure of compensatory mechanisms.14
of L-dopa leads to a 3-fold increase in the concentration of
Advanced nigrostriatal denervation in animal models and
dopamine in the striatum.24 As soon as the striatum cannot
advanced disease in PD patients are both associated with
sufficiently buffer exogenously administered and endogenous
faster onset and more severe LID following the initiation of
DA levels, DA synaptic availability starts to mirror the short
L-dopa treatment.15–19
plasma half-life of L-dopa, and non-physiological dopamine
receptor stimulation follows, contributing to the occurrence of
peak-dose LID. Furthermore, 18F-dopa PET scans have demon-
PATHOPHYSIOLOGY OF LID strated greater impairments in dopamine synthesis, storage,
The neural mechanisms that underlie LID in PD are not com- and release in younger-onset patients, leading to larger swings
pletely understood, but our current insights have been greatly in synaptic DA levels, which may contribute to the more com-
helped through the detailed study of basal ganglia anatomy mon occurrence of LID in younger-onset PD patients.25
and physiology in the normal and dopamine-depleted states. Although L-dopa administration will continue to enhance
Dopamine activates the direct striatal output pathway through dopamine synthesis and release by the surviving dopamin-
D1-type receptors and inhibits the indirect striatal output path- ergic neurons, exogenous L-dopa is also decarboxylated and
way through D2-type receptors (Figure 1A). In PD, the progres- released as dopamine by serotonergic terminals, striatal capil-
sive death of SNc neurons upsets the equilibrium between the laries, noradrenrgic neurons, and non-aminergic striatal

ENJ 2010; 2:(2). August 2010 92 www.slm-neurology.com


L-dopa-induced dyskinesias in Parkinson’s disease

CEREBRAL CORTEX

STRIATUM D2
D1 Thalamus

Indirect ptw

Direct ptw
GPe STN
Nigro striatal
dopaminergic GPi
pathway SN
Pars reticulata
SN
Pars compacta

CEREBRAL CORTEX

STRIATUM D2
D1 Thalamus

Indirect ptw
Direct ptw

GPe STN
Nigro-striatal
dopaminergic GPi
pathway SN
Pars reticulata

SN
Pars compacta

Figure 1. A) Direct and indirect pathways in normal physiological conditions. B) Rate model of PD: It was originally proposed that with SN pars compacta
neuronal loss, overexcitability of the direct pathway compared with the indirect would lead to hyperactivity of GPi and STN, and increased inhibitory basal
ganglia outflow toward the thalamus. The reality of basal ganglia dysfunction in PD is far more complex. SN pars compacta, substantia nigra pars compacta;
Gpe, globus pallidus pars externa; STN, subthalamic nucleus; Gpi, globus pallidus pars interna; SN pars reticulata, substantia nigra pars reticulata; Ptw, pathway.

interneurons.24,26–28 The chaotic unregulated release of dopa- and induces receptor internalization, and it is hypothesized
mine as a false neurotransmitter from these non-dopaminergic that in LID, this desensitization and internalization process is
terminals leads to further non-physiological dopamine recep- impaired.33
tor stimulation contributing to LID appearance.29 Upon persistent stimulation, D1Rs undergo desensitization
through a two-step process: activation-dependent receptor
phophorylation by G-protein-coupled receptor kinases (GRKs)
DOPAMINE RECEPTOR SUPERSENSITIVITY followed by the binding of uncoupling proteins called arrestins
Chronic denervation of dopamine receptors also leads to that induce internalization. The D1 receptor–arrestin interac-
receptor supersensitivity and increased expression of receptors tion promotes the recruitment of an endocytic complex and
on the postsynaptic membrane of medium spiny neurons.30,31 consequently the internalization of the receptor in an endocytic
A direct relationship between D1 receptor (D1R) supersensi- vesicle (Figure 2). This is modified in LID, and D1R expression
tivity and LID severity has been identified.32 Persistent stim- is increased and their subcellular distribution is modified. This
ulation of dopamine receptors leads to their desensitization has been proven in dyskinetic animals, as there are more D1R at

www.slm-neurology.com 93 ENJ 2010; 2:(2). August 2010


European Neurological Journal

Striatal Medium Spiny


Neuron CEREBRAL CORTEX

D1R
Arrestin
D1R
P P Arrestin

GRK

cAMP
D1R
DA AC
PKA PP

DARPP-32
BG output
L-dopa

Figure 2. Upper part of figure: desensitization process of D1R. D1R is phosphorylated by GRKs and once phosphorylated binds to arrestin. This induces
the formation of an endocytic complex. GRK, G protein receptor kinase. Bottom part of figure: Loss of depotentiation after chronic non-physiological D1
receptor stimulation. L-dopa, levodopa; DA, dopamine; D1R, D1 receptor; AC, adenylyl cyclase; PKA, protein kinase A; DARPP-32, dopamine- and cAMP-regulated
phosphoprotein of molecular weight 32 KDa: PP, protein phosphatase; LTD, long-term depotentiation; LTP, long-term potentiation; BG, basal ganglia.

the plasma membrane.34,35 However, it is clear that LID do not of processes culminates in the phosphorylation of intracellular
occur simply as a result of abnormal D1 receptor expression or proteins and glutamatergic receptors40 (Figure 2).
sensitivity alone because D2-selective agonists can also provoke LTP and LTD are fully abolished after the genetic disruption of
dyskinesia.36 DARPP-32.41 It is hypothesized that the disrupted motor control
underlying dyskinesia is attributable to specific changes occur-
ring along the D1/PKA/DARPP-32 signaling pathway leading
SYNAPTIC PLASTICITY to the inhibition of PP-1 activity and loss of synaptic depotentia-
tion at corticostriatal synapses.37 Disruption of normal synaptic
Two main output pathways arise from the striatal medium plasticity is strongly linked to the appearance of dyskinesia.42–46
spiny neurons: the direct pathway that projects from the stria- The loss of synaptic depotentiation at corticostriatal synapses
tum to globus pallidus pars interna (GPi) and substantia nigra leads to synaptic saturation and to the development of non-
pars reticulata (SNr), and the indirect one that projects to physiological motor circuits within the basal ganglia.37
GPi/SNr through the globus pallidus pars externa (GPe) and
subthalamic nucleus (STN) (Figure 1). Physiological dopamin- L-dopa can restore normal synaptic plasticity among individ-
ergic input from nigrostriatal neurons onto the striatal medium uals free of dyskinesia, but not when dyskinesias are already
spiny neurons plays an important role in the potentiation developed.47 It has been proposed that patients with LID have
and de-potentiation of synapses of the corticostriatal pathway. lost the ability to depotentiate synapses normally, that is, they
Repetitive stimulation can cause either a long-lasting increase have lost the mechanism that underlies “synaptic forgetting,”
in synaptic strength, known as long-term potentiation (LTP), or leading to the pathological storage of information that would
an enduring decrease, known as long-term depression (LTD), normally be erased and the development of abnormal motor
a phenomenon known as synaptic plasticity.37,38 This process patterns, that is, LID.43,48
allows the strengthening of desirable motor programs and the
deletion of unwanted or unnecessary connections.
The striatal neurons of the direct pathway express D1R and GENETIC INFLUENCES ON LID DEVELOPMENT
produce dynorphin and substance P, and the medium spiny Although much of the variation in LID risk can be attributed
neurons of the indirect pathway are richer in D2R and express to L-dopa dose and pattern of administration, based on the
enkephalin. The stimulation of D1R and D2R after L-dopa ther- preceding data, it is likely that genetic polymorphisms that
apy induces the activation or inhibition of adenylyl cyclase influence dopaminergic transmission or changes in synaptic
(AC), respectively, inducing opposite effects on the intracellu- plasticity may also contribute to susceptibility in developing
lar levels of cAMP.39 The increase in cAMP activates protein dyskinesia.49–51 Genetic variations of the dopamine receptor
kinase A (PKA) which in turn phosphorylates DA and cAMP- genes, DRD2, DRD3, and DRD4, have been studied but do not
regulated phospoprotein 32 kDa (DARPP-32) which acts as a seem to influence the occurrence of L-dopa -induced adverse
potent inhibitor of protein-phosphatase-1(PP-1). This cascade effects. However, there is limited evidence in support of a

