Sei sulla pagina 1di 226

Dx/Rx:

Brain Tumors

Eudocia Quant, MD
Center for Neuro-Oncology
Dana-Farber/Brigham and Women’s Cancer Center
Boston, MA
Series Editor: Kevin N. Sheth, MD
Division of Neuro-Critical Care & Stroke
University of Maryland Medical Center
R. Adams Cowley Shock Trauma Center
Baltimore, MD

73724_FM00_Printer.indd i 03/29/09 2:40:00 PM


World Headquarters Jones and Bartlett Publishers Canada
Jones and Bartlett Publishers 6339 Ormindale Way
40 Tall Pine Drive Mississauga, Ontario L5V 1J2
Sudbury, MA 01776 Canada
978-443-5000
info@jbpub.com Jones and Bartlett Publishers International
www.jbpub.com Barb House, Barb Mews
London W6 7PA
United Kingdom
Jones and Bartlett’s books and products are available through most bookstores and online
booksellers. To contact Jones and Bartlett Publishers directly, call 800-832-0034,
fax 978-443-8000, or visit our website, www.jbpub.com.

Substantial discounts on bulk quantities of Jones and Bartlett’s publications are available
to corporations, professional associations, and other qualified organizations. For details
and specific discount information, contact the special sales department at Jones and
Bartlett via the above contact information or send an email to specialsales@jbpub.com.

Copyright © 2011 by Jones and Bartlett Publishers, LLC

All rights reserved. No part of the material protected by this copyright may be reproduced or
utilized in any form, electronic or mechanical, including photocopying, recording, or by any
information storage and retrieval system, without written permission from the copyright owner.

The authors, editor, and publisher have made every effort to provide accurate information.
However, they are not responsible for errors, omissions, or for any outcomes related to the
use of the contents of this book and take no responsibility for the use of the products
and procedures described. Treatments and side effects described in this book may not be
applicable to all people; likewise, some people may require a dose or experience a side
effect that is not described herein. Drugs and medical devices are discussed that may have
limited availability controlled by the Food and Drug Administration (FDA) for use only in a
research study or clinical trial. Research, clinical practice, and government regulations often
change the accepted standard in this field. When consideration is being given to use of any
drug in the clinical setting, the healthcare provider or reader is responsible for determining
FDA status of the drug, reading the package insert, and reviewing prescribing information
for the most up-to-date recommendations on dose, precautions, and contraindications, and
determining the appropriate usage for the product. This is especially important in the case
of drugs that are new or seldom used.

Production Credits
Senior Acquisitions Editor: Nancy Anastasi Duffy Cover Design: Anne Spencer
Editorial Assistant: Sara Cameron Cover Image: © Basov Mikhail/
Production Director: Amy Rose ShutterStock, Inc.
Associate Production Editor: Jessica deMartin Composition: diacriTech, Chennai,
Senior Marketing Manager: Barb Bartoszek India
V.P., Manufacturing and Inventory Control: Printing and Binding: Malloy, Inc.
Therese Connell Cover Printing: Malloy, Inc.

Library of Congress Cataloging-in-Publication Data


Quant, Eudocia.
Dx/Rx. Brain tumors / Eudocia Quant.
p. ; cm. — (Dx/Rx neurology series)
Other title: Brain tumors
Includes bibliographical references and index.
ISBN-13: 978-0-7637-7372-4
ISBN-10: 0-7637-7372-7
1. Brain—Tumors—Handbooks, manuals, etc. I. Title. II. Title: Brain
tumors. III. Series: Jones and Bartlett Publishers Dx/Rx neurology series.
[DNLM: 1. Brain Neoplasms—diagnosis—Handbooks. 2. Brain Neoplasms—
therapy—Handbooks. WL 39 Q15d 2011]
RC280.B7.Q83 2011
616.99'481—dc22
2010000301
6048

Printed in the United States of America


14 13 12 11 10 10 9 8 7 6 5 4 3 2 1

73724_FM00_Printer.indd ii 03/29/09 2:40:00 PM


Contents

Editor’s Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . vii

Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix

1 Primary Brain Tumors . . . . . . . . . . . . . . . . . . . . . . . 1


Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Histological Classification of Primary Brain Tumors . . . . 1
Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Gliomas. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Meningioma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
Primary CNS Lymphoma (PCNSL) . . . . . . . . . . . . . . . 28
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35

2 Brain Metastases . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
Clinical Features in Parenchymal Metastases . . . . . . . . 43
Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
Prognosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 44
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
Brain Metastases from Breast Cancer . . . . . . . . . . . . . . 53
Brain Metastases from Non-Small Cell
Lung Cancer (NSCLC) . . . . . . . . . . . . . . . . . . 55
Brain Metastases from Melanoma . . . . . . . . . . . . . . . . 57
Brain Metastases from Renal Cell
Carcinoma (RCC) . . . . . . . . . . . . . . . . . . . . . . . 58
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59

3 Spinal Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
Extradural Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . 68
Intradural Tumors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80
iii

73724_FM00_Printer.indd iii 03/29/09 2:40:00 PM


iv Contents

4 Leptomeningeal Metastases . . . . . . . . . . . . . . . . 83
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
Prognosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90

5 Neurologic Complications
of Radiation Therapy . . . . . . . . . . . . . . . . . . . . . . . 93
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 93
Radiation Effects on the Central
Nervous System . . . . . . . . . . . . . . . . . . . . . . . . 94
Radiation Effects on the Peripheral
Nervous System . . . . . . . . . . . . . . . . . . . . . . . 101
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103

6 Neurologic Complications
of Chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . 107
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
Common Chemotherapy Agents . . . . . . . . . . . . . . . . . 107
Central Nervous System . . . . . . . . . . . . . . . . . . . . . . . 110
Peripheral Nervous System . . . . . . . . . . . . . . . . . . . . . 115
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123

7 Paraneoplastic Disorders . . . . . . . . . . . . . . . . . . 127


Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 127
Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 128
Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 130
Common Paraneoplastic Disorders . . . . . . . . . . . . . . . 131
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 152

8 Cerebrovascular Complications . . . . . . . . . . . . 161


Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161
Ischemic Strokes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 161

73724_FM00_Printer.indd iv 03/29/09 2:40:00 PM


Contents v

Embolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 163
Thrombotic Disease (Including Small, Medium,
and Large Vessel Disease) . . . . . . . . . . . . . . . . 166
Intracranial Hemorrhages . . . . . . . . . . . . . . . . . . . . . . 169
Cerebral Venous Thrombosis (CVT) . . . . . . . . . . . . . . 173
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176

9 Medical Complications of Brain


Tumor Patients . . . . . . . . . . . . . . . . . . . . . . . . . . . 181
Seizures. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 181
Venous Thromboembolism (VTE). . . . . . . . . . . . . . . . 186
Cerebral Edema . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 190
Pain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 192
Cognitive Impairment . . . . . . . . . . . . . . . . . . . . . . . . . 194
Fatigue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 195
Treatment Complications and Their Management . . . 196
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 202

Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207

73724_FM00_Printer.indd v 03/29/09 2:40:00 PM


73724_FM00_Printer.indd vi 03/29/09 2:40:00 PM
Editor’s Preface

Welcome to the Dx/Rx Neurology series. This is a new series


of books focusing on common neurological disorders. This
book, Dx/Rx: Brain Tumors, is a comprehensive overview of
tumors of the central nervous system and complications of
cancer and cancer therapy. Dr. Eudocia Quant provides a
succinct and easy-to-read overview for the neurological
trainee, neurologist, and practicing oncologist as well as the
general internist. The additional focus on complications of
nervous system tumors and their associated therapies is
most useful in addressing the significant morbidity patients
with brain tumors face. I believe you will find this, and the
entire neurology series, an invaluable resource to you and
your colleagues.

Kevin N. Sheth, MD

vii

73724_FM00_Printer.indd vii 03/29/09 2:40:00 PM


73724_FM00_Printer.indd viii 03/29/09 2:40:00 PM
Preface

This book focuses on two difficult issues in neuro-oncology:


brain tumors and neurologic complications of cancer. The
former represents a heterogeneous group that includes
benign and malignant histologies as well as primary and
metastatic lesions.
The latter is often a source of significant morbidity for
cancer patients. While some patients may ultimately be
referred to neuro-oncologists, it often falls upon general
internists, neurologists, or oncologists to initially manage
these issues. This book is intended for them.

Eudocia Quant, MD

ix

73724_FM00_Printer.indd ix 03/29/09 2:40:00 PM


73724_FM00_Printer.indd x 03/29/09 2:40:00 PM
C H A P T E R 1

Primary Brain Tumors

Epidemiology
■ Central Brain Tumor Registry of the United Status
(CBTRUS)1
• The overall incidence rate for 2004–2005 in the
United States for primary brain and CNS tumors was
23.62 per 100,000 person-years for adults (ages 201
years).
• The most frequently reported histology is meningioma
(33.4%), followed by glioblastoma (17.6%).
■ Mortality rates from nervous system tumors (including
meningiomas) in North America, Western Europe, and
Australia are approximately 4 to 7 per 100,000 persons
per year in men and 3 to 5 per 100,000 persons per year
in women.2

Histological Classification of Primary


Brain Tumors
■ Primary brain tumors rarely metastasize outside of the
CNS.
■ Grading of primary brain tumors is as follows:
• Conventional TNM grading is not used for primary
brain tumors.
• Several grading systems exist for primary brain tumors,
but the WHO classification is the most widely adopted
(Table 1-1).3
■ Tumors are graded I to IV, which correlates with

the degree of malignancy.


■ Subclassification is based on the morphologic

appearance of tumor cells.

73724_CH01_Printer.indd 1 03/29/09 2:35:00 PM


2 Chapter 1

Table 1-1: WHO Classification of Primary Brain Tumors

Classification WHO Grade (I–IV)

Astrocytic tumors

Pilocytic astrocytoma I

Diffuse astrocytoma II

Anaplastic astrocytoma III

Glioblastoma IV

Gliosarcoma IV

Gliomatosis cerebri

Oligodendroglial tumors

Oligodendroglioma II

Anaplastic oligodendroglioma III

Oligoastrocytic tumors

Oligoastrocytoma II

Anaplastic oligoastrocytoma III

Ependymal tumors

Subependymoma I

Myxopapillary ependymoma I

Ependymoma II

Anaplastic ependymoma III

Choroid plexus tumors

Choroid plexus papilloma I

Atypical choroid plexus papilloma II

Choroid plexus carcinoma III

(Continues)

73724_CH01_Printer.indd 2 03/29/09 2:35:00 PM


Primary Brain Tumors 3

Table 1-1: WHO Classification of Primary Brain


Tumors (Continued)

Classification WHO Grade (I–IV)

Neuronal and mixed neuronal-glial tumors

Gangliocytoma I

Ganglioglioma I

Anaplastic ganglioglioma III

Central neurocytoma II

Pineal tumors

Pineocytoma I

Pineoblastoma IV

Embryonal tumors

Medulloblastoma IV

Tumors of the cranial and peripheral nerves

Schwannoma I

Neurofibroma I

Malignant peripheral nerve II–IV


sheath tumor

Meningeal tumors

Meningioma I

Atypical meningioma II

Anaplastic/malignant meningioma III

Hemangioblastoma I

Tumors of the hematopoietic system

Primary CNS lymphoma

(Continues)

73724_CH01_Printer.indd 3 03/29/09 2:35:00 PM


4 Chapter 1

Table 1-1: WHO Classification of Primary Brain


Tumors (Continued)

Classification WHO Grade (I–IV)

Germ cell tumors

Germinoma

Teratoma

Choriocarcinoma

Tumors of the sellar region

Craniopharyngioma I

Data are from Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, eds.
WHO Classification of Tumors of the Central Nervous System, 4th ed.
Lyon: International Agency for Research on Cancer; 2007.

Risk Factors4
■ Genetic syndromes account for only a few cases.
• Neurofibromatosis 1: an autosomal dominant disorder
that is characterized by multiple neurofibromas, malig-
nant peripheral nerve sheath tumors, optic nerve
gliomas, café-au-lait spots, axillary/inguinal freckling,
and iris hamartomas.2
• Neurofibromatosis 2: an autosomal dominant disorder
that is characterized by schwannomas, meningiomas,
and gliomas.
• Tuberous sclerosis: an autosomal dominant disor-
der that is characterized by cortical tubers, facial
angiofibroma, subependymal nodules, and giant cell
astrocytomas.
• Retinoblastoma
• Li-Faumeni (TP53): an autosomal dominant disorder
that is characterized by multiple primary neoplasms
in children and young adults, with a predominance
of soft-tissue sarcomas, osteosarcomas, and breast
cancer, as well as an increased incidence of brain
tumors (mostly astrocytic gliomas).2

73724_CH01_Printer.indd 4 03/29/09 2:35:00 PM


Primary Brain Tumors 5

• Turcot syndrome: an autosomal dominant disorder


that is characterized by adenomatous colorectal pol-
yps, colon carcinomas, and malignant neuroepithelial
tumors.2
• Multiple hamartoma syndrome
■ Genetic polymorphisms: several polymorphisms have
been implicated, but too few studies have been con-
ducted to assure consistency.
■ Ionizing radiation exposure is a generally accepted cause
of primary brain tumors.
• This is based on data from bomb studies, nuclear test
fallout data, therapeutic radiation for cancer and benign
conditions, and occupational and environmental studies.
• Cranial irradiation (even low doses) can increase the
incidence of meningiomas by a factor of 10 and the
incidence of glial tumors by a factor of 3 to 7, with a
latency period of 10 years to more than 20 years.5
■ Nonionizing radiation exposure: the association of elec-
tromagnetic fields or radiofrequency cell phones with
brain tumors remains unresolved because of inconsis-
tency between studies.2,4

Diagnosis
■ MRI with gadolinium is generally the test of choice in
primary brain tumors.
• Imaging is useful for tumor diagnosis, preoperative
planning, intraoperative imaging, postoperative care,
and treatment response.6
• The appearance varies depending on histology (dis-
cussed later in this chapter).
• Imaging cannot substitute for tissue diagnosis because
it is difficult to differentiate between glioma histolo-
gies based on imaging alone.
■ Pathology (obtained via surgical biopsy or resection) is
often required for definitive diagnosis.