ENJ 2010; 2:(2). August 2010 94 www.slm-neurology.com


L-dopa-induced dyskinesias in Parkinson’s disease

specific allele of the dopamine transporter gene (VNTR-DAT) agonists are generally to be avoided in view of long-term risks
being a predictor of LID in L-dopa-treated patients.52 In line of pulmonary and valvular fibrosis,61,62 notwithstanding the fact
with the evidence that LID develop because of abnormal synap- that no long-term (14-year) difference in dyskinesia induction
tic plasticity (which is in turn influenced by the release of has been reported comparing Bromocriptine versus L-dopa as
brain-derived neurotrophic factor (BDNF)),53 a functional poly- initial treatment.63
morphism in the BDNF gene (val66met) has also been pro- CDS using a long-acting transdermally administered agonist,
posed to influence time of onset of LID in one cohort of PD Rotigotine, has shown reduced dyskinesia induction compared
patients.54 These preliminary observations need to be replicated with pulsatile administration of the same drug and far less
in further large cohorts of PD patients. dyskinesia than pulsatile administration of L-dopa alone in
drug-naïve MPTP-treated animal models.64 Whether there is
Clinical Implications any advantage of one dopamine agonist over another has yet
For some patients dyskinesias are an acceptable inconve- to be studied, but the additional development of slow-release
nience, whereas for others dyskinesias can become severe, preparations of Ropinirole and Pramipexole, in addition to
exhausting, and disabling, interfering with the daily activi- Rotigotine, provides the opportunity to explore whether these
ties and producing gait disturbances or postural instability.55,56 drug formulations might confer long-term benefits on dyski-
Delaying the onset or reducing the severity of LID, while main- nesia risk.
taining effective relief of PD symptoms, represents a vital com-
The monoamine-oxidase B inhibitors, Selegiline and Rasa-
ponent of contemporary PD treatment.
giline, have been the subject of extensive research specifically
studying whether they possess any disease-modifying prop-
REDUCING THE RISK OF DYSKINESIA erties. They irreversibly inhibit MAO-B, the main dopamine-
metabolizing enzyme in the brain, and enhance dopamine
DEVELOPMENT
bioavailability, thus potentially leading to CDS. Both have been
CDS shown to improve PD symptoms65,66 and recently a small but
In the healthy state, dopamine release occurs in both a tonic sustained advantage has been reported as a result of the ear-
and a phasic manner57 ; however, in PD patients receiving short- lier initiation of treatment,67–69 but any advantage of long-term
acting drugs, the stimulation of dopamine receptors becomes LID risk is currently unproven. In the long-term follow-up of
discontinuous and pulsatile. Pulsatile administration of L-dopa the TEMPO trial the time for 25% of subjects to develop dysk-
has been shown to increase the development of LID in MPTP inesia showed no difference between the early-start group and
primates12 and in human studies13 and have led to the devel- the delayed-start group.70
opment of therapies to maximize “Continuous Dopaminergic Another theoretical way of improving CDS is through
Stimulation.” “Dopaminergic agonists” have been successfully the use of catechol-O-methyl transferase (COMT) inhibitors.
used for the control of motor symptoms of PD since their avail- This enzyme is the main mechanism of peripheral L-dopa
ability in the 1970s.58 These drugs were developed with the metabolism.71 COMT inhibition slows L-dopa plasma degrada-
expectation that on the basis of a longer plasma half-life lead- tion and increases its bioavailability without increasing max-
ing to a more physiological pattern of CDS, and as a substitutive imal concentration, allowing a L-dopa dose reduction by up
or concomitant treatment with L-dopa, PD symptoms could be to 30%.72 Entacapone and Tolcapone are selective COMT
relieved while reducing the risk of long-term motor fluctua- inhibitors that were developed for clinical use for the treat-
tions and dyskinesias.59 There is no doubt that the dopamine ment of PD. Tolcapone should be reserved for patients who
agonists have lower efficacy than L-dopa and more trouble- have inadequate benefit from Entacapone in view of rare but
some short-term side effects, and although these drugs can potentially fatal hepatotoxicity.73 Animal models showed that
themselves lead to the development of involuntary dyskinetic early coadministration of Entacapone with L-dopa allows good
movements, this is generally to a lesser extent than with L-dopa. motor control while decreasing the severity of all dyskine-
There is still debate about the magnitude of any advantage sia subtypes and delaying their onset.74,75 However, this sup-
of using dopamine agonists rather than L-dopa as initial ther- posed preventive effect has not been replicated in human clin-
apy in PD. The ELLDOPA study clearly demonstrated that high ical trials.76 The unpublished STRIDE-PD trial unfortunately
doses of L-dopa lead to dyskinesia development early on in showed that the time to dyskinesia was shorter in Carbidopa/
treatment. The recent 10-year follow-up of PD patients initi- L-dopa /Entacapone-treated patients, possibly because of
ated with Ropinirole versus L-dopa36 showed a significantly excessive L-dopa dosage among patients receiving COMT
lower incidence of dyskinesia in the Ropinirole group and a inhibitors.
significantly longer median time to dyskinesia, but no signif- Any strategy that reduces the pulsatility of dopaminergic stim-
icant difference in “disabling” dyskinesias. Furthermore by 10 ulation is likely to be an effective way of delaying time to onset
years, 93% of patients initiated on Ropinirole were also tak- of LID. The effectiveness and tolerability of any drug will vary
ing L-dopa, almost identical to the percentage in the group from one patient to the next, emphasizing the need to tai-
initiated on L-dopa, and having started on Ropinirole did not lor the drug regime to the individual. Combination therapies
translate to any significant advantage in activities of daily living. that afford symptom relief while keeping the dose of L-dopa
Similar results have also been shown with Pramipexole60 versus relatively low represent the optimal way of reducing pulsatile
L-dopa as the initial treatment choice. Ergot-derived dopamine dopaminergic receptor stimulation.

www.slm-neurology.com 95 ENJ 2010; 2:(2). August 2010


European Neurological Journal

TREATING LID ONCE THEY HAVE DEVELOPED these drugs prior to L-dopa prescription. Adding a dopamine
Following the onset of LID, the general principles of CDS agonist to a patient on L-dopa therapy generally increases
already outlined should be continued where possible to prevent peak dose dyskinesia unless the L-dopa dose is reduced.88
increases in LID frequency or severity. Beyond this, in order to However, this strategy has been successfully demonstrated with
implement effective treatment strategies for the treatment of both Ropinirole89 and Pramipexole.90,91 The addition of the
existing LID, it is important to establish which type of dyski- Rotigotine patch has been shown to increase “on” time with-
nesia the patient is describing, through a careful history of the out exacerbating dyskinesia in advanced PD patients although
type, distribution, and timing of the involuntary movements. reduction in pre-existing dyskinesia has not been reported.92
Patients with painful off-period dystonia in the early morning
can be helped by supplementary prescriptions of long-acting
Modification of Oral L-dopa dopamine agonists taken the preceding night. Patients with
Administration/Metabolism diphasic dyskinesias need to maintain sufficient dopaminer-
A short-term approach to reduce peak dose dyskinesias gic medication levels (L-dopa or agonist) throughout the day to
when they appear for the first time is to diminish the dose of reduce the regularity of their occurrence. However, care needs
dopaminergic agents to coincide with the time of day of LID to be taken when using this strategy to ensure that symptoms
occurrence. However, this strategy may be counter-productive of dopa-dysregulation do not supervene.
because of increases in “off” time and dose failures.77
Increasing the frequency of smaller doses of L-dopa can help
in the early stages but with continuing disease progression,
Adding Amantadine
recurrence of fluctuations between the off state and the on This drug was used originally as an antiviral agent, and it
state with accompanying dyskinesia demands further therapy was only through serendipity that it was observed to moder-
changes. Long-acting L-dopa preparations were developed to ately alleviate PD symptoms.93 Its mechanism of action is still
try and ameliorate fluctuations,78 but these preparations can not clear, but it is thought to be mediated through the N-
lead to increased numbers of “dose failures” and unpredictable methyl-D-aspartate receptor antagonist effect seen with high
L-dopa accumulation that further compounds the problem.79 doses (300–400 mg/day). Amantadine still remains the only
Patients describing off-period dystonia, for example, painful drug proven to have direct anti-dyskinetic effects, with improve-
posturing of a foot in the early morning prior to their medica- ments demonstrated of up to 50%.94–96 Efficacy has been shown
tion, can be helped with the prescription of dispersible forms of for up to 1 year97 ; however, loss of efficacy after 3–8 months
L-dopa, but these should not be used throughout the day as they has also been reported in a double-blind evaluation.98 Although
are likely to increase the pulsatility of dopaminergic receptor efficacy is most clearly demonstrated at higher dose levels, it
stimulation. is wise to try and keep the dose to the lowest useful level par-
ticularly among elderly patients or individuals with evidence of
MAO-B inhibitors have only mild effects on motor symptoms
cognitive impairment because of cognitive side effects.
in advanced PD but can allow a decrease in required L-dopa
dosage without deterioration in “off-time.” Entacapone has
also demonstrated its efficacy in prolonging the L-dopa clinical Switching to Apomorphine
response in patients with motor fluctuations.80,81 Nevertheless Apomorphine is the most potent dopamine agonist used
dyskinesia increase can occur because of the accumulating lev- in clinical practice and has a very fast onset (5–15 minutes)
els of plasma L-dopa necessitating careful titrated reduction but short duration of effect (∼40 minutes).99 It can be used,
of L-dopa dose.82,83 The LARGO trial revealed a significant through intermittent subcutaneous administration, as a suc-
but slight increase in daily-on time without troublesome dysk- cessful rescue treatment of severe unpredictable “off” periods
inesia in both Rasagiline and Entacapone groups compared in advanced PD patients with motor fluctuations.100 It has also
with placebo in adjunct treatment with L-dopa. No differences been used as a continuous infusion through a pump since
between Rasagiline and Entacapone were reported.84 1995,101 and there is robust data to demonstrate good control of
PD symptoms together with the reduction of peak-dose dysk-
Adding or Increasing an Oral Agonist inesia, by using this therapy as a means of lowering L-dopa
requirements.102–105 Cognitive side effects, nausea, hypersom-
Whether shifting therapy toward the dopamine agonists can
nolence, and skin nodules may occur as well as a rare hemolytic
reverse dyskinesia severity without worsening “off time” is
anemia and these factors may limit the number of patients that
yet to be convincingly established. Among the MPTP-treated
tolerate its use over the long term.
marmosets with established dyskinesias provoked by pulsatile
Rotigotine or L-dopa, continuous Rotigotine administration
led to reversal in dyskinesia.85 In the same animal models, Ablative Neurosurgery
switching to Ropinirole86 or adding Pramipexole87 to L-dopa Creating lesions in the thalamus and globus pallidum for
(with its dosage disminution) has also significantly reduced the treatment of PD was pioneered in the 1950s but then
pre-existing LID while maintaining therapeutic benefit. displaced for the treatment of PD with the introduction of L-
From the practical perspective, most patients suitable for dopa. A major renaissance of surgery followed the publication
dopamine agonist treatment have already been prescribed that posteroventral pallidotomy could improve not only the