Gliomas (Table 1-2)


■ These arise from glial cells (astrocytes, oligodendrocytes,
ependymal cells, etc.).

73724_CH01_Printer.indd 5 03/29/09 2:35:00 PM


Table 1-2: Astrocytic Versus Oligodendroglial Tumors

Astrocytic Tumors Oligodendroglial Tumors

Diffuse Anaplastic Anaplastic

73724_CH01_Printer.indd 6
6 Chapter 1

Astrocytoma Astrocytoma Glioblastoma Oligodendroglioma Oligodendroglioma

WHO grade Grade II Grade III Grade IV Grade II Grade III

Demographics Peak incidence ages Mean age of diagnosis is Peak incidence ages Peak incidence ages Peak incidence ages
30–40, slight male approximately 45 years, 45–75, slight male 40–45, slight male 45–50, slight male
predominance slight male predominance predominance predominance predominance

Histopathology Well-differentiated Compared with Poorly differentiated, Diffusely infiltrating Compared with
neoplastic astrocytes diffuse astrocytoma, often pleomorphic gliomas of moderate oligodendroglioma
on background of increased cellularity, astrocytic tumor cells cellularity, composed of WHO grade II, mitotic
loosely structured distinct nuclear atypical, with marked nuclear monomorphic cells with activity, prominent
matrix mitotic activity. atypia and high mitotic uniform round nuclei and microvascular
Microvascular activity. Prominent perinuclear halos on proliferation, or
proliferation and necrosis microvascular paraffin sections. Other conspicuous necrosis
are absent. proliferation and/or features include
necrosis are essential. microcalcifications,
mucoid/cystic
degeneration, and a
dense network of
branching capillaries.

(continues)

03/29/09 2:35:00 PM
Table 1-2: Astrocytic Versus Oligodendroglial Tumors (Continued)

Astrocytic Tumors Oligodendroglial Tumors

Typical MRI Nonenhancing mass, Irregularly shaped mass Irregularly shaped, Nonenhancing mass, Heterogenous patterns

73724_CH01_Printer.indd 7
appearance T1-hypointense, with peritumoral edema, contrast-enhancing mass T1-hypointense, due to variable
T2-hyperintense typically contrast with necrotic center. T2-hyperintense presence of necrosis,
enhancing with necrotic Typically more extensive cystic degeneration,
center peritumoral edema than intratumoral
anaplastic gliomas hemorrhages, and
calcifications. Contrast
enhancement may be
patchy or
homogenous.

Median 5–10 years 2–3 years 9–12 years Depends on 1p/19q status: median survival in
survival patients with 1p/19q codeletions is more than
7 years compared with 2.8 years in patients with
1p/19q retained

Data are from Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, eds. WHO Classification of Tumors of the Central Nervous System, 4th ed. Lyon: International Agency
for Research on Cancer; 2007. Wen PY, Kesari S. Malignant gliomas in adults. N Engl J Med. 2008;359(5):494–507.
Primary Brain Tumors 7

03/29/09 2:35:00 PM
8 Chapter 1

■ Glioblastomas account for 54% of all gliomas.1


■ Most low-grade gliomas undergo malignant transforma-
tion to higher grade tumors.
■ The incidence of gliomas has increased slightly over the
past 2 decades, primarily because of improved diagnostic
imaging.7
■ They rarely spread outside of the CNS.

Astrocytoma
Pilocytic Astrocytoma (WHO Grade I/IV)
■ These are mostly seen in the pediatric population.

Diffuse Astrocytoma (WHO Grade II/IV)


■ Biology

• p53 tumor suppressor mutations are found in more


than 60% of astrocytomas.8
• The combined loss of 1p and 19q is a rare event in
low-grade astrocytomas.9
■ Clinical features

• The mean age of presentation is 34 years.6


• Patients may present with generalized or focal signs/
symptoms.
• There is a high rate of seizures in low-grade tumors
because of frequent involvement of the cortex and
slower growth than higher grade tumors.
■ MRI appearance: typically nonenhancing mass, hypoin-

tense on T1-weighted images, hyperintense on


T2-weighted images
■ Treatment for low-grade gliomas

• There is no standard of care, and treatment recom-


mendations vary widely.
• Studies of low-grade glioma often combine astrocy-
toma, oligoastrocytoma, and oligodendroglioma patient
populations.
• Surgery
■ Surgery is important for establishing a diagnosis.

■ It is controversial whether the extent of resection

improves outcomes.

73724_CH01_Printer.indd 8 03/29/09 2:35:00 PM


Primary Brain Tumors 9

• Gliomas are often infiltrative, diffuse, and/or


near eloquent brain regions.
• There is an increasing trend among neurosur-
geons toward aggressive resection.
• No randomized studies are available on the
extent of resection in low-grade gliomas, but
several retrospective reviews show improved
survival with aggressive tumor resection.10,11
■ Benefits of more extensive resection include

• Minimizing tumor burden


• Reducing symptoms
• Decreasing sampling error and improving diag-
nostic accuracy12
■ Results from a phase II study of 111 “good-risk”

patients with low-grade glioma (neurosurgeon-


defined gross total resection and age #40) followed
with serial MRI imaging13:
• Overall 5-year survival rate, 93%
• Five-year progression-free survival rate, 48%
• Factors associated with poorer progression-free
survival in multivariate analysis: preoperative
tumor diameter $4 cm, astrocytoma or oligoas-
trocytoma histologic type, and residual tumor
$1 cm on an MRI
• Radiation therapy
■ Radiation therapy is considered standard treatment

for low-grade gliomas.


■ The total dose is typically 50–54 Gy.

• A randomized, multicenter study of 379 patients


with low-grade glioma comparing lower dose
(45 Gy over 5 weeks) versus higher dose (59.4 Gy
over 6.6 weeks) conventional, limited-field,
external-beam radiation demonstrated no differ-
ence in overall survival or 5-year progression-
free survival.14
• A randomized, multicenter study of 211 patients
with low-grade glioma comparing lower dose
(50.5 Gy in 28 fractions) versus higher dose

73724_CH01_Printer.indd 9 03/29/09 2:35:00 PM


10 Chapter 1

(64.8 Gy in 36 fractions) conventional, limited-


field, external-beam radiation demonstrated no
difference in overall survival or progression-free
survival but did demonstratemore radiation-
induced toxicity with higher dose.15
■ Timing of radiation therapy

• A randomized, multicenter trial of 311 patients


with low-grade glioma comparing immediate
radiation therapy after resection versus delayed
radiation until tumor progression demonstrated
a significant difference in 5-year progression-
free survival in the early radiation arm over the
delayed radiation arm (55% vs. 34.6%) but no
difference in overall survival and it is unclear if
there is any impact on quality of life16
■ Long-term side effects of radiation

• There is concern for an effect on cognitive


functioning.
• Results from a longitudinal neuropsychological
study of patients with radiographically and clini-
cally stable low-grade glioma:
■ At mean of 6 years after diagnosis, results

suggested that the tumor itself (not radia-


tion) had the most deleterious effect on
cognitive functioning, and only high frac-
tion dose radiation (daily fractions .2 Gy)
was associated with additional cognitive
deterioration.17
■ At mean of 12 years after diagnosis, the

patients who received radiation therapy (even


those with daily fractions #2 Gy) showed a
progressive decline in attentional function-
ing, whereas those who never received radia-
tion therapy had stable radiological and
cognitive status.18
• Chemotherapy
■ There is interest in developing effective chemo-

therapy for low-grade glioma in an attempt to


spare patients the late toxicities associated with
radiation.

73724_CH01_Printer.indd 10 03/29/09 2:35:00 PM


Primary Brain Tumors 11

■ Randomized studies evaluating chemotherapy in


low-grade glioma
• A randomized, multicenter trial of radiation with
or without lomustine showed no difference in
survival.19
• A randomized, multicenter trial of high-risk
patients with low-grade glioma (defined as resid-
ual tumor or age .40) of radiation with or with-
out six cycles of adjuvant procarbazine, CCNU,
and vincristine (PCV) chemotherapy.20
■ The overall median survival and progression-

free survival were similar for both arms between


the years 0 and 2.
■ Beyond 2 years, the overall survival and

progression-free survival curves separated sig-


nificantly, favoring PCV and radiation therapy.
• Results are pending on a randomized, multi-
center trial of patients with low-grade glioma
comparing radiation versus temozolomide.
■ Phase II studies have demonstrated objective

responses with the following regimens.9


• Procarbazine, CCNU, and vincristine (PCV)
• Temozolomide
■ Prognosis
• Based on a Swiss population-based study, the median
overall survival for patients with low-grade astrocytoma
was 5.6 years.21
• In a study of newly diagnosed low-grade gliomas trea-
ted with temozolomide, methylated methylguanine-
methytransferase (versus unmethylated MGMT) and
loss of 1p/19q were associated with longer progression-
free survival.22

Anaplastic Astrocytoma (WHO Grade III/IV)


■ Biology

• They frequently arise from lower grade astrocytomas.


• They frequently undergo malignant transformation
into glioblastomas.
• p53 mutations and platelet-derived growth factor
receptor (PDGFR) overexpression are common.23

73724_CH01_Printer.indd 11 03/29/09 2:35:00 PM


12 Chapter 1

■ Clinical features
• Patients may present with generalized or focal signs/
symptoms.
• The median age at the time of diagnosis is 40 to 50 years.
■ MRI appearance: irregularly shaped mass with associated
edema, typically contrast enhancing
■ Treatment
• Most trials to date that include anaplastic astrocy-
tomas have lumped grade III and grade IV astrocy-
tomas together, although ongoing trials are beginning
to separate grade III from grade IV astrocytomas.
• Treatment decisions are often extrapolated from glio-
blastoma trials.
• Surgery
■ Surgery is important for establishing diagnosis.

■ Pathology should be taken from the contrast-

enhancing margin of the lesion rather than the


necrotic core.23
■ It is controversial whether the extent of resection

improves outcomes.
• Retrospective studies suggest that more aggres-
sive resections increase survival, but such stud-
ies are prone to bias.
• In a randomized study of 30 older patients (age
.65) with grade III or grade IV gliomas compar-
ing stereotactic biopsy with open craniotomy
and resection, median survival was longer after
resection (171 days vs. 85 days).24
• Radiation therapy
■ The role of radiation therapy is clearly established

in anaplastic astrocytoma.
■ For patients with adequate performance status, a

radiation therapy dose of 60 Gy in 30 fractions/day


is the standard of care.25
• Chemotherapy
■ The role of chemotherapy in newly diagnosed ana-

plastic astrocytoma is less well established than the


role of radiation therapy.