ENJ 2010; 2:(2). August 2010 96 www.slm-neurology.com


L-dopa-induced dyskinesias in Parkinson’s disease

main PD symptoms, but LID as well,106,107 and this has been however been shown to improve dyskinesia control in up to
demonstrated as part of randomized trials.108,109 Bilateral pal- 50% without worsening of motor symptoms.131–133 The dis-
lidotomy causes swallowing difficulties,110 and with the advent advantage of this drug is the need for regular blood testing
of implantable pulse generators to enable delivery of chronic because of the risk of agranulocytosis.
stimulation, lesioning techniques are now rarely performed.
FUTURE THERAPIES
Deep Brain Stimulation
Further trials are underway to evaluate novel approaches to
Deep brain stimulation has become a widely used, safe, and
reduce LID in PD. These include putative antidyskinetic drugs
effective technique for the treatment of the cardinal motor
such as D2/D3 receptor “partial” agonists, adenosine A2a
symptoms of advanced PD.111,112 When dyskinesia is the main
antagonists, and metabotropic glutamate receptor antagonists.
problem, the choice of targets lies between the STN and the
In addition, there are four major gene therapy programs being
GPi.113 There is general acceptance that dyskinesia reduction
conducted to evaluate whether the complications of PD can
is greatest following GPi-deep brain stimulation (DBS), with
be ameliorated through growth factor administration,134 GAD
improvement rates of up to 89% following GPi-DBS compared
enzyme delivery to the STN,135 or attempts to improve upon stri-
with 62% in the STN.114,115 The improvement in “off” symptoms
atal CDS.136,137 Furthermore there are renewed attempts to apply
however tends to be less with GPi-DBS, and therefore medica-
cell therapies to patients with PD before LID development in an
tion requirements usually remain high postoperatively.114,115 It
attempt to allow restoration of normal physiological dopamin-
seems therefore that dyskinesia improvement following GPi-
ergic stimulation and avoid the long-term complications of
DBS is directly related to the stimulation effect by interfering
pharmacological dopaminergic replacement.
with pallidal outflow.
In contrast, STN-DBS frequently leads to reduction in the dose CONCLUSIONS
of L-dopa required and this is certainly one explanation why
dyskinesia severity has been shown to decrease.116,117 Over a 5- After more than 40 years of use and despite the develop-
year follow-up period, reduction in both peak-dose dyskinesia, ment of a wide range of newer treatments, L-dopa remains
biphasic dyskinesia, and off dystonia has been reported with the most efficacious agent to treat the motor symptoms of PD.
STN-DBS,118 and it has been shown that the response of the Nevertheless the disabling motor complications, particularly
patient to a L-dopa challenge also changes implying that there dyskinesias, can be disabling and limit its long-term thera-
is possibly also a direct effect of chronic STN stimulation.119 peutic use. The issue regarding whether and when to initiate
This may perhaps be mediated by changes in synaptic plas- treatment with L-dopa has been the topic of frequent discussion
ticity downstream from the STN,120 although this may equally to avoid long-term motor complications but without depriv-
be related to the stimulation of adjacent local pallidothalamic, ing patients the most effective treatment for the relief of their
pallidosubthalamic, and subthalamopallidal fiber projections motor symptoms. PD is a heterogeneous disease and individual
rather than STN neurons per se.117,121 patients will respond and tolerate prescribed drugs in differ-
ent ways with variable risks of dyskinesia development. L-dopa
Intra-Jejunal L-dopa should not be feared but when required should be used where
The best evidence that CDS can relieve LID comes from the possible in combination with the range of other available thera-
use of the enteral L-dopa infusion. A L-dopa -Carbidopa gel is pies unless or until alternative (more physiological) treatments
administered directly into the duodenum or the upper jejunum, become available.
bypassing the stomach (and thus its irregular pattern of empty- Acknowledgments: Dr Foltynie is supported by grants from the
ing and drug absorption), with a delivery controlled by an exter- Parkinson’s Appeal and Parkinson’s UK.
nal portable pump. The system has demonstrated a reduction in
Disclosure: Dr Foltynie has received honoraria for speaking at meet-
plasma L-dopa fluctuations compared with oral treatment.122–125 ings from Teva, UCB, GSK, and Orion Pharma.
This therapy is limited by the need for the patient to have a
permanent gastrostomy tube with jejunal extension, the con-
sequent risk of local complications,126 and its considerable REFERENCES
financial cost. At the current time, it is considered as a good 1. Cotzias GC, Van Woert MH, Schiffer LM. Aromatic amino acids and
alternative for those patients unsuitable to DBS or intolerant of modification of parkinsonism. N Engl J Med. 1967;276(7):374–379.
Apomorphine. No randomized controlled trials have been per- 2. Hornykiewicz OD. Physiologic, biochemical, and pathological back-
formed to directly compare the efficacy and tolerability of these grounds of levodopa and possibilities for the future. Neurology.
1970;20(12):1–5.
three therapies, and it is likely that any advantage in the mean 3. Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and
outcome of one therapy against another127–130 needs to be con- motor fluctuations as estimated from the cumulative literature. Mov Disord.
sidered very carefully in the context of tailoring treatment to the 2001;16(3):448–458.
individual patient. 4. Schrag A, Quinn N. Dyskinesias and motor fluctuations in Parkinson’s
disease. A community-based study. Brain. 2000;123(Pt. 11):2297–2305.
Clozapine 5. Kurlan R. “Levodopa phobia”: a new iatrogenic cause of disability in
Parkinson disease. Neurology. 2005;64(5):923–924.
Traditional antipsychotic drugs should not be used in the 6. Chapuis S, Ouchchane L, Metz O, Gerbaud L, Durif F. Impact of the motor
treatment of PD patients because of the inevitable deteriora- complications of Parkinson’s disease on the quality of life. Mov Disord.
tion in parkinsonism. The atypical antipsychotic clozapine has 2005;20(2):224–230.