73724_CH01_Printer.indd 12 03/29/09 2:35:00 PM


Primary Brain Tumors 13

■ Preliminary results from a randomized, multicenter


trial of patients with newly diagnosed anaplastic
glioma randomized to sequential radiochemother-
apy (radiation therapy upfront followed by PCV or
temozolomide at first progression versus PCV or
temozolomide upfront followed by radiation therapy
at first progression):26
• There was no difference in time to failure (pro-
gression after radiotherapy and chemotherapy
in either sequence) or median overall survival
between initial radiotherapy and initial chemo-
therapy and between temozolomide and PCV.
• Patients with anaplastic astrocytoma had a worse
time to failure than those with anaplastic astro-
cytoma or anaplastic oligoastrocytoma.
■ Based on data from the glioblastoma population,27
there is an increasing trend toward using radia-
tion therapy with concurrent and adjuvant temo-
zolomide.
■ Results from a randomized, multicenter trial of 193
patients with newly diagnosed anaplastic astrocy-
toma comparing radiation therapy with concurrent
and adjuvant BCNU and dibromodulcitol (DBD)
versus radiation therapy alone:28
• This study demonstrated no statistically signifi-
cant difference in overall survival or progression-
free survival.
• The study terminated early because of decreas-
ing accrual.
• The 2-year survival rate was 56% in the com-
bined therapy arm versus 49% in the radiation
therapy-only arm.
• At central pathology review, 53% of the locally
diagnosed cases of anaplastic astrocytoma could
not be confirmed.
■ In a randomized multicenter trial of 290 patients
with newly diagnosed anaplastic oligodendro-
glioma or anaplastic oligoastrocytoma comparing

73724_CH01_Printer.indd 13 03/29/09 2:35:00 PM


14 Chapter 1

PCV chemotherapy followed by radiation therapy


versus radiation therapy alone, there was no
difference in overall survival or progression-free
survival.29
■ Results are pending on an ongoing randomized

phase III trial in anaplastic astrocytoma of radia-


tion therapy with temozolomide versus radiation
therapy with a nitrosourea.
• Recurrent disease
■ Chemotherapy is frequently given for recurrent

disease.
■ Preliminary results from a multicenter, randomized

phase III trial of 447 patients with chemotherapy-


naïve, recurrent grade III or grade IV astrocytomas
or oligoastrocytomas comparing temozolomide ver-
sus PCV chemotherapy:30
• The study demonstrated no difference in over-
all median survival comparing temozolomide
to PCV.
• There was no clear evidence of differential
treatment effect in either GBM or anaplastic
astrocytoma.
• A 5-day regimen of temozolomide was supe-
rior to a 21-day regimen of temozolomide
with respect to progression-free and overall
survival.
■ In a multicenter phase II trial of patients with ana-

plastic astrocytoma or anaplastic oligoastrocytoma


who recurred after radiation therapy, temozolo-
mide demonstrated good activity with 6-month
progression-free survival rate of 46% and an objec-
tive response rate of 35%.31
■ Several investigational agents have also been tested

in phase II clinical trials for malignant glioma


(i.e., grade III and grade IV gliomas), although none
has become a standard therapy yet.7
■ Prognosis
• Based on European population studies for patients
diagnosed during 1990–1994, the 1-year survival was
44%, and the 5-year survival was 16%.23

73724_CH01_Printer.indd 14 03/29/09 2:35:00 PM


Primary Brain Tumors 15

• Based on a Swiss population-based study, the median


overall survival for anaplastic astrocytoma was
1.6 years.21
• Based on a phase III clinical trial of radiation with
or without DBD and BCNU, the median overall
survival for anaplastic astrocytoma was 2 to 2.6
years.28
• Good prognostic factors are younger age (,50 years
old), good performance status, and intact neurologic
function.

Glioblastoma (WHO Grade IV/IV)


■ Biology

• Other histologic variants include gliosarcoma, giant-


cell glioblastoma, small-cell glioblastoma, and glio-
blastoma with oligodendroglial features.
• It may present de novo (primary GBM) or arise from a
lower grade tumor (secondary GBM).
• Genetic differences exist between primary and second-
ary GBMs.7
■ Primary GBMs are characterized by epidermal

growth factor receptor (EGFR) amplification and


mutations, loss of heterozygosity of chromosome
10q, deletion of PTEN, and p16 deletion.
■ Secondary GBMs are characterized by p53 muta-

tions, overexpression of PDGFR, abnormalities in


p16 and retinoblastoma pathways, and loss of
heterozygosity of chromosome 10q.
■ Clinical features

• GBMs may present with focal or generalized signs/


symptoms.
• The median age at the time of diagnosis is 64 years,7
although patients presenting with a primary GBM are
generally older than patients with a secondary GBM.
■ MRI appearance

• Heterogeneous enhancement with nonenhancing


necrotic center, surrounding edema, and mass effect
• Most commonly disseminate along white matter
tracts, especially the corpus callosum, to involve the
contralateral hemisphere

73724_CH01_Printer.indd 15 03/29/09 2:35:00 PM


16 Chapter 1

■ Treatment
• Primary and secondary GBMs are currently treated
similarly despite their genetic differences, but this
may change in the future.
• Surgery
■ This is important for establishing a diagnosis.

■ It is somewhat controversial about whether the

extent of resection improves outcomes, although


recent prospective studies suggest a survival benefit
for more extensive resections.
• Because of the infiltrating nature of GBMs, it is
not possible to remove the tumor in its entirety,
and thus, the extent of resection is defined by
the contrast-enhancing portion of the tumor on
postoperative MRI with contrast.
• Retrospective studies suggest that more aggres-
sive resections increase survival, but such stud-
ies are prone to bias.
• In a randomized study of 30 older patients (age
.65) with grade III or grade IV gliomas compar-
ing stereotactic biopsy with open craniotomy
and resection, median survival was longer after
resection (171 days vs. 85 days).24
• Results from a randomized study of 322 patients
with grade III or IV glioma comparing the extent
of resection by fluorescence-guided resection
with 5-aminolevulinic acid versus conventional
microsurgery:
■ Interim analysis suggested that patients with-

out residual contrast-enhancing tumor had a


higher overall median survival than those
with residual-enhancing tumor (17.9 months
vs. 12.9 months).32
■ Subgroup analysis of 243 patients with GBM

demonstrated treatment bias regarding com-


plete versus incomplete resection, but when
patients were stratified based on age and
eloquent location, survival advantages from
complete resection remained significant.33

73724_CH01_Printer.indd 16 03/29/09 2:35:00 PM


Primary Brain Tumors 17

• Newly diagnosed glioblastoma


■ The current standard of care for newly diagnosed

glioblastoma is radiation plus concomitant temozo-


lomide followed by adjuvant temozolomide.27
■ Based on a randomized phase III clinical trial,

initial treatment with radiation plus concomitant


temozolomide followed by adjuvant temozolo-
mide for six cycles increased the median overall
survival compared with radiation alone (15 vs.
12 months).
■ The regimen is well tolerated, with myelosuppres-

sion occurring in less than 10%.


■ There is no data showing an improvement in over-

all survival with prolonged adjuvant temozolomide.


■ Several studies have demonstrated correlations

between treatment outcome and expression of the


DNA repair enzyme methylguanine methyltrans-
ferase (MGMT).34
■ There are several phase II trials of promising tar-

geted molecular therapies added to the standard


radiation/temozolomide regimen, but none has
been adopted as a new standard of care.7
• Recurrent glioblastoma
■ There is no clear standard of care, but a bevacizumab-

containing regimen is often given to patients with


recurrent GBM.
■ FDA approval of bevacizumab for recurrent glio-

blastoma is based on several phase II clinical trials


with bevacizumab monotherapy35,36 or bevacizumab
plus irinotecan36–38 demonstrating prolongation
of progression-free survival and improvement
in response rates compared with historical
controls.
■ There are several phase II trials of other promising

targeted molecular therapies,7 but none has been


as widely adopted as bevacizumab.
■ Prognosis
• Based on a Swiss population-based study, the median
survival for glioblastoma was 0.4 years.21

73724_CH01_Printer.indd 17 03/29/09 2:35:00 PM


18 Chapter 1

• Based on the phase III clinical trial of radiation with


or without temozolomide, the median survival for glio-
blastoma was 12 to 15 months.27

Oligodendroglioma
Oligodendroglioma (WHO Grade II/IV)
■ Biology

• Loss of heterozygosity of 1p and 19q chromosomes is


associated with response to treatment and good
prognosis but is studied less extensively in low-grade
oligodendrogliomas than in anaplastic oligodendro-
gliomas.9
• Other less common genetic abnormalities in low-grade
oligodendroglioma include overexpression of platelet-
derived growth factor or receptor, epidermal growth
factor receptor, or vascular endothelial growth factor.39
• They arise in white matter, commonly in the frontal
lobes.
■ Clinical features

• Oligodendrogliomas may present with generalized or


focal signs/symptoms.
• There is a high rate of seizures in low-grade tumors
because of the frequent involvement of the cortex and
slower growth than higher grade tumors.
• The median age of presentation is 35 to 40 years.
■ MRI appearance

• MRI typically shows a nonenhancing mass, hyperin-


tense on T2-weighted imaging.
• Calcifications may be present and may be more con-
spicuous on gradient echo sequences.6
• Features that favor oligodendroglioma over astrocy-
toma include cortical involvement, the presence of
calcifications, and a heterogeneous signal.6
■ Treatment

• No standard of care exists for low-grade oligodendro-


glioma, and treatment recommendations vary widely.
• Treatment options are similar to low-grade astrocy-
toma (discussed previously in this chapter).

73724_CH01_Printer.indd 18 03/29/09 2:35:00 PM


Primary Brain Tumors 19

■ Prognosis
• Low-grade oligodendrogliomas have a better prognosis
than low-grade astrocytomas.
• Based on a Swiss population-based study, the median
overall survival for a low-grade oligodendroglioma was
11.6 years.21

Anaplastic Oligodendroglioma (WHO Grade III/IV)


■ Biology
• Loss of heterozygosity of 1p and 19q chromosomes is
associated with an increased response to treatment
and good prognosis.39
■ The incidence of either 1p or 19q deletions in oli-

godendrogliomas is approximately 75%.40


■ The incidence of combined 1p 19q loss in oligo-

dendrogliomas is 60% to 70%.40


■ The combined loss of 1p 19q is mediated by trans-

location of 1 and 19q.41,42


• Other less common genetic changes include amplifi-
cation of CDK4 or MYC, mutations of CDKN2A or
CDKN2C, and deletions on chromosome 10.39
• Progression from low-grade oligodendroglioma to ana-
plastic oligodendroglioma is associated with defects in
PTEN, retinoblastoma, p53, and cell cycle pathways.7
■ Clinical features: generalized or focal signs/symptoms

■ MRI appearance6

• Marked enhancement is associated with anaplastic


grades.
• Calcifications may be present and may be more con-
spicuous on gradient echo sequences.
• Features that favor oligodendroglioma over astrocy-
toma include cortical involvement, the presence of
calcifications, and a heterogeneous signal.
■ Treatment

• No standard of care is set for anaplastic oligodendro-


glioma.
• The best treatment regimen is unknown, and treat-
ment recommendations vary widely.
• Surgery

73724_CH01_Printer.indd 19 03/29/09 2:35:00 PM


20 Chapter 1

■This is important for establishing a diagnosis.


■Pathology should be taken from the contrast-
enhancing margin of the lesion rather than the
necrotic core.23
■ It is controversial whether the extent of resection

improves outcomes.
• Radiation therapy
■ Randomized trials in malignant gliomas demonstrate

a survival advantage with adjuvant radiation therapy,


but no trials have specifically addressed radiation
therapy in the anaplastic oligodendroglioma.40
• Chemotherapy
■ Two large multicenter randomized phase III trials

of newly diagnosed anaplastic oligodendroglioma


showed that the addition of PCV chemotherapy to
radiation does not prolong overall survival but does
increase progression-free survival.
• One study included 368 patients with anaplas-
tic oligoastrocytoma and oligodendroglioma
randomized to standard radiation therapy alone
or radiation therapy followed by six cycles of
adjuvant PCV (procarbazine, lomustine,
vincristine).43
• The other study included 289 patients with ana-
plastic oligodendroglioma randomized to stan-
dard radiation therapy alone or four cycles of
neoadjuvant dose-intensive PCV followed by
radiation therapy.29
• In both trials, 80% of patients assigned to radia-
tion therapy alone were treated with chemother-
apy at progression.
• There was no effect of treatment on survival
even in the subgroup with 1p 19q codeletion.
• The subgroup with 1p 19q codeletion had a lon-
ger median survival than those with 1p 19q
retained.
• The addition of PCV to radiation was associated
with greater toxicity than radiation therapy
alone.

73724_CH01_Printer.indd 20 03/29/09 2:35:00 PM


Primary Brain Tumors 21

■ Temozolomide is effective in newly diagnosed


anaplastic oligodendroglioma (based on phase II
trials44–46) and is less toxic than PCV chemotherapy,26
but it is unclear whether temozolomide is equiva-
lent, inferior, or superior to PCV chemotherapy.
■ Results are pending in several ongoing randomized

phase III trials in anaplastic oligodendroglioma.


• CATNON: radiation plus temozolomide versus
radiation alone in anaplastic glioma without 1p
19q loss.
• Codeleted trial: radiation alone versus temozolo-
mide alone versus radiation plus temozolomide
in newly diagnosed anaplastic oligodendro-
glioma or anaplastic oligoastrocytoma with 1p
19q codeletions.
• Recurrent anaplastic oligodendroglioma
■ 1p 19q codeleted tumors that previously responded

to chemotherapy will often respond again, although


for a shorter period.
■ Procarbazine, CCNU, and vincristine chemother-

apy (PCV) produced favorable responses in several


phase II studies of recurrent anaplastic oligo-
dendroglioma.47,48
• Radiographic response rates were 62% to 73%.
• Median time to progression was 12 to 24
months.
• Toxicities, including hematologic and gastroin-
testinal side effects, limit the duration of PCV
administration.
■ Temozolomide produced favorable responses in

several phase II studies of recurrent anaplastic oli-


godendroglioma.
• Response rates were 25% to 44% following pro-
gression on PCV chemotherapy.49,50
• The response rate was 53% after progression on
surgery and radiation.51
■ Prognosis
• Anaplastic oligodendrogliomas have a better prognosis
than anaplastic astrocytomas.

73724_CH01_Printer.indd 21 03/29/09 2:35:00 PM


22 Chapter 1

• Based on a Swiss population-based study, median


overall survival for anaplastic oligodendroglioma was
3.5 years.21
• Based on randomized phase III trials of radiation ther-
apy with or without PCV chemotherapy in anaplastic
oligodendroglioma and oligoastrocytoma, median
overall survival was 2.6 to 4.9 years.29,43
■ The difference in overall survival between two trials

may be explained by stricter selection criteria in


one trial resulting in a larger number of patients
with 1p 19q codeletions and with fewer anaplastic
features.

Meningioma
■ Tumors arising from arachnoid (meningothelial) cap cells
■ Most common type of primary brain tumor, accounting
for 33.4% of all primary brain tumors, with an annual
incidence rate of 6.03 per 100,000 person-years1
■ Twice as common in women than men1

Risk Factors
■ Ionizing radiation52
• Cranial radiotherapy for gliomas, leukemias, lympho-
mas, or cerebral metastases is associated with the
development of meningiomas within the previous radi-
ation field after a median latency period of 24 years.
• Most meningiomas associated with radiotherapy are
higher grade with a high proliferation index.
■ Hereditary syndromes, including neurofibromatosis type 2,

Gorlin syndrome, and Cowden syndrome


■ Other suspected factors with insufficient evidence to

determine definitively whether they increase the risk for


meningiomas include cellular phones and head injury.

Clinical Features
■ The most common locations (in descending order) are

the convexities, parasagittal areas, sphenoid and middle


cranial fossa, frontal base, posterior fossa, cerebellar

73724_CH01_Printer.indd 22 03/29/09 2:35:00 PM


Primary Brain Tumors 23

convexity, cerebellopontine angle, intraventricular, and


clivus.53
■ Spinal meningiomas are most often found in the thoracic
spine.
■ Symptoms depend on location (Table 1-3).
• Parasagittal meningiomas occur most frequently in the
frontal lobe and can grow to a large size before causing
symptoms such as seizures or papilledema.
• Frontal skull base meningiomas may present with
vision changes, headache, anosmia, mental status
changes, and seizures.

Pathology
■ WHO grades

• Benign meningiomas (WHO grade I/IV)


■ Most common grade

■ Subtypes: meningothelial, fibrous (fibroblastic),

transitional (mixed), psammomatous, angiomatous,


microcystic, secretory, lymphoplasmacyte rich, and
metaplastic
• Atypical meningiomas (WHO grade II/IV)
■ Represent 5% to 7% of all meningiomas

■ Subtypes: atypical, clear cell, and choroid

■ Compared with benign meningiomas, increased

mitotic activity, increased cellularity, small cells


with high nucleus:cytoplasm ratio, prominent
nucleoli, pattern-less or sheet-like growth, or foci
of spontaneous or geographic necrosis
• Anaplastic/malignant meningiomas (WHO grade III/IV)
■ Less common

■ Subtypes: rhabdoid, papillary, and anaplastic

(malignant)
■ Other pathologic changes

• Genetic changes
■ Deletion of chromosome 22q, which contains the

NF2 gene
• Sex hormones54
■ Progesterone receptors are found in 76% of menin-

giomas, mostly benign meningiomas.

73724_CH01_Printer.indd 23 03/29/09 2:35:00 PM


Table 1-3: Meningioma Location and Associated Typical Clinical Presentations

Meningioma Location Clinical Presentation

73724_CH01_Printer.indd 24
24 Chapter 1

Parasagittal and falcine Anterior one third Headache, mental status changes

Middle one third Jacksonian seizures, progressive hemiparesis

Posterior one third Headache, visual symptoms, seizures, mental status changes

Sphenoid wing Lateral/pterional Similar to convexity tumors

Middle one third (alar) Hemiparesis, dysphagia

Medial (clinoidal) Visual acuity/field disturbance due to optic nerve compression, proptosis, cranial
nerve dysfunction (III, IV, V, VI)

Olfactory groove Foster Kennedy syndrome (anosmia, ipsilateral optic atrophy with contralateral
papilledema), frontal lobe syndromes, mental status changes, urinary
incontinence, seizure

(continues)

03/29/09 2:35:00 PM
Table 1-3: Meningioma Location and Associated Typical Clinical Presentations (Continued)

Meningioma Location Clinical Presentation

Tuberculum sella/ Visual acuity/field disturbance, anosmia, hydrocephalus, endocrinologic

73724_CH01_Printer.indd 25
suprasellar syndromes

Cavernous sinus Cranial nerve deficits (III, IV, V, VI)

Cerebellopontine angle Hearing loss, facial pain/numbness/weakness/spasms, headaches,


cerebellar signs

Foramen magnum Unilateral cervical pain, extremity motor and sensory loss (clockwise
involvement), cold and clumsy hands with intrinsic hand atrophy

Petroclival Hearing loss, vertigo, tinnitus, facial pain, cranial nerve deficits (V, VI, VIII, VIII)

Adapted from Asthagiri AR, Helm GA, Sheehan JP. Current concepts in management of meningiomas and schwannomas. Neurol Clin. 2007;
25(4):1209–1230.
Primary Brain Tumors 25

03/29/09 2:35:00 PM
26 Chapter 1

■ More aggressive meningiomas are associated with


low numbers or absence of progesterone receptors.
■ Estrogen receptors are found in 19% of meningiomas.

Imaging
■ Characteristic findings on MRI with gadolinium include
the following6:
• Sessile or pedunculated mass that homogeneously and
intensely enhances
• CSF cleft sign between the tumor and the brain
• Involvement of the dura with a dural tail sign in 40%
to 60%
• Intratumoral calcification may be present
• Presence of peritumoral edema is variable
■ Staging for metastatic disease

• There are no consensus guidelines regarding systemic


staging but meningioma metastases are uncommon;
thus, further staging is unnecessary in most cases.
• The most common sites of metastases are lung, bone,
liver, and lymph nodes.
• If lung metastases are suspected, then staging should
include chest CT with contrast.

Treatment
■ Surgery

• Approximately 80% of meningiomas (mostly grade I


meningiomas) can be cured by surgery.52
• The goal is for complete resection, including the dural
attachment and infiltrated bone.
• The extent of resection is associated with the recur-
rence rate.53
• Complete resection is not achievable in some patients
because of inaccessibility or proximity to vital struc-
tures, such as the cavernous sinus and clivus.
• Preoperative embolization may facilitate removal and
improve surgical outcomes by reducing vascularity and
operative blood loss, but embolization is associated with
a risk of ischemic and hemorrhagic complications.

73724_CH01_Printer.indd 26 03/29/09 2:35:00 PM


Primary Brain Tumors 27

■ Radiation therapy
• Focal external beam radiation therapy is considered
standard treatment for recurrent meningiomas and
after surgery for atypical and malignant meningiomas.
• Stereotactic radiosurgery can also be considered for
small lesions (,3 cm) and/or for locations not amena-
ble to surgery (i.e., cavernous sinus).
• The use and timing of radiation after resection of a
benign meningioma are controversial.
■ Some centers withhold radiation, even after subto-

tal resection of benign meningioma, until the time


of progression.
■ In patients with WHO grade I meningiomas, radi-

ation in single daily doses of 1.8–2.0 Gy to total


doses of 45–60 Gy after subtotal resection pro-
duces comparable 5-year progression-free survival
rates to rates after complete surgical resection.52
• Radiation is often provided after resection, even after
total resection, in atypical and anaplastic meningiomas.
■ Medical therapy52,55
• Mostly used as investigational agents or after pro-
gression despite multiple surgeries and/or radiation
treatments
• Agents with possible benefit (mostly based on pilot
studies and phase II studies) include the following:
■ Interferon-a

■ Somatostatin analogues

■ Hydroxyurea

■ Observation
• There are no clear guidelines on observation versus
treatment.
• The decision for treatment depends on clinical history,
severity of symptoms, rate of growth, amenability of
the tumor to surgery, and the estimated benefit of
treatment.
■ Patients with small asymptomatic meningiomas

can be followed with serial imaging.


■ Atypical or anaplastic meningiomas should be

treated.

73724_CH01_Printer.indd 27 03/29/09 2:35:00 PM


28 Chapter 1

Prognosis
■ The overall 5-year survival ranges from 69% to 90%
depending on the study.52
■ Atypical and anaplastic meningiomas

• Poorer prognosis than benign meningiomas


• Usually demonstrate recurrence and aggressive growth
rates regardless of treatment modality (extirpation,
radiosurgery, or external beam radiation therapy)53
■ Unfavorable prognostic factors include old age, male

gender, and significant comorbidities56


■ Tumor growth rates based on volumetric analysis57

• In study of patients with WHO grade I meningiomas


who underwent subtotal resection, doubling time was
5.2 years.
• Growth rates were higher in young patients.
• Growth rates were lower in meningiomas with calcifi-
cations and in asymptomatic meningiomas.
■ Recurrences

• High recurrence rates are associated with high histo-


logical grading, papillary and hemangiopericytic mor-
phology, large tumor size, and high mitotic index.52
• In one series of 936 meningiomas, the 5-year recur-
rence rate after complete resection was 3% in benign
meningiomas, 38% in atypical meningiomas, and 78%
in anaplastic meningiomas.

Primary CNS Lymphoma (PCNSL)


■ Rare form of extranodal non-Hodgkin lymphoma con-
fined to the brain, leptomeninges, spinal cord, and eyes
■ Accounted for 2.5% of all primary brain and CNS tumors
diagnosed in the United States between 2004 and 20051

Risk Factors
■ Congenital or acquired immunodeficiency (discussed
later in this chapter) is a risk factor.
■ HIV infection is associated with a 3,600-fold increased

risk of developing PCNSL.58

73724_CH01_Printer.indd 28 03/29/09 2:35:00 PM


Primary Brain Tumors 29

Pathology
■ Ninety percent of PCNSL are diffuse large B-cell
lymphoma (DLBCL), with the remaining 10% poorly
characterized low-grade lymphomas, Burkitt lymphomas,
and T-cell lymphomas.59
■ The DLBCL type is composed of immunoblasts or cen-

troblasts with a predilection for blood vessels, resulting in


lymphoid clustering around small cerebral vessels.60
■ They are classified as stage I disease according to the Ann

Arbor staging system, but the prognostic significance of


the Ann Arbor staging system does not apply to PCNSL.

Clinical Features
■ The median age of diagnosis in immunocompetent pati-

ents is 53 to 57 years.60
■ Patients present with neurologic signs and symptoms

depending on the location of the tumor, including focal


deficits, neuropsychiatric symptoms, increased intracra-
nial pressure, seizures, and ocular symptoms.
■ Sixteen percent to 42% of patients with PCNSL have

leptomeningeal involvement at diagnosis, but most do


not show clinical signs of leptomeningeal disease.60
■ Twenty percent have ocular involvement, presenting as

floaters, blurred vision, diminished visual acuity, and


painful red eyes.60
■ Systemic B symptoms (fever, weight loss, night sweats)

are less common, unlike systemic DLBCL.


■ Occult systemic disease has been reported in up to 8% of

patients initially thought to have isolated PCNSL.61

Diagnosis61
■ A definitive diagnosis usually requires a stereotactic

biopsy.
■ MRI with gadolinium of the entire neuroaxis

• Lesions appear clearly delineated and are isointense


to hypointense on T2-weighted MRI because of high
cell density and scant cytoplasm.

73724_CH01_Printer.indd 29 03/29/09 2:35:00 PM


30 Chapter 1

• Lesions are typically homogeneously enhancing on


postcontrast T1-weighted MRI in immunocompetent
patients.
• Lesions are solitary in 65% of patients and multifocal
in 35%.60
• Locations include cerebral hemispheres (38%),
thalamus/basal ganglia (16%), corpus callosum (14%),
ventricular region (12%), and cerebellum (9%).60
• Lesions may appear to vanish with steroids because
steroids reduce swelling and cause tumor lysis.
■ Lumbar puncture with cytology, flow cytometry, and
immunoglobulin heavy-chain gene rearrangement studies
• CSF typically demonstrates increased WBC count,
high protein concentrations, and low glucose.
■ Complete ophthalmologic examination, including dilated
fundus and slip-lamp examinations
■ Other recommended studies to look for systemic disease
• Chest, abdomen, and pelvic CT scan with contrast
• Bone marrow biopsy
• Clinical and ultrasound testicular examination in men
• Clinical examination of peripheral lymph nodes
• Routine blood work including HIV testing, CBC,
LDH
• Possibly PET scan

Treatment
■ PCNSLs are sensitive to corticosteroids, but responses

are not durable.


■ There is no standard treatment.

■ There are no randomized, controlled phase III trials in

PCNSL.
■ Treatment options are based on the results from phase II

clinical trials.62
• Newly diagnosed PCNSL
■ Surgery

• Stereotactic biopsy is indicated for diagnosis.