www.slm-neurology.com 97 ENJ 2010; 2:(2). August 2010


European Neurological Journal

7. Kostic V, Przedborski S, Flaster E, Sternic N. Early development of 29. Carta M, Carlsson T, Munoz A, Kirik D, Bjorklund A. Serotonin-dopamine
levodopa-induced dyskinesias and response fluctuations in young-onset interaction in the induction and maintenance of L-DOPA-induced dyskine-
Parkinson’s disease. Neurology. 1991;41(2(Pt. 1)):202–205. sias. Prog Brain Res. 2008;172:465–478.
8. Wagner ML, Fedak MN, Sage JI, Mark MH. Complications of disease and 30. Lee T, Seeman P, Rajput A, Farley IJ, Hornykiewicz O. Receptor
therapy: a comparison of younger and older patients with Parkinson’s basis for dopaminergic supersensitivity in Parkinson’s disease. Nature.
disease. Ann Clin Lab Sci. 1996;26(5):389–395. 1978;273(5657):59–61.
9. Lucking CB, Durr A, Bonifati V, et al. Association between early-onset 31. Shinotoh H, Hirayama K, Tateno Y. Dopamine D1 and D2 receptors in
Parkinson’s disease and mutations in the parkin gene. N Engl J Med. Parkinson’s disease and striatonigral degeneration determined by PET.
2000;342(21):1560–1567. Adv Neurol. 1993;60:488–493.
10. Khan NL, Graham E, Critchley P, et al. Parkin disease: a phenotypic study 32. Aubert I, Guigoni C, Hakansson K, et al. Increased D1 dopamine recep-
of a large case series. Brain. 2003;126(Pt. 6):1279–1292. tor signaling in levodopa-induced dyskinesia. Ann Neurol. 2005;57(1):
11. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of 17–26.
Parkinson’s disease. N Engl J Med. 2004;351(24):2498–2508. 33. Guigoni C, Bezard E. Involvement of canonical and non-canonical D1
12. Smith LA, Jackson MJ, Hansard MJ, Maratos E, Jenner P. Effect of pulsatile dopamine receptor signalling pathways in L-dopa-induced dyskinesia.
administration of levodopa on dyskinesia induction in drug-naive MPTP- Parkinsonism Relat Disord. 2009;15(Suppl. 3):S64–S67.
treated common marmosets: effect of dose, frequency of administration, 34. Berthet A, Porras G, Doudnikoff E, et al. Pharmacological analysis
and brain exposure. Mov Disord. 2003;18(5):487–495. demonstrates dramatic alteration of D1 dopamine receptor neuronal dis-
13. Nutt JG, Obeso JA, Stocchi F. Continuous dopamine-receptor stimula- tribution in the rat analog of L-DOPA-induced dyskinesia. J Neurosci.
tion in advanced Parkinson’s disease. Trends Neurosci. 2000;23(10 Suppl.): 2009;29(15):4829–4835.
S109–S115. 35. Guigoni C, Doudnikoff E, Li Q, Bloch B, Bezard E. Altered D(1) dopamine
14. Bezard E, Brotchie JM, Gross CE. Pathophysiology of levodopa-induced receptor trafficking in parkinsonian and dyskinetic non-human primates.
dyskinesia: potential for new therapies. Nat Rev Neurosci. 2001;2(8): Neurobiol Dis. 2007;26(2):452–463.
577–588. 36. Hauser RA, Rascol O, Korczyn AD, et al. Ten-year follow-up of Parkinson’s
15. Rodriguez M, Barroso-Chinea P, Abdala P, Obeso J, Gonzalez-Hernandez disease patients randomized to initial therapy with ropinirole or levodopa.
T. Dopamine cell degeneration induced by intraventricular administration Mov Disord. 2007;22(16):2409–2417.
of 6-hydroxydopamine in the rat: similarities with cell loss in parkinson’s 37. Calabresi P, Di Filippo M, Ghiglieri V, Picconi B. Molecular
disease. Exp Neurol. 2001;169(1):163–181. mechanisms underlying levodopa-induced dyskinesia. Mov Disord.
16. Marin C, Aguilar E, Mengod G, Cortes R, Obeso JA. Effects of early vs. late 2008;23(Suppl. 3):S570–S579.
initiation of levodopa treatment in hemiparkinsonian rats. Eur J Neurosci. 38. Di Filippo M, Picconi B, Tantucci M, et al. Short-term and long-term plas-
2009;30(5):823–832. ticity at corticostriatal synapses: implications for learning and memory.
17. Bezard E, Gross CE. Compensatory mechanisms in experimental and Behav Brain Res. 2009;199(1):108–118.
human parkinsonism: towards a dynamic approach. Prog Neurobiol. 39. Greengard P, Allen PB, Nairn AC. Beyond the dopamine recep-
1998;55(2):93–116. tor: the DARPP-32/protein phosphatase-1 cascade. Neuron. 1999;23(3):
18. Langston JW, Ballard P. Parkinsonism induced by 1-methyl-4- 435–447.
phenyl-1,2,3,6-tetrahydropyridine (MPTP): implications for treatment 40. Santini E, Valjent E, Usiello A, et al. Critical involvement of cAMP/DARPP-
and the pathogenesis of Parkinson’s disease. Can J Neurol Sci. 1984;11 32 and extracellular signal-regulated protein kinase signaling in L-DOPA-
(1 Suppl.):160–165. induced dyskinesia. J Neurosci. 2007;27(26):6995–7005.
19. Kuoppamaki M, Al-Barghouthy G, Jackson MJ, Smith LA, Quinn N, 41. Calabresi P, Gubellini P, Centonze D, et al. Dopamine and cAMP-regulated
Jenner P. L-dopa dose and the duration and severity of dyskine- phosphoprotein 32 kDa controls both striatal long-term depression and
sia in primed MPTP-treated primates. J Neural Transm. 2007;114(9): long-term potentiation, opposing forms of synaptic plasticity. J Neurosci.
1147–1153. 2000;20(22):8443–8451.
20. Wichmann T, DeLong MR. Pathophysiology of Parkinson’s disease: 42. Linazasoro G. New ideas on the origin of L-dopa-induced dyskinesias: age,
the MPTP primate model of the human disorder. Ann N Y Acad Sci. genes and neural plasticity. Trends Pharmacol Sci. 2005;26(8):391–397.
2003;991:199–213. 43. Picconi B, Centonze D, Hakansson K, et al. Loss of bidirectional
21. Mitchell IJ, Boyce S, Sambrook MA, Crossman AR. A 2-deoxyglucose striatal synaptic plasticity in L-DOPA-induced dyskinesia. Nat Neurosci.
study of the effects of dopamine agonists on the parkinsonian 2003;6(5):501–506.
primate brain. Implications for the neural mechanisms that medi- 44. Calabresi P, Galletti F, Saggese E, Ghiglieri V, Picconi B. Neuronal
ate dopamine agonist-induced dyskinesia. Brain. 1992;115(Pt. 3): networks and synaptic plasticity in Parkinson’s disease: beyond motor
809–824. deficits. Parkinsonism Relat Disord. 2007;13(Suppl. 3):S259–S262.
22. Laitinen LV, Bergenheim AT, Hariz MI. Ventroposterolateral pallido- 45. Chase TN. Striatal plasticity and extrapyramidal motor dysfunction.
tomy can abolish all parkinsonian symptoms. Stereotact Funct Neurosurg. Parkinsonism Relat Disord. 2004;10(5):305–313.
1992;58(1–4):14–21. 46. Ueki Y, Mima T, Kotb MA, et al. Altered plasticity of the human motor
23. Conn PJ, Battaglia G, Marino MJ, Nicoletti F. Metabotropic gluta- cortex in Parkinson’s disease. Ann Neurol. 2006;59(1):60–71.
mate receptors in the basal ganglia motor circuit. Nat Rev Neurosci. 47. Morgante F, Espay AJ, Gunraj C, Lang AE, Chen R. Motor cortex plas-
2005;6(10):787–798. ticity in Parkinson’s disease and levodopa-induced dyskinesias. Brain.
24. Lee J, Zhu WM, Stanic D, et al. Sprouting of dopamine terminals and 2006;129(Pt. 4):1059–1069.
altered dopamine release and uptake in Parkinsonian dyskinaesia. Brain. 48. Picconi B, Paille V, Ghiglieri V, et al. L-DOPA dosage is critically
2008;131(Pt. 6):1574–1587. involved in dyskinesia via loss of synaptic depotentiation. Neurobiol Dis.
25. Sossi V, de la Fuente-Fernandez R, Schulzer M, Adams J, Stoessl J. Age- 2008;29(2):327–335.
related differences in levodopa dynamics in Parkinson’s: implications for 49. Lin JJ, Yueh KC, Lin SZ, Harn HJ, Liu JT. Genetic polymorphism of the
motor complications. Brain. 2006;129(Pt. 4):1050–1058. angiotensin converting enzyme and L-dopa-induced adverse effects in
26. Melamed E, Hefti F, Pettibone DJ, Liebman J, Wurtman RJ. Aromatic Parkinson’s disease. J Neurol Sci. 2007;252(2):130–134.
L-amino acid decarboxylase in rat corpus striatum: implications for action 50. Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-
of L-dopa in parkinsonism. Neurology. 1981;31(6):651–655. year study of the incidence of dyskinesia in patients with early Parkinson’s
27. Ng KY, Chase TN, Colburn RW, Kopin IJ. L-dopa-induced release of disease who were treated with ropinirole or levodopa. 056 Study Group.
cerebral monoamines. Science. 1970;170(953):76–77. N Engl J Med. 2000;342(20):1484–1491.
28. Carta M, Carlsson T, Munoz A, Kirik D, Bjorklund A. Involvement of the 51. Zappia M, Annesi G, Nicoletti G, et al. Sex differences in clinical and
serotonin system in L-dopa-induced dyskinesias. Parkinsonism Relat Disord. genetic determinants of levodopa peak-dose dyskinesias in Parkinson
2008;14(Suppl. 2):S154–S158. disease: an exploratory study. Arch Neurol. 2005;62(4):601–605.