• Gross total resection or tumor debulking confers
no survival benefit over surgery alone.60

73724_CH01_Printer.indd 30 03/29/09 2:35:00 PM


Primary Brain Tumors 31

■ High-dose methotrexate monotherapy


• CNS penetration of methotrexate is poor at con-
ventional doses.
• High doses (3 to 8 g/m2) can achieve cytocidal
concentrations of methotrexate in cerebrospinal
fluid.63
• High-dose methotrexate monotherapy is associ-
ated with response rates of 35% to 74% and with
median progression-free survival of 12.8–13.7
months.
• High doses $3 g/m2 require intensive inpatient
management and good renal function.
■ Methotrexate-based multidrug regimens
• The incremental benefit from additional chemo-
therapy beyond high-dose methotrexate is
unclear.
■ Preliminary results from a randomized phase

II study of high-dose methotrexate (followed


by whole brain radiation) with or without
high-dose cytarabine found that the addition
of cytarabine increased response rates from
40% to 69%.64
• Radiographic response rates and disease-free
survival may be slightly higher in trials with
methotrexate-based multidrug regimens com-
pared with trials with methotrexate monother-
apy, but rates often overlap.62
• Multidrug regimens may be associated with
higher toxicity than methotrexate monotherapy.
• Common regimens include methotrexate, pro-
carbazine, and vincristine (MPV); high-dose
cytarabine and methotrexate; and temozolomide
and methotrexate in older patients (age .60).62
■ Intrathecal chemotherapy
• Intrathecal chemotherapy is standard treatment
for patients with proven leptomeningeal disease.
• However, the use of adjuvant intrathecal che-
motherapy with high-dose methotrexate-based
regimens is controversial because methotrexate

73724_CH01_Printer.indd 31 03/29/09 2:35:00 PM


32 Chapter 1

doses $3 g/m2 can achieve adequate concentra-


tions in the CSF.62
■ Adjuvant corticosteroids
• Corticosteroids are useful for edema and symp-
tomatic management.
• Incorporation of corticosteroids into multidrug
regimens has not increased response rates com-
pared with non-steroid-containing regimens and
is associated with side effects.
■ High-dose chemotherapy with autologous stem-cell
transplantation
• Based on results from several phase II clinical
trials, this approach is feasible in a selected pop-
ulation (younger and chemotherapy sensitive).
• It is unclear whether patients in transplantation
trials would have done as well with standard
high-dose methotrexate-based chemotherapy
regimens.
• Studies show that transplantation is associated
with higher mortality and morbidity than chemo-
therapy regimens without transplantation.
■ Whole-brain radiation (WBRT)
• WBRT was historically the standard of care in
PCNSL but has been replaced by high-dose
methotrexate-based regimens.
• WBRT is often deferred, especially in patients
older than 60 years of age and depending on
response to chemotherapy.
■ WBRT alone does not produce durable remis-

sions in most patients.65


■ It is associated with neurotoxicity, including

dementia, gait disturbance, and urinary incon-


tinence (see Chapter 5 for additional infor-
mation regarding radiation-related toxicities).
■ Adjuvant rituximab
• Rituximab is a humanized monoclonal antibody
to CD20, a cell surface protein found on mature
B cells.
• It may be incorporated into treatment regimens
for patients with DLBCL, which express
CD20.

73724_CH01_Printer.indd 32 03/29/09 2:35:00 PM


Primary Brain Tumors 33

• Response rates with rituximab monotherapy in


PCNSL are modest.
• Rituximab combined with methotrexate, procar-
bazine, vincristine, and cytarabine produced a
high response rate of 93% in a phase II clinical
trial.66
• There are several ongoing trials of rituximab in
PCNSL.
■ Recurrent or refractory PCNSL
• Up to 50% of patients with PCNSL will relapse,
and 10% to 15% of patients have primary refractory
PCNSL.62
• Optimal management is unclear because of limited
data.
• Options include whole-brain radiation (for patients
who had this treatment option initially deferred),
reinduction with high-dose methotrexate, single
agent topotecan, and single agent temozolomide.
■ Ocular lymphoma
• Ocular lymphoma may occur in isolation or in com-
bination with parenchymal PCNSL.
• Symptoms are similar to a nonspecific uveitis but
eventually become refractory to topical steroids.
• Definitive diagnosis is made with vitreal cytology.
• For those with isolated ocular lymphoma, the risk of
developing brain involvement is as high as 80%.62
• Treatment may include the following:
■ Primary intraocular lymphoma: directed ocular

treatment with ocular radiotherapy or intraocu-


lar methotrexate.
■ Parenchymal brain and ocular lymphoma: the

addition of directed ocular treatment may


improve disease control, but a methotrexate dose
of 8 g/m2 can also produce cytocidal concentra-
tions in the vitreous body.

Prognosis
■ Durable complete responses and long-term survival are

possible with treatment.


■ Outcomes are worse compared with patients with a simi-

larly staged systemic non-Hodgkin lymphoma.

73724_CH01_Printer.indd 33 03/29/09 2:35:00 PM


34 Chapter 1

Table 1-4: Prognostic Factors in PCNSL and Estimated


2-Year Overall Survival Rates

Poor Prognostic • Age .60 years


Factors • Performance status of 1 on the Eastern
Cooperative Oncology Group (ECOG)
performance status scale
• Elevated serum LDH
• High CSF protein concentration
• Tumor location in the deep brain regions
(periventricular regions, basal ganglia,
brainstem and/or cerebellum)

Number of Poor Prognostic


Factors Two-Year Overall Survival Rates

0–1 80%

2–3 48%

4–5 15%

Data are from Ferreri AJ, Blay JY, Reni M, et al. Prognostic scoring system
for primary CNS lymphomas: the International Extranodal Lymphoma
Study Group experience. J Clin Oncol. 2003;21(2):266–272.

■ Poor prognostic factors in PCNSL and 2-year survival


rates are listed in Table 1-4.67

HIV-Related PCNSL
■ Nearly 6% of the AIDS population will be afflicted with
PCNSL.6
■ The disease incidence has decreased during the highly

active antiretroviral therapy (HAART) era.68


■ MRI with gadolinium in HIV-related PCNSL

• This may demonstrate ring enhancing lesions, which


makes it difficult to differentiate from toxoplasmosis.
• Compared with PCNSL in immunocompetent
patients, there is a higher frequency of multiple lesions,
cortical-based lesions, lesions with irregular margins,
heterogeneity, and hemorrhage.6

73724_CH01_Printer.indd 34 03/29/09 2:35:00 PM


Primary Brain Tumors 35

■ Pathophysiology involves Epstein-Bar virus (EBV).


■ Optimal management is unclear, but high-dose methotrex-
ate, whole-brain radiation, and/or HAART may be used.62

References
1. CBTRUS. Statistical report: primary brain and central
nervous system tumors diagnosed in the United Status in
2004–2005. Hinsdale, IL: Central Brain Tumor Registry of
the United States; 2009. http://www.cbtrus.org/reports//
2007–2008/2007report.pdf. Accessed August 15, 2009.
2. Ohgaki H. Epidemiology of brain tumors. Methods Mol Biol.
2009;472:323–342.
3. Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, eds. WHO
Classification of Tumours of the Central Nervous System, 4th ed.
Lyon: International Agency for Research on Cancer; 2007.
4. Bondy ML, Scheurer ME, Malmer B, et al. Brain tumor epi-
demiology: consensus from the Brain Tumor Epidemiology
Consortium. Cancer. 2008;113(7 Suppl):1953–1968.
5. DeAngelis LM. Brain tumors. N Engl J Med. 2001;344(2):
114–123.
6. Mechtler L. Neuroimaging in neuro-oncology. Neurol Clin.
2009;27(1):171–201, ix.
7. Wen PY, Kesari S. Malignant gliomas in adults. N Engl J Med.
2008;359(5):492–507.
8. Okamoto Y, Di Patre PL, Burkhard C, et al. Population-based
study on incidence, survival rates, and genetic alterations of
low-grade diffuse astrocytomas and oligodendrogliomas. Acta
Neuropathol. 2004;108(1):49–56.
9. Lang FF, Gilbert MR. Diffusely infiltrative low-grade gliomas
in adults. J Clin Oncol. 2006;24(8):1236–1245.
10. Claus EB, Horlacher A, Hsu L, et al. Survival rates in patients
with low-grade glioma after intraoperative magnetic reso-
nance image guidance. Cancer. 2005;103(6):1227–1233.
11. Smith JS, Chang EF, Lamborn KR, et al. Role of extent of
resection in the long-term outcome of low-grade hemi-
spheric gliomas. J Clin Oncol. 2008;26(8):1338–1345.
12. Jackson RJ, Fuller GN, Abi-Said D, et al. Limitations of ste-
reotactic biopsy in the initial management of gliomas. Neuro
Oncol. 2001;3(3):193–200.
13. Shaw EG, Berkey B, Coons SW, et al. Recurrence following
neurosurgeon-determined gross-total resection of adult
supratentorial low-grade glioma: results of a prospective clin-
ical trial. J Neurosurg. 2008;109(5):835–841.

73724_CH01_Printer.indd 35 03/29/09 2:35:00 PM


36 Chapter 1

14. Karim AB, Maat B, Hatlevoll R, et al. A randomized trial on


dose-response in radiation therapy of low-grade cerebral
glioma: European Organization for Research and Treatment
of Cancer (EORTC) Study 22844. Int J Radiat Oncol Biol
Phys. 1996;36(3):549–556.
15. Shaw E, Arusell R, Scheithauer B, et al. Prospective ran-
domized trial of low- versus high-dose radiation therapy in
adults with supratentorial low-grade glioma: initial report
of a North Central Cancer Treatment Group/Radiation
Therapy Oncology Group/Eastern Cooperative Oncology
Group study. J Clin Oncol. 2002;20(9):2267–2276.
16. van den Bent MJ, Afra D, de Witte O, et al. Long-term effi-
cacy of early versus delayed radiotherapy for low-grade astro-
cytoma and oligodendroglioma in adults: the EORTC 22845
randomised trial. Lancet. 2005;366(9490):985–990.
17. Klein M, Heimans JJ, Aaronson NK, et al. Effect of radio-
therapy and other treatment-related factors on mid-term to
long-term cognitive sequelae in low-grade gliomas: a com-
parative study. Lancet. 2002;360(9343):1361–1368.
18. Douw L, Klein M, Fagel SS, et al. Cognitive and radiological
effects of radiotherapy in patients with low-grade glioma:
long-term follow-up. Lancet Neurol. 2009;8(9):810–818.
19. Eyre HJ, Crowley JJ, Townsend JJ, et al. A randomized trial
of radiotherapy versus radiotherapy plus CCNU for incom-
pletely resected low-grade gliomas: a Southwest Oncology
Group study. J Neurosurg. 1993;78(6):909–914.
20. Shaw E, Wang M, Coons SW, et al. Final report of Radiation
Therapy Oncology Group (RTOG) protocol 9802: Radiation
therapy (RT) versus RT + procarbazine, CCNU, and vin-
cristine (PCV) chemotherapy for adult low-grade glioma
(LGG). J Clin Oncol. 2008;26(Suppl):abstr 2006.
21. Ohgaki H, Kleihues P. Population-based studies on inci-
dence, survival rates, and genetic alterations in astrocytic
and oligodendroglial gliomas. J Neuropathol Exp Neurol.
2005;64(6):479–489.
22. Everhard S, Kaloshi G, Criniere E, et al. MGMT methyla-
tion: a marker of response to temozolomide in low-grade
gliomas. Ann Neurol. 2006;60(6):740–743.
23. Stupp R, Reni M, Gatta G, Mazza E, Vecht C. Anaplastic
astrocytoma in adults. Crit Rev Oncol Hematol. 2007;63(1):
72–80.
24. Vuorinen V, Hinkka S, Farkkila M, Jaaskelainen J. Debulking
or biopsy of malignant glioma in elderly people - a ran-
domised study. Acta Neurochir (Wien). 2003;145(1):5–10.

73724_CH01_Printer.indd 36 03/29/09 2:35:00 PM


Primary Brain Tumors 37

25. Bleehen NM, Stenning SP. A Medical Research Council


trial of two radiotherapy doses in the treatment of grades 3
and 4 astrocytoma. The Medical Research Council Brain
Tumour Working Party. Br J Cancer. 1991;64(4):769–774.
26. Wick W, Weller M, Neurooncology Working Group of the
German Cancer Society. Randomized phase III study of
sequential radiochemotherapy of oligoastrocytic tumors of
WHO-grade III with PCV or temozolomide: NOA-04. J Clin
Oncol. 2008;26(Suppl):abstr LBA 2007.
27. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy
plus concomitant and adjuvant temozolomide for glioblas-
toma. N Engl J Med. 2005;352(10):987–996.
28. Hildebrand J, Gorlia T, Kros JM, et al. Adjuvant dibromod-
ulcitol and BCNU chemotherapy in anaplastic astrocytoma:
results of a randomised European Organisation for Research
and Treatment of Cancer phase III study (EORTC study
26882). Eur J Cancer. 2008;44(9):1210–1216.
29. Cairncross G, Berkey B, Shaw E, et al. Phase III trial of che-
motherapy plus radiotherapy compared with radiotherapy
alone for pure and mixed anaplastic oligodendroglioma:
Intergroup Radiation Therapy Oncology Group Trial 9402.
J Clin Oncol. 2006;24(18):2707–2714.
30. Brada M, Gabe R, Stenning S, Thompson L, Lee SM.
A randomised trial of temozolomide vs PCV chemotherapy
for recurrent malignant glioma (MRC BR12). Paper pre-
sented at the 33rd European Society for Medical Oncology
(ESMO) Congress; September 12–16, 2008; Stockholm,
Sweden.
31. Yung WK, Prados MD, Yaya-Tur R, et al. Multicenter phase
II trial of temozolomide in patients with anaplastic astrocy-
toma or anaplastic oligoastrocytoma at first relapse.
Temodal Brain Tumor Group. J Clin Oncol. 1999;17(9):
2762–2771.
32. Stummer W, Pichlmeier U, Meinel T, Wiestler OD, Zanella F,
Reulen HJ. Fluorescence-guided surgery with 5-aminolevulinic
acid for resection of malignant glioma: a randomised controlled
multicentre phase III trial. Lancet Oncol. 2006;7(5):392–401.
33. Stummer W, Reulen HJ, Meinel T, et al. Extent of resection
and survival in glioblastoma multiforme: identification
of and adjustment for bias. Neurosurgery. 2008;62(3):
564–576.
34. Stupp R, Hegi ME, Gilbert MR, Chakravarti A. Chemoradio-
therapy in malignant glioma: standard of care and future
directions. J Clin Oncol. 2007;25(26):4127–4136.

73724_CH01_Printer.indd 37 03/29/09 2:35:00 PM


38 Chapter 1

35. Kreisl TN, Kim L, Moore K, et al. Phase II trial of single-


agent bevacizumab followed by bevacizumab plus irinotecan
at tumor progression in recurrent glioblastoma. J Clin Oncol.
2009;27(5):740–745.
36. Cloughesy TF, Prados MD, Wen PY, et al. A phase II, ran-
domized, non-comparative clinical trial of the effect of beva-
cizumab (BV) alone or in combination with irinotecan (CPT)
on 6-month progression free survival (PFS6) in recurrent,
treatment-refractory glioblastoma (GBM). J Clin Oncol
(Meeting Abstracts). 2008;26(Suppl):abstr 2010b.
37. Vredenburgh JJ, Desjardins A, Herndon JE II, et al. Phase II
trial of bevacizumab and irinotecan in recurrent malignant
glioma. Clin Cancer Res. 2007;13(4):1253–1259.
38. Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, et al. Beva-
cizumab plus irinotecan in recurrent glioblastoma multiforme.
J Clin Oncol. 2007;25(30):4722–4729.
39. Cairncross G, Jenkins R. Gliomas with 1p/19q codeletion:
a.k.a. oligodendroglioma. Cancer J. 2008;14(6):352–357.
40. van den Bent MJ, Reni M, Gatta G, Vecht C. Oligoden-
droglioma. Crit Rev Oncol Hematol. 2008;66(3):262–272.
41. Griffin CA, Burger P, Morsberger L, et al. Identification of
der(1;19)(q10;p10) in five oligodendrogliomas suggests
mechanism of concurrent 1p and 19q loss. J Neuropathol Exp
Neurol. 2006;65(10):988–994.
42. Jenkins RB, Blair H, Ballman KV, et al. At (1;19)(q10;p10)
mediates the combined deletions of 1p and 19q and predicts
a better prognosis of patients with oligodendroglioma. Cancer
Res. 2006;66(20):9852–9861.
43. van den Bent MJ, Carpentier AF, Brandes AA, et al. Adjuvant
procarbazine, lomustine, and vincristine improves progression-
free survival but not overall survival in newly diagnosed
anaplastic oligodendrogliomas and oligoastrocytomas: a ran-
domized European Organisation for Research and Treat-
ment of Cancer phase III trial. J Clin Oncol. 2006;24(18):
2715–2722.
44. Taliansky-Aronov A, Bokstein F, Lavon I, Siegal T. Temozolo-
mide treatment for newly diagnosed anaplastic oligoden-
drogliomas: a clinical efficacy trial. J Neurooncol. 2006;79(2):
153–157.
45. Vogelbaum MA, Berkey B, Peereboom D, et al. Phase II trial
of preirradiation and concurrent temozolomide in patients

73724_CH01_Printer.indd 38 03/29/09 2:35:00 PM


Primary Brain Tumors 39

with newly diagnosed anaplastic oligodendrogliomas and


mixed anaplastic oligoastrocytomas: RTOG BR0131. Neuro
Oncol. 2009;11(2):167–175.
46. Mikkelsen T, Doyle T, Anderson J, et al. Temozolomide
single-agent chemotherapy for newly diagnosed anaplastic
oligodendroglioma. J Neurooncol. 2009;92(1):57–63.
47. Soffietti R, Ruda R, Bradac GB, Schiffer D. PCV chemo-
therapy for recurrent oligodendrogliomas and oligoastrocy-
tomas. Neurosurgery. 1998;43(5):1066–1073.
48. Cairncross G, Macdonald D, Ludwin S, et al. Chemotherapy
for anaplastic oligodendroglioma. National Cancer Institute
of Canada Clinical Trials Group. J Clin Oncol. 1994;12(10):
2013–2021.
49. Chinot OL, Honore S, Dufour H, et al. Safety and efficacy
of temozolomide in patients with recurrent anaplastic oligo-
dendrogliomas after standard radiotherapy and chemother-
apy. J Clin Oncol. 2001;19(9):2449–2455.
50. van den Bent MJ, Chinot O, Boogerd W, et al. Second-line
chemotherapy with temozolomide in recurrent oligodendro-
glioma after PCV (procarbazine, lomustine and vincristine)
chemotherapy: EORTC Brain Tumor Group phase II study
26972. Ann Oncol. 2003;14(4):599–602.
51. van den Bent MJ, Taphoorn MJ, Brandes AA, et al. Phase II
study of first-line chemotherapy with temozolomide in recur-
rent oligodendroglial tumors: the European Organization for
Research and Treatment of Cancer Brain Tumor Group
Study 26971. J Clin Oncol. 2003;21(13):2525–2528.
52. Marosi C, Hassler M, Roessler K, et al. Meningioma. Crit
Rev Oncol Hematol. 2008;67(2):153–171.
53. Asthagiri AR, Helm GA, Sheehan JP. Current concepts in
management of meningiomas and schwannomas. Neurol
Clin. 2007;25(4):1209–1230, xi.
54. Huisman TW, Tanghe HL, Koper JW, et al. Progesterone,
oestradiol, somatostatin and epidermal growth factor recep-
tors on human meningiomas and their CT characteristics.
Eur J Cancer. 1991;27(11):1453–1457.
55. Sioka C, Kyritsis AP. Chemotherapy, hormonal therapy, and
immunotherapy for recurrent meningiomas. J Neurooncol.
2009;92(1):1–6.
56. Sankila R, Kallio M, Jaaskelainen J, Hakulinen T. Long-term
survival of 1986 patients with intracranial meningioma

73724_CH01_Printer.indd 39 03/29/09 2:35:00 PM


40 Chapter 1

diagnosed from 1953 to 1984 in Finland. Comparison of the


observed and expected survival rates in a population-based
series. Cancer. 1992;70(6):1568–1576.
57. Nakamura M, Roser F, Michel J, Jacobs C, Samii M.
Volumetric analysis of the growth rate of incompletely
resected intracranial meningiomas. Zentralbl Neurochir.
2005;66(1):17–23.
58. Cote TR, Manns A, Hardy CR, Yellin FJ, Hartge P.
Epidemiology of brain lymphoma among people with or with-
out acquired immunodeficiency syndrome. AIDS/Cancer
Study Group. J Natl Cancer Inst. 1996;88(10):675–679.
59. Miller DC, Hochberg FH, Harris NL, Gruber ML,
Louis DN, Cohen H. Pathology with clinical correlations
of primary central nervous system non-Hodgkin’s lymphoma.
The Massachusetts General Hospital experience
1958–1989. Cancer. 1994;74(4):1383–1397.
60. Batchelor T, Loeffler JS. Primary CNS lymphoma. J Clin
Oncol. 2006;24(8):1281–1288.
61. Abrey LE, Batchelor TT, Ferreri AJ, et al. Report of an inter-
national workshop to standardize baseline evaluation and
response criteria for primary CNS lymphoma. J Clin Oncol.
2005;23(22):5034–5043.
62. Morris PG, Abrey LE. Therapeutic challenges in primary
CNS lymphoma. Lancet Neurol. 2009;8(6):581–592.
63. Lippens RJ, Winograd B. Methotrexate concentration levels
in the cerebrospinal fluid during high-dose methotrexate
infusions: an unreliable prediction. Pediatr Hematol Oncol.
1988;5(2):115–124.
64. Ferreri AJ, Reni M, Martelli M, et al. Randomized phase II
trial on primary chemotherapy with high-dose methotrexate
(HD-MTX) alone or associated with high-dose cytarabine
(HD-araC) for patients with primary CNS lymphoma
(I.E.L.S.G. #20 Trial): Tolerability, activity, and survival
analyses. J Clin Oncol (Meeting Abstracts). 2009;27(Suppl):
abstr 8545.
65. Nelson DF, Martz KL, Bonner H, et al. Non-Hodgkin’s lym-
phoma of the brain: can high dose, large volume radiation
therapy improve survival? Report on a prospective trial by
the Radiation Therapy Oncology Group (RTOG): RTOG
8315. Int J Radiat Oncol Biol Phys. 1992;23(1):9–17.
66. Shah GD, Yahalom J, Correa DD, et al. Combined immuno-
chemotherapy with reduced whole-brain radiotherapy for
newly diagnosed primary CNS lymphoma. J Clin Oncol.
2007;25(30):4730–4735.

73724_CH01_Printer.indd 40 03/29/09 2:35:00 PM


Primary Brain Tumors 41

67. Ferreri AJ, Blay JY, Reni M, et al. Prognostic scoring system
for primary CNS lymphomas: the International Extranodal
Lymphoma Study Group experience. J Clin Oncol. 2003;
21(2):266–272.
68. Marti-Carvajal AJ, Cardona AF, Lawrence A. Interventions
for previously untreated patients with AIDS-associated non-
Hodgkin’s lymphoma. Cochrane Database Syst Rev. 2009(3):
CD005419.

73724_CH01_Printer.indd 41 03/29/09 2:35:00 PM


73724_CH01_Printer.indd 42 03/29/09 2:35:00 PM
C H A P T E R 2

Brain Metastases

Epidemiology
■ Brain metastases are the most common intracranial
neoplasm in adults.1
■ The exact incidence is unknown, but it is estimated to be
as high as 200,000 cases per year in the United States.2
■ The incidence of CNS metastases appears to be rising
because of more effective systemic treatments resulting
in longer survival, earlier detection, and better imaging
modalities.
■ Most brain metastases originate from the following pri-
mary malignancies.1
• Lung cancer (40% to 50%)
• Breast cancer (15% to 25%)
• Melanoma (5% to 20%)
■ Most brain metastases occur through hematogenous and
spread with a predilection for vascular border zones and
gray-white matter junction.
■ The distribution parallels blood flow: 80% in cerebral
hemispheres, 15% in cerebellum, and 5% in brainstem.3

Clinical Features in Parenchymal Metastases


■ Headache
■ Neurologic deficits depending on location of lesion
(i.e., ataxia with cerebellar lesions, motor deficits with
corticospinal tract involvement)
■ Cognitive and mental status changes
■ Seizures

43

73724_CH02_Printer.indd 43 03/29/09 2:40:00 PM


44 Chapter 2

Diagnosis
■ Brain MRI with gadolinium is the preferred imaging
modality for evaluation of brain metastases.
• Lesions typically are well circumscribed and enhance
with gadolinium because of an impaired blood–brain
barrier.
• A brain MRI should not be used in emergency situa-
tions if the patient is too unstable.
• Posterior fossa and leptomeninges are better visual-
ized on MRI than CT.
■ In an acute presentation, head CT without contrast is
appropriate to identify life-threatening pathology, includ-
ing intracranial hemorrhage, acute hydrocephalus, or
herniation.
■ Brain biopsy is indicated if the diagnosis is in question.
• Multiple, widespread brain metastases in a patient
with cancer are strongly suggestive of brain metasta-
ses, but brain abscesses can have a similar appear-
ance, especially in immunosuppressed or septic
patients.
• Single or solitary brain lesions may be more difficult to
diagnose.
■ In one study, 11% of patients with cancer undergo-

ing surgery for a single brain lesion had pathology


consistent with a primary brain tumor, an infec-
tious disease, or an inflammatory disease.4

Prognosis
■ Important prognostic factors include age, Karnofsky per-
formance status (KPS) (Table 2-1), number of brain
metastases (single or multiple), primary tumor type, sys-
temic tumor activity (controlled or uncontrolled), and
time since primary tumor diagnosis.5,6

73724_CH02_Printer.indd 44 03/29/09 2:40:00 PM


Brain Metastases 45

Table 2-1: Karnofsky Performance Status Scale

Percentage Description

100 Normal, no complaints, no evidence of disease

90 Able to carry on normal activity; minor signs or


symptoms of disease

80 Normal activity with effort; some signs or symptoms


of disease

70 Cares for self, unable to carry on normal activity or


to do active work

60 Requires occasional assistance but is able to care for


most of his or her needs

50 Requires considerable assistance and frequent


medical care

40 Disabled, requires special care and assistance

30 Severely disabled, hospitalization indicated; death


not imminent

20 Very sick, hospitalization indicated; death not


imminent

10 Moribund, fatal processes progressing rapidly

0 Dead

■ Recursive partitioning analysis (RPA) classes (Table 2-2)5


• Patients can be divided into subgroups based on
prognosis.
• Subgroups are based on a review of 1,200 patients in
the Radiation Therapy Oncology Group (RTOG) data-
base who received whole-brain radiation therapy.