ENJ 2010; 2:(2). August 2010 98 www.slm-neurology.com


L-dopa-induced dyskinesias in Parkinson’s disease

52. Kaiser R, Hofer A, Grapengiesser A, et al. L-dopa-induced adverse Mov Disord. 2005;20(3):306–314.
effects in PD and dopamine transporter gene polymorphism. Neurology. 76. Hauser RA, Panisset M, Abbruzzese G, Mancione L, Dronamraju
2003;60(11):1750–1755. N, Kakarieka A. Double-blind trial of levodopa/carbidopa/entacapone
53. Wardle RA, Poo MM. Brain-derived neurotrophic factor modulation of versus levodopa/carbidopa in early Parkinson’s disease. Mov Disord.
GABAergic synapses by postsynaptic regulation of chloride transport. 2009;24(4):541–550.
J Neurosci. 2003;23(25):8722–8732. 77. Metman LV, van den Munckhof P, Klaassen AA, Blanchet P, Mouradian
54. Foltynie T, Cheeran B, Williams-Gray CH, et al. BDNF val66met influ- MM, Chase TN. Effects of supra-threshold levodopa doses on dyskinesias
ences time to onset of levodopa induced dyskinesia in Parkinson’s disease. in advanced Parkinson’s disease. Neurology. 1997;49(3):711–713.
J Neurol Neurosurg Psychiatry. 2009;80(2):141–144. 78. Pacchetti C, Martignoni E, Sibilla L, Bruggi P, Turla M, Nappi G.
55. Marras C, Lang A, Krahn M, Tomlinson G, Naglie G. Quality of life in early Effectiveness of Madopar HBS plus Madopar standard in patients with
Parkinson’s disease: impact of dyskinesias and motor fluctuations. Mov fluctuating Parkinson’s disease: two years of follow-up. Eur Neurol.
Disord. 2004;19(1):22–28. 1990;30(6):319–323.
56. Sethi KD. The impact of levodopa on quality of life in patients with 79. Koller WC, Hutton JT, Tolosa E, Capilldeo R. Immediate-release
Parkinson disease. Neurologist. 2010;16(2):76–83. and controlled-release carbidopa/levodopa in PD: a 5-year random-
57. Grace AA, Bunney BS. The control of firing pattern in nigral dopamine ized multicenter study. Carbidopa/Levodopa Study Group. Neurology.
neurons: single spike firing. J Neurosci. 1984;4(11):2866–2876. 1999;53(5):1012–1019.
58. Calne DB, Teychenne PF, Claveria LE, Eastman R, Greenacre JK, Petrie 80. Rinne UK, Larsen JP, Siden A, Worm-Petersen J. Entacapone enhances the
A. Bromocriptine in Parkinsonism. Br Med J. 1974;4(5942):442–444. response to levodopa in parkinsonian patients with motor fluctuations.
59. Fahn S. How do you treat motor complications in Parkinson’s disease: Nomecomt Study Group. Neurology. 1998;51(5):1309–1314.
medicine, surgery, or both? Ann Neurol. 2008;64(Suppl. 2):S56–S64. 81. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s
60. Holloway RG, Shoulson I, Fahn S, et al. Pramipexole vs levodopa as initial disease patients. Parkinson Study Group. Ann Neurol. 1997;42(5):
treatment for Parkinson disease: a 4-year randomized controlled trial. Arch 747–755.
Neurol. 2004;61(7):1044–1053. 82. Linazasoro G, Kulisevsky J, Hernandez B. Should levodopa dose be
61. Tintner R, Manian P, Gauthier P, Jankovic J. Pleuropulmonary fibrosis after reduced when switched to stalevo? Eur J Neurol. 2008;15(3):257–261.
long-term treatment with the dopamine agonist pergolide for Parkinson 83. Gordin A, Kaakkola S, Teravainen H. Clinical advantages of COMT
Disease. Arch Neurol. 2005;62(8):1290–1295. inhibition with entacapone—a review. J Neural Transm. 2004;111(10–11):
62. Antonini A, Poewe W. Fibrotic heart-valve reactions to dopamine-agonist 1343–1363.
treatment in Parkinson’s disease. Lancet Neurol. 2007;6(9):826–829. 84. Rascol O, Brooks DJ, Melamed E, et al. Rasagiline as an adjunct to
63. Katzenschlager R, Head J, Schrag A, Ben-Shlomo Y, Evans A, Lees AJ. levodopa in patients with Parkinson’s disease and motor fluctuations
Fourteen-year final report of the randomized PDRG-UK trial comparing (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once
three initial treatments in PD. Neurology. 2008;71(7):474–480. daily, study): a randomised, double-blind, parallel-group trial. Lancet.
64. Stockwell KA, Scheller D, Rose S, et al. Continuous 2005;365(9463):947–954.
administration of rotigotine to MPTP-treated com- 85. Stockwell KA, Scheller DK, Smith LA, et al. Continuous rotigotine
mon marmosets enhances anti-parkinsonian activity and administration reduces dyskinesia resulting from pulsatile treatment with
reduces dyskinesia induction. Exp Neurol. 2009;219(2): rotigotine or L-DOPA in MPTP-treated common marmosets. Exp Neurol.
533–542. 2010;221(1):79–85.
65. Elizan TS, Yahr MD, Moros DA, Mendoza MR, Pang S, Bodian CA. 86. Jackson MJ, Smith LA, Al-Barghouthy G, Rose S, Jenner P. Decreased
Selegiline as an adjunct to conventional levodopa therapy in Parkinson’s expression of L-dopa-induced dyskinesia by switching to ropini-
disease. Experience with this type B monoamine oxidase inhibitor in 200 role in MPTP-treated common marmosets. Exp Neurol. 2007;204(1):
patients. Arch Neurol. 1989;46(12):1280–1283. 162–170.
66. Parkinson Study Group. A controlled, randomized, delayed-start study of 87. Tayarani-Binazir KA, Jackson MJ, Rose S, Olanow CW, Jenner P.
rasagiline in early Parkinson disease. Arch Neurol. 2004;61(4):561–566. Pramipexole combined with levodopa improves motor function but
67. Olanow CW, Hauser RA, Jankovic J, et al. A randomized, double-blind, reduces dyskinesia in MPTP-treated common marmosets. Mov Disord.
placebo-controlled, delayed start study to assess rasagiline as a dis- 2010;25(3):377–384.
ease modifying therapy in Parkinson’s disease (the ADAGIO study): 88. Talati R, Baker WL, Patel AA, Reinhart K, Coleman CI. Adding a
rationale, design, and baseline characteristics. Mov Disord. 2008;23(15): dopamine agonist to preexisting levodopa therapy vs. levodopa therapy
2194–2201. alone in advanced Parkinson’s disease: a meta analysis. Int J Clin Pract.
68. Palhagen S, Heinonen E, Hagglund J, Kaugesaar T, Maki-Ikola O, Palm R. 2009;63(4):613–623.
Selegiline slows the progression of the symptoms of Parkinson disease. 89. Cristina S, Zangaglia R, Mancini F, Martignoni E, Nappi G, Pacchetti C.
Neurology. 2006;66(8):1200–1206. High-dose ropinirole in advanced Parkinson’s disease with severe dyski-
69. Parkinson Study Group. A controlled trial of rasagiline in early Parkinson nesias. Clin Neuropharmacol. 2003;26(3):146–150.
disease: the TEMPO Study. Arch Neurol. 2002;59(12):1937–1943. 90. Guttman M. Double-blind comparison of pramipexole and bromocriptine
70. Hauser RA, Lew MF, Hurtig HI, Ondo WG, Wojcieszek J, Fitzer-Attas CJ. treatment with placebo in advanced Parkinson’s disease. International
Long-term outcome of early versus delayed rasagiline treatment in early Pramipexole-Bromocriptine Study Group. Neurology. 1997;49(4):
Parkinson’s disease. Mov Disord. 2009;24(4):564–573. 1060–1065.
71. Axelrod J, Tomchick R. Enzymatic O-methylation of epinephrine and other 91. Brodsky MA, Park BS, Nutt JG. Effects of a dopamine agonist on
catechols. J Biol Chem. 1958;233(3):702–705. the pharmacodynamics of levodopa in Parkinson disease. Arch Neurol.
72. Nutt JG, Woodward WR, Beckner RM, et al. Effect of peripheral catechol- 2010;67(1):27–32.
O-methyltransferase inhibition on the pharmacokinetics and pharmaco- 92. Rektor I, Babic T, Boothmann B, Polivka J, Boroojerdi B, Randerath O.
dynamics of levodopa in parkinsonian patients. Neurology. 1994;44(5): High doses of rotigotine transdermal patch: results of an open-label,
913–919. dose-escalation trial in patients with advanced-stage, idiopathic Parkinson
73. Assal F, Spahr L, Hadengue A, Rubbia-Brandt L, Burkhard PR. Tolcapone disease. Clin Neuropharmacol. 2009;32(4):193–198.
and fulminant hepatitis. Lancet. 1998;352(9132):958. 93. Schwab RS, England AC, Jr., Poskanzer DC, Young RR. Amantadine in the
74. Marin C, Aguilar E, Obeso JA. Coadministration of entacapone with lev- treatment of Parkinson’s disease. JAMA. 1969;208(7):1168–1170.
odopa attenuates the severity of dyskinesias in hemiparkinsonian rats. Mov 94. Luginger E, Wenning GK, Bosch S, Poewe W. Beneficial effects of aman-
Disord. 2006;21(5):646–653. tadine on L-dopa-induced dyskinesias in Parkinson’s disease. Mov Disord.
75. Smith LA, Jackson MJ, Al-Barghouthy G, et al. Multiple small doses of 2000;15(5):873–878.
levodopa plus entacapone produce continuous dopaminergic stimulation 95. Verhagen Metman L, Del Dotto P, van den Munckhof P, Fang J, Mouradian
and reduce dyskinesia induction in MPTP-treated drug-naive primates. MM, Chase TN. Amantadine as treatment for dyskinesias and motor