73724_CH02_Printer.indd 45 03/29/09 2:40:00 PM


46 Chapter 2

Table 2-2: Recursive Partitioning Analysis (RPA) Classes in


Patients with Brain Metastases

Class I Class II Class III

KPS $70 ,70

Age ,65 Any patient Any


not in class I
Extent of disease Controlled or class III Any
primary tumor,
no extracranial
metastases

Median overall 7.1 months 4.2 months 2.3 months


survival

Modified from Gaspar L, Scott C, Rotman M, et al. Recursive


partitioning analysis (RPA) of prognostic factors in three Radiation
Therapy Oncology Group (RTOG) brain metastases trials. Int J Radiat
Oncol Biol Phys. 1997;37(4):745–751.

Treatment
■ The treatment approach often depends on tumor histol-
ogy, number of brain metastases, the control of systemic
disease, and the status of neurologic function.
■ Main treatment options include whole-brain radiation,
surgical resection, radiosurgery, and systemic treatments.
■ Fifty percent of patients with CNS metastasis have a
single brain metastasis.

Whole-Brain Radiation Therapy (WBRT)


■ This is the mainstay of treatment for patients with brain

metastases.
■ Indications for WBRT are as follows:

• Curative intent
• Palliation
• Consolidation to reduce neurological morbidity
• Prophylaxis in specific patients with SCLC, NSCLC,
or breast cancer with curative intent

73724_CH02_Printer.indd 46 03/29/09 2:40:00 PM


Brain Metastases 47

■ Nonrandomized studies suggest that WBRT increases


median survival by 3 to 4 months over no treatment (the
median survival is approximately 1 month with no
treatment).1
■ Radiographic response rate (complete responses or par-
tial responses) to WBRT is approximately 60% in RTOG
randomized controlled trials.7
■ Symptom stabilization or improvement is achieved in
approximately 60% of patients, although this may be
overestimated because corticosteroids are often adminis-
tered with WBRT.8
■ Neurologic complications from radiation therapy are fur-
ther discussed in Chapter 5.

WBRT Treatment Regimens


■ The most common regimen used is 35 Gy delivered in
2.5 Gy fractions over 14 treatment days.
■ Fractions of .3 Gy increase the risk of neurotoxicity.

■ Differences in dose, timing, and fractionation do not alter

the median survival of patients receiving WBRT for brain


metastases.9

WBRT 1 Radiation Sensitizers


■ Most radiosensitizers (lonidamine, metronidazole, mis-

onidazole, gadolinium, or bromodeoxyuridine) have dem-


onstrated no benefit over WBRT alone in terms of local
brain tumor control or overall survival.
■ In a randomized phase III study of WBRT with or without

motexafin gadolinium for treatment of brain metastases


from solid tumors, there were no significant differences
between the two arms in terms of survival or time to neu-
rologic progression.10
• In the follow-up trial in patients with NSCLC, the
motexafin arm exhibited a nonsignificant trend toward
improved neurologic outcomes (time to neurologic
progression 10.0 months for WBRT vs. 15.4 months
for WBRT 1 motexafin, P 5 0.122).11
■ In a randomized phase III study of WBRT and supplemental

oxygen with or without efaproxiral for treatment of brain

73724_CH02_Printer.indd 47 03/29/09 2:40:00 PM


48 Chapter 2

metastases from solid tumors, there was no significant


difference between the two arms for survival.12
• In subgroup analysis of patients with breast cancer,
the efaproxiral arm had significantly reduced death
rates by 46% and improved quality of life.13
• Preliminary results from the follow-up phase III open-
label trial in patients with breast cancer, however, sug-
gest no difference in overall survival, response rate,
performance status, or neurologic signs/symptoms.14

WBRT 1 Chemotherapy
■ Several chemotherapeutic agents yield higher response

rates at the expense of greater toxicity and no benefit in


overall survival, including chloroethylnitrosoureas, tega-
fur, fotemustine, and teniposide.
■ Efficacy and safety of temozolomide and radiation in

brain metastases have been evaluated in several phase II


trials, but the combination does not seem to improve
survival.
• Antonadou et al. reported significant improvement in
the response rate with temozolomide 1 WBRT com-
pared with WBRT alone (96% vs. 67%) but survival
data were not evaluated.15
• In a randomized phase II trial of temozolomide 1 WBRT
versus WBRT alone, overall survival and response rates
were similar in both arms, but the percentage of patients
with progression-free survival of brain metastases at
90 days was significantly higher for the temozolomide 1
WBRT arm (72% vs. 54%).16

Stereotactic Radiosurgery (SRS)


■ SRS uses multiple convergent beams to deliver a single

high dose of focal radiation to a small target volume.


■ This is a common treatment modality for newly diagnosed

brain metastases, alone or in combination with WBRT,


and as salvage therapy after WBRT.
■ The most common delivery systems include linear accel-

erator, gamma knife, and cyclotron.

73724_CH02_Printer.indd 48 03/29/09 2:40:00 PM


Brain Metastases 49

■ Maximum tolerated doses are based on the RTOG 90-05


protocol.17
• Tumors 31 to 40 mm: 15 Gy
• Tumors 21 to 30 mm: 18 Gy
• Tumors less than or equal to 20 mm: 24 Gy but subse-
quent retrospective analysis suggested that doses
.20 Gy increase neurotoxicity and do not improve
local control.18
■ One-year actuarial local control rates for single and
multiple brain metastases with SRS alone are 71%
to 79%.18–21
■ Based on a small series, SRS at recurrence is an accept-
able treatment for patients with good functional status
and controlled/indolent extracranial disease with local
control rates of 57% to 100% and overall brain control
rates of 65% to 78%.17,22–25
■ SRS does not prevent distant failure.
■ The complications from SRS are the following:
• Early treatment-induced cerebral edema (4% to 6%
patients within 1 to 2 weeks of treatment)
• Seizures (2% to 6% patients within the first 24 to
48 hours)
• Delayed radiation necrosis (2% to 17%)
■ There is a higher risk for radiation necrosis with

larger tumor volume, higher radiation dose, and prior


radiotherapy.

WBRT with or without SRS


■ Based on three randomized controlled trials and two

retrospective studies, SRS boost after WBRT improves


survival in select patients with single brain metastases.1
■ In patients with two to three brain metastases, the role of

SRS boost is less clear.


• Based on subgroup analysis of a large randomized con-
trolled trial, SRS boost after WBRT did not improve
survival or local control.26
• Based on a smaller randomized study 27 and retrospec-
tive series,28 SRS boost did improve survival.

73724_CH02_Printer.indd 49 03/29/09 2:40:00 PM


50 Chapter 2

SRS with or without WBRT


■ It is controversial whether patients can be treated with
SRS alone in order to spare the risk for late neurotoxicity
from WBRT.
■ Based on a randomized controlled trial, SRS alone as

compared with WBRT 1 SRS results in worse local con-


trol and worse distant intracranial disease control with no
difference in overall survival.19
■ Preliminary results from the SRS arm of the European

Organization for Research and Treatment of Cancer


(EORTC) study 22952-26001 (patients with good per-
formance status and 1–3 brain metastases with stable
systemic cancer or asymptomatic synchronous primary
tumor without metastases outside the CNS)29:
• No difference in overall survival between SRS alone
versus SRS 1 WBRT
• Significant reduction of intracranial progression after
WBRT in both arms of the study
■ There is a risk of distant brain failure in patients who

receive SRS alone.30


• The low-risk (median time to distant brain failure
47.9 weeks) group had three or fewer metastases, an
absence of extracranial disease, and nonmelanoma
histologic characteristics.
• The median time to distant brain failure for all other
possible groupings ranged from 12.3 to 28.7 weeks.

Surgery
■ The benefits of surgery include pathology for definitive

diagnosis, rapid relief of neurologic symptoms caused by


mass effect, and local control.
■ Resection of single brain metastasis is a standard option

in patients with good, functional status and controlled or


indolent extracranial disease.31
■ The in-hospital mortality for resection of brain metasta-

ses in high-volume centers is as low as 1.8%.31

73724_CH02_Printer.indd 50 03/29/09 2:40:00 PM


Brain Metastases 51

Surgery for a Single Brain Metastasis


■ Three randomized controlled trials have compared surgical
resection 1 WBRT versus WBRT alone in patients with a
single brain metastasis (Table 2-3).4,32,33
• Patchell et al. 1990: resection 1 WBRT versus biopsy
1 WBRT associated with longer median survival time,
better local control rate, longer duration of functional
independence (KPS $ 70) and longer freedom from
death due to neurologic causes.
• Vetch et al. 1993: resection 1 WBRT associated with
longer median survival time as compared with WBRT
alone.
• Mintz et al. 1996: no survival difference between
two groups but patients had a poorer functional status
and more active extracranial disease than other studies.

Table 2-3: Results from Randomized Controlled Trials


of Patients with a Single Brain Metastasis
Comparing Surgical Resection ⴙ WBRT
Versus WBRT Alone

Resection ⴙ
WBRT Median WBRT Median
Study Overall Survival Overall Survival

Patchell et al., 1990 9.2 months 3.4 months

Vecht et al., 1993 10.0 months 6.0 months

Mintz et al., 1996 5.6 months 6.3 months

Data are from Patchell RA, Tibbs PA, Walsh JW, et al. A randomized trial
of surgery in the treatment of single metastases to the brain. N Engl
J Med. 1990;322(8):494–500. Vecht CJ, Haaxma-Reiche H, Noordijk EM,
et al. Treatment of single brain metastasis: radiotherapy alone or
combined with neurosurgery? Ann Neurol. 1993;33(6):583–590.
Mintz AH, Kestle J, Rathbone MP, et al. A randomized trial to assess the
efficacy of surgery in addition to radiotherapy in patients with a single
cerebral metastasis. Cancer. 1996;78(7):1470–1476.

73724_CH02_Printer.indd 51 03/29/09 2:40:00 PM


52 Chapter 2

Surgery for Multiple Brain Metastases


■ Patients with good prognostic features and two to three
metastases may benefit from resection of the dominant
lesion.34,35
■ A highly selected subset of patients with multiple metas-

tases may benefit from resection of all lesions.36

Surgery with or without WBRT


■ Based on a randomized controlled trial, resection of a

single brain metastasis 1 WBRT versus resection alone


is associated with better local, distant, and overall intrac-
ranial control rates.37
■ No benefit is seen in overall survival from combined ther-

apy, although the patients who received WBRT were less


likely to die from neurologic causes.
■ Preliminary results from the surgery arm of EORTC

22952-26001 (patients with one to three brain metasta-


ses with stable systemic cancer or asymptomatic synchro-
nous primary tumor without metastases outside of the
CNS)29:
• No difference in overall survival between surgery alone
versus surgery 1 WBRT
• Significant reduction of intracranial progression with
WBRT

Surgery Versus SRS


■ There are no prospective studies directly comparing sur-

gery versus SRS.


■ Most retrospective studies demonstrate no difference in

survival between surgery and SRS.38


■ Surgery is probably better for larger tumors with exten-

sive edema and mass effect or tumors requiring rapid


relief of neurologic symptoms.
■ SRS is noninvasive and can treat tumors that are not

amenable to surgery.

Chemotherapy
■ This is traditionally reserved for patients who have
failed other treatment modalities or tumors that are

73724_CH02_Printer.indd 52 03/29/09 2:40:00 PM


Brain Metastases 53

chemosensitive (lymphoma, SCLC, germ cell tumors


more chemosensitive than breast cancer).
■ Because brain metastases represent a late event in the
natural history of most cancers, tumors may have acquired
resistance to many drugs.
■ For patients with active systemic disease and brain metas-
tases, chemotherapy provides an opportunity to treat both
compartments.39
■ Delivery to the CNS is limited by the blood–brain barrier,
efflux by P-glycoprotein, nonuniform drug distribution in
the tumor (preferential concentration in necrotic areas),
and poor drug accumulation (due to tumor interstitial
fluid gradients).40
■ This is discussed further later in this chapter.

Brain Metastases from Breast Cancer


Epidemiology
■ Breast cancer is the second most frequent cause of brain
metastasis.41
■ Up to 30% patients with breast cancer will develop CNS

metastases.42–44
■ Breast cancer is the solid tumor most commonly associ-

ated with leptomeningeal metastases.44


■ Leptomeningeal involvement is found at autopsy in 5% to

16% of breast cancer patients.44


■ In the majority of breast cancer patients, metastases to

the CNS develop following extracranial metastases,


although the CNS may be the first site of recurrence in
20% to 39% of patients.45

Incidence in Patients Receiving Trastuzumab


■ Trastuzumab is a monoclonal antibody against the HER2/

neu oncoprotein used to treat HER2/neu-positive breast


cancer.
■ Some studies report a higher incidence of CNS metasta-

ses in patients who received trastuzumab-based regimens


for HER2 overexpressing metastatic breast cancer,46–49
whereas other studies did not find a higher incidence.