www.slm-neurology.com 99 ENJ 2010; 2:(2). August 2010


European Neurological Journal

fluctuations in Parkinson’s disease. Neurology. 1998;50(5):1323–1326. Direct effect of subthalamic nucleus stimulation on levodopa-induced
96. Rajput AW, Rajput AH. 18 month prospective study of amantadine for peak-dose dyskinesia in patients with Parkinson’s disease. Stereotact Funct
levodopa induced dyskinesia in Parkinson’s Disease. Can J Neurol Sci. Neurosurg. 2006;84(4):176–179.
1997;24:S23. 118. Simonin C, Tir M, Devos D, et al. Reduced levodopa-induced complica-
97. Metman LV, Del Dotto P, LePoole K, Konitsiotis S, Fang J, Chase TN. tions after 5 years of subthalamic stimulation in Parkinson’s disease: a
Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. second honeymoon. J Neurol. 2009;256(10):1736–1741.
Arch Neurol. 1999;56(11):1383–1386. 119. Piboolnurak P, Lang AE, Lozano AM, et al. Levodopa response in long-
98. Thomas A, Iacono D, Luciano AL, Armellino K, Di Iorio A, Onofrj M. term bilateral subthalamic stimulation for Parkinson’s disease. Mov Disord.
Duration of amantadine benefit on dyskinesia of severe Parkinson’s dis- 2007;22(7):990–997.
ease. J Neurol Neurosurg Psychiatry. 2004;75(1):141–143. 120. Moro E, Esselink RJ, Benabid AL, Pollak P. Response to levodopa in parkin-
99. Duby SE, Cotzias GC, Papavasiliou PS, Lawrence WH. Injected apomor- sonian patients with bilateral subthalamic nucleus stimulation. Brain.
phine and orally administered levodopa in Parkinsonism. Arch Neurol. 2002;125(Pt. 11):2408–2417.
1972;27(6):474–480. 121. Alterman RL, Shils JL, Gudesblatt M, Tagliati M. Immediate and sustained
100. Chen JJ, Obering C. A review of intermittent subcutaneous apomorphine relief of levodopa-induced dyskinesias after dorsal relocation of a deep
injections for the rescue management of motor fluctuations associated brain stimulation lead. Case report. Neurosurg Focus. 2004;17(1):E6.
with advanced Parkinson’s disease. Clin Ther. 2005;27(11):1710–1724. 122. Nyholm D, Askmark H, Gomes-Trolin C, et al. Optimizing levodopa phar-
101. Gancher ST, Nutt JG, Woodward WR. Apomorphine infusional therapy macokinetics: intestinal infusion versus oral sustained-release tablets. Clin
in Parkinson’s disease: clinical utility and lack of tolerance. Mov Disord. Neuropharmacol. 2003;26(3):156–163.
1995;10(1):37–43. 123. Stocchi F, Vacca L, Ruggieri S, Olanow CW. Intermittent vs continuous
102. Manson AJ, Turner K, Lees AJ. Apomorphine monotherapy in the treat- levodopa administration in patients with advanced Parkinson disease: a
ment of refractory motor complications of Parkinson’s disease: long-term clinical and pharmacokinetic study. Arch Neurol. 2005;62(6):905–910.
follow-up study of 64 patients. Mov Disord. 2002;17(6):1235–1241. 124. Antonini A, Isaias IU, Canesi M, et al. Duodenal levodopa infusion for
103. Stocchi F, Vacca L, De Pandis MF, Barbato L, Valente M, Ruggieri S. advanced Parkinson’s disease: 12-month treatment outcome. Mov Disord.
Subcutaneous continuous apomorphine infusion in fluctuating patients 2007;22(8):1145–1149.
with Parkinson’s disease: long-term results. Neurol Sci. 2001;22(1):93–94. 125. Nyholm D, Nilsson Remahl AI, Dizdar N, et al. Duodenal levodopa infu-
104. Colzi A, Turner K, Lees AJ. Continuous subcutaneous waking day apo- sion monotherapy vs oral polypharmacy in advanced Parkinson disease.
morphine in the long term treatment of levodopa induced interdose dysk- Neurology. 2005;64(2):216–223.
inesias in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1998;64(5): 126. Nyholm D, Lewander T, Johansson A, Lewitt PA, Lundqvist C, Aquilonius
573–576. SM. Enteral levodopa/carbidopa infusion in advanced Parkinson disease:
105. Katzenschlager R, Hughes A, Evans A, et al. Continuous subcuta- long-term exposure. Clin Neuropharmacol. 2008;31(2):63–73.
neous apomorphine therapy improves dyskinesias in Parkinson’s dis- 127. De Gaspari D, Siri C, Landi A, et al. Clinical and neuropsychological follow
ease: a prospective study using single-dose challenges. Mov Disord. up at 12 months in patients with complicated Parkinson’s disease treated
2005;20(2):151–157. with subcutaneous apomorphine infusion or deep brain stimulation of the
106. Laitinen LV, Bergenheim AT, Hariz MI. Leksell’s posteroventral pallido- subthalamic nucleus. J Neurol Neurosurg Psychiatry. 2006;77(4):450–453.
tomy in the treatment of Parkinson’s disease. J Neurosurg. 1992;76(1): 128. Clarke CE, Worth P, Grosset D, Stewart D. Systematic review of apomor-
53–61. phine infusion, levodopa infusion and deep brain stimulation in advanced
107. Guridi J, Obeso JA, Rodriguez-Oroz MC, Lozano AA, Manrique M. Parkinson’s disease. Parkinsonism Relat Disord. 2009;15(10):728–741.
L-dopa-induced dyskinesia and stereotactic surgery for Parkinson’s dis- 129. Nyholm D, Constantinescu R, Holmberg B, Dizdar N, Askmark H.
ease. Neurosurgery. 2008;62(2):311–323;discussion 323–325. Comparison of apomorphine and levodopa infusions in four patients
108. de Bie RM, de Haan RJ, Nijssen PC, et al. Unilateral pallidotomy in with Parkinson’s disease with symptom fluctuations. Acta Neurol Scand.
Parkinson’s disease: a randomised, single-blind, multicentre trial. Lancet. 2009;119(5):345–348.
1999;354(9191):1665–1669. 130. Deuschl G, Schade-Brittinger C, Krack P, et al. A randomized trial of
109. Vitek JL, Bakay RA, Freeman A, et al. Randomized trial of pallidotomy deep-brain stimulation for Parkinson’s disease. N Engl J Med. 2006;355(9):
versus medical therapy for Parkinson’s disease. Ann Neurol. 2003;53(5): 896–908.
558–569. 131. Bennett JP, Jr., Landow ER, Schuh LA. Suppression of dyskinesias
110. Merello M, Starkstein S, Nouzeilles MI, Kuzis G, Leiguarda R. Bilateral in advanced Parkinson’s disease. II. Increasing daily clozapine doses
pallidotomy for treatment of Parkinson’s disease induced corticobul- suppress dyskinesias and improve parkinsonism symptoms. Neurology.
bar syndrome and psychic akinesia avoidable by globus pallidus lesion 1993;43(8):1551–1555.
combined with contralateral stimulation. J Neurol Neurosurg Psychiatry. 132. Pierelli F, Adipietro A, Soldati G, Fattapposta F, Pozzessere G, Scoppetta
2001;71(5):611–614. C. Low dosage clozapine effects on L-dopa induced dyskinesias in parkin-
111. Limousin P, Pollak P, Benazzouz A, et al. Effect of parkinsonian signs sonian patients. Acta Neurol Scand. 1998;97(5):295–299.
and symptoms of bilateral subthalamic nucleus stimulation. Lancet. 133. Durif F, Debilly B, Galitzky M, et al. Clozapine improves dyskinesias in
1995;345(8942):91–95. Parkinson disease: a double-blind, placebo-controlled study. Neurology.
112. Rodriguez-Oroz MC, Obeso JA, Lang AE, et al. Bilateral deep brain stimu- 2004;62(3):381–388.
lation in Parkinson’s disease: a multicentre study with 4 years follow-up. 134. Marks WJ, Jr., Ostrem JL, Verhagen L, et al. Safety and tolerability of
Brain. 2005;128(Pt. 10):2240–2249. intraputaminal delivery of CERE-120 (adeno-associated virus serotype 2-
113. Deep-brain stimulation of the subthalamic nucleus or the pars interna of neurturin) to patients with idiopathic Parkinson’s disease: an open-label,
the globus pallidus in Parkinson’s disease. N Engl J Med. 2001;345(13): phase I trial. Lancet Neurol. 2008;7(5):400–408.
956–963. 135. Kaplitt MG, Feigin A, Tang C, et al. Safety and tolerability of gene ther-
114. Burchiel KJ, Anderson VC, Favre J, Hammerstad JP. Comparison of pallidal apy with an adeno-associated virus (AAV) borne GAD gene for Parkinson’s
and subthalamic nucleus deep brain stimulation for advanced Parkinson’s disease: an open label, phase I trial. Lancet. 2007;369(9579):2097–2105.
disease: results of a randomized, blinded pilot study. Neurosurgery. 136. Christine CW, Starr PA, Larson PS, et al. Safety and tolerability of putam-
1999;45(6):1375–1382;discussion 1382–1384. inal AADC gene therapy for Parkinson disease. Neurology. 2009;73(20):
115. Anderson VC, Burchiel KJ, Hogarth P, Favre J, Hammerstad JP. Pallidal 1662–1669.
vs subthalamic nucleus deep brain stimulation in Parkinson disease. Arch 137. Jarraya B, Boulet S, Ralph GS, et al. Dopamine gene therapy for
Neurol. 2005;62(4):554–560. Parkinson’s disease in a nonhuman primate without associated dyskine-
116. Moro E, Scerrati M, Romito LM, Roselli R, Tonali P, Albanese A. Chronic sia. Sci Transl Med. 2009;1(2):2ra4.
subthalamic nucleus stimulation reduces medication requirements in
Parkinson’s disease. Neurology. 1999;53(1):85–90.
117. Katayama Y, Oshima H, Kano T, Kobayashi K, Fukaya C, Yamamoto T.

ENJ 2010; 2:(2). August 2010 100 www.slm-neurology.com

Potrebbero piacerti anche