73724_CH02_Printer.indd 53 03/29/09 2:40:00 PM


54 Chapter 2

■ The proposed causes for possible higher incidence are as


follows:
• Trastuzumab does not cross the blood–brain barrier,
and therefore, the CNS may act as a sanctuary site for
metastatic disease.
• There is prolonged survival in HER2-positive meta-
static breast cancer due to trastuzumab, resulting in
an increased risk for CNS relapse.
• There is an inherent biology of HER2-positive breast
cancer (which may be associated with higher risk for
CNS metastases).
■ Continued use of trastuzumab in patients with brain
metastases may improve overall survival compared with
discontinuation of trastuzumab, possibly because of bet-
ter control of systemic disease.50

Risk Factors for the Development of CNS Metastases


in Breast Cancer44,45,51
■ Young age

■ Negative hormone receptor status

■ Invasive ductal carcinoma

Treatment Options for Brain Metastases


from Breast Cancer
■ Treatment recommendations are similar to brain metas-

tases from other primary tumors.

Surgery
■ This is indicated for urgent decompression and solitary
brain metastasis.
■ Surgery is typically reserved for patients with a single

brain lesion, minimal extracranial disease, and/or better


performance status.
■ Surgical resection for single brain metastases followed by

WBRT is superior to WBRT alone with stable/absent


extracranial disease.4

SRS
■ SRS is typically recommended for #3 brain metastases,

with each lesion measuring #3 cm, but the failure rate


outside the SRS field is 37%.

73724_CH02_Printer.indd 54 03/29/09 2:40:00 PM


Brain Metastases 55

■ There are no randomized trials comparing SRS with


surgery for treatment of single brain metastasis.
■ Combining SRS 1 WBRT is better than WBRT alone.44

WBRT
■ Seventy-five percent to 85% of patients who receive

WBRT experience improvement or stabilization of


symptoms (seizures and headaches most effectively
palliated).44
■ Postoperative WBRT does not extend survival but does

reduce brain recurrences.37

Systemic Treatments
■ This is generally recommended after failure with surgery

and radiation.
■ Selected chemotherapy agents such as capecitabine,

5-fluorouracil, platinum analogues, temozolomide, meth-


otrexate, topotecan, bendamustine have known activity
in the CNS.40,51
■ Lapatinib has modest antitumor activity against CNS

metastases in HER2-positive breast cancer patients.52


■ Capecitabine in combination with temozolomide or lapa-

tinib may have activity in new and recurrent brain metas-


tases, but larger studies are needed.50

Brain Metastases from Non-Small Cell Lung


Cancer (NSCLC)
Epidemiology
■ Brain metastases may develop in 20% to 40% of patients

with NSCLC.53
■ NSCLC is usually chemoresistant, and thus, patients

who develop brain metastases usually have been heavily


pretreated.
■ RPA classification scheme (Table 2-2) was based on a

study in which lung cancer accounted for 61% of the


patients studied (almost all had NSCLC).5
■ Although survival times are short for patients with brain

metastases, patients with brain metastases from NSCLC


may have a worse prognosis than patients with brain
metastases from other solid tumors.54

73724_CH02_Printer.indd 55 03/29/09 2:40:00 PM


56 Chapter 2

Risk Factors for Development of CNS Metastases


in NSCLC
■ Age5

■ Clinical stage (later stage)55

■ Adenocarcinoma histology56

■ CEA serum levels $40 ng/mL56

Treatment Options for Brain Metastases from NSCLC


■ Treatment decisions in NSCLC are often based on the

extent of disease characterized as follows:


• Single brain metastasis with controlled or controllable
primary disease
• Oligometastatic disease (primary disease and less than
three distant metastases)
• Multiple metastases
■ Carefully selected patients with resectable N0,1 primary

NSCLC and an isolated brain metastasis as the only site


of metastatic disease may benefit from aggressive therapy
of the primary site and brain lesion (surgery or SRS) to
prolong survival.57
■ In patients with a single brain metastasis and previously

resected primary NSCLC, surgical resection or SRS of


the isolated brain metastasis should be considered.57
■ Select patients with oligometastatic disease may be

treated with surgical resection or radiosurgery followed


by WBRT.58
■ Patients with multiple brain metastases from NSCLC

are generally treated with WBRT.

Systemic Treatments
■ For patients with brain metastases, chemotherapy and
targeted therapies have a greater role for patients with
active systemic disease or contraindications to other
treatment modalities.53
■ For patients with brain metastases but controlled or

stable systemic disease, surgery and radiation are more


commonly used.
■ Chemotherapy-naïve patients with brain metastases may

benefit from cisplatin alone (radiographic response rates

73724_CH02_Printer.indd 56 03/29/09 2:40:00 PM


Brain Metastases 57

30%)59 or in combination with other agents (radiographic


response rates 28% to 45%).60–64
■ Patients with recurrent brain metastases may benefit
from TMZ alone (RR 0% to 20%)65–68 or in combination
with vinorelbine (higher RR but more toxicity).69
■ Data on the response of brain metastases to small
molecular epidural growth factor receptor (EGFR) inhib-
itors (erlotinib, gefitinib) is limited, but responses to
these agents likely depend on the presence of EGFR
mutations.70

Brain Metastases from Melanoma


Epidemiology
■ CNS metastases detectable on imaging develop in nearly

50% of patients with metastatic melanoma.71


■ Patients with metastatic melanoma to the brain have a

medial survival of 4 to 6 months.72


■ Brain metastases from melanoma tend to be hemorrhagic.

Imaging
■ The typical melanotic pattern on MRI consists of high

signal intensity on T1-weighted images and low signal


intensity on T2-weighted images.
• The MRI appearance is attributed to free radicals in
melanin as well as blood products.
■ An amelanotic pattern is also frequently described

consisting of a hypointense/isointense appearance on


T1-weighted images and hyperintense/isointense appear-
ance on T2-weighted images.
■ Dural involvement by malignant melanoma is rare.

Treatment Options for Brain Metastases from


Melanoma
■ Whole brain radiation therapy is moderately effective for

the control of melanoma metastatic to the brain (a com-


plete response rate of less than 20% and a partial response
rate of approximately 50%).72

73724_CH02_Printer.indd 57 03/29/09 2:40:00 PM


58 Chapter 2

■ Select patients with limited disease may benefit from


stereotactic surgery for local control.
■ Chemotherapy does not significantly increase the local
control rate.
• Fotemustine (response rate 5% to 25%) and temozolo-
mide (response rate of 6% to 10%) are the most active
agents.38
■ A select subset of patients achieve disease stabilization
with aggressive local control 1 WBRT.1

Brain Metastases from Renal Cell


Carcinoma (RCC)
Epidemiology
■ Only 4% to 11% of patients with RCC develop brain
metastases.73
■ Over 90% of patients with brain metastases from RCC

are symptomatic (headaches, mental status changes,


confusion, and seizures).74
■ Development of brain metastases suggests late-stage dis-

ease, and patients frequently have extracranial disease.


■ Brain metastases from RCC tend to be hemorrhagic.

Treatment Options for Brain Metastases from RCC


■ A select subset of patients, particularly those with one to

two brain metastases and an absence of extracranial dis-


ease, can achieve long-term survival with surgical resec-
tion 1 radiation.75
■ Brain metastases from RCC are considered “radioresis-

tant,” although in a published case series of patients with


up to five lesions, SRS with or without WBRT has
produced good local control and prolonged survival.76
Prospective studies are needed to confirm these results
and to characterize better which patients would benefit
most from SRS treatment.
■ The role of vascular endothelial growth factor (VEGF)

and VEGF receptor inhibitors in brain metastases from


RCC is unclear because most published phase II and
phase III clinical trials of systemic disease excluded
patients with brain metastases.

73724_CH02_Printer.indd 58 03/29/09 2:40:00 PM


Brain Metastases 59

References
1. Eichler AF, Loeffler JS. Multidisciplinary management of
brain metastases. Oncologist. 2007;12(7):884–898.
2. Gavrilovic IT, Posner JB. Brain metastases: epidemiology
and pathophysiology. J Neurooncol. 2005;75(1):5–14.
3. Delattre JY, Krol G, Thaler HT, Posner JB. Distribution of
brain metastases. Arch Neurol. 1988;45(7):741–744.
4. Patchell RA, Tibbs PA, Walsh JW, et al. A randomized trial
of surgery in the treatment of single metastases to the brain.
N Engl J Med. 1990;322(8):494–500.
5. Gaspar L, Scott C, Rotman M, et al. Recursive partitioning
analysis (RPA) of prognostic factors in three Radiation
Therapy Oncology Group (RTOG) brain metastases trials.
Int J Radiat Oncol Biol Phys. 1997;37(4):745–751.
6. Lagerwaard FJ, Levendag PC, Nowak PJ, Eijkenboom WM,
Hanssens PE, Schmitz PI. Identification of prognostic fac-
tors in patients with brain metastases: a review of 1292
patients. Int J Radiat Oncol Biol Phys. 1999;43(4):795–803.
7. Khuntia D, Brown P, Li J, Mehta MP. Whole-brain radio-
therapy in the management of brain metastasis. J Clin
Oncol. 2006;24(8):1295–1304.
8. Bezjak A, Adam J, Barton R, et al. Symptom response after
palliative radiotherapy for patients with brain metastases.
Eur J Cancer. 2002;38(4):487–496.
9. Tsao MN, Lloyd N, Wong R, Chow E, Rakovitch E,
Laperriere N. Whole brain radiotherapy for the treatment of
multiple brain metastases. Cochrane Database Syst Rev.
2006;3:CD003869.
10. Mehta MP, Rodrigus P, Terhaard CH, et al. Survival and
neurologic outcomes in a randomized trial of motexafin gad-
olinium and whole-brain radiation therapy in brain metasta-
ses. J Clin Oncol. 2003;21(13):2529–2536.
11. Mehta MP, Shapiro WR, Phan SC, et al. Motexafin gado-
linium combined with prompt whole brain radiotherapy pro-
longs time to neurologic progression in non-small-cell lung
cancer patients with brain metastases: results of a phase III
trial. Int J Radiat Oncol Biol Phys. 2009;73(4):1069–1076.
12. Suh JH, Stea B, Nabid A, et al. Phase III study of efaproxiral
as an adjunct to whole-brain radiation therapy for brain
metastases. J Clin Oncol. 2006;24(1):106–114.
13. Scott C, Suh J, Stea B, Nabid A, Hackman J. Improved sur-
vival, quality of life, and quality-adjusted survival in breast
cancer patients treated with efaproxiral (Efaproxyn) plus

73724_CH02_Printer.indd 59 03/29/09 2:40:00 PM


60 Chapter 2

whole-brain radiation therapy for brain metastases. Am J


Clin Oncol. 2007;30(6):580–587.
14. Suh JH, Stea B, Tankel K, et al. Results of the phase III
ENRICH (RT-016) study of efaproxiral administered con-
current with whole brain radiation therapy (WBRT) in
women with brain metastases from breast cancer. Int J
Radiat Oncol Biol Phys. 2008;72(Suppl 1):S50–S51.
15. Antonadou D, Paraskevaidis M, Sarris G, et al. Phase II ran-
domized trial of temozolomide and concurrent radiotherapy
in patients with brain metastases. J Clin Oncol. 2002;20(17):
3644–3650.
16. Verger E, Gil M, Yaya R, et al. Temozolomide and concomi-
tant whole brain radiotherapy in patients with brain metas-
tases: a phase II randomized trial. Int J Radiat Oncol Biol
Phys. 2005;61(1):185–191.
17. Shaw E, Scott C, Souhami L, et al. Single dose radiosurgical
treatment of recurrent previously irradiated primary brain
tumors and brain metastases: final report of RTOG protocol
90-05. Int J Radiat Oncol Biol Phys. 2000;47(2):291–298.
18. Shehata MK, Young B, Reid B, et al. Stereotatic radiosurgery
of 468 brain metastases , or 5 2 cm: implications for SRS
dose and whole brain radiation therapy. Int J Radiat Oncol
Biol Phys. 2004;59(1):87–93.
19. Aoyama H, Shirato H, Tago M, et al. Stereotactic radiosur-
gery plus whole-brain radiation therapy vs stereotactic
radiosurgery alone for treatment of brain metastases: a ran-
domized controlled trial. JAMA. 2006;295(21):2483–2491.
20. Hasegawa T, Kondziolka D, Flickinger JC, Germanwala A,
Lunsford LD. Brain metastases treated with radiosurgery
alone: an alternative to whole brain radiotherapy?
Neurosurgery. 2003;52(6):1318–1326; discussion 1326.
21. Sneed PK, Lamborn KR, Forstner JM, et al. Radiosurgery
for brain metastases: is whole brain radiotherapy necessary?
Int J Radiat Oncol Biol Phys. 1999;43(3):549–558.
22. Flickinger JC, Kondziolka D, Lunsford LD, et al. A multi-
institutional experience with stereotactic radiosurgery for
solitary brain metastasis. Int J Radiat Oncol Biol Phys. 1994;
28(4):797–802.
23. Fuller BG, Kaplan ID, Adler J, Cox RS, Bagshaw MA.
Stereotaxic radiosurgery for brain metastases: the impor-
tance of adjuvant whole brain irradiation. Int J Radiat Oncol
Biol Phys. 1992;23(2):413–418.

73724_CH02_Printer.indd 60 03/29/09 2:40:00 PM


Brain Metastases 61

24. Loeffler JS, Kooy HM, Wen PY, et al. The treatment of
recurrent brain metastases with stereotactic radiosurgery.
J Clin Oncol. 1990;8(4):576–582.
25. Noel G, Proudhom MA, Valery CA, et al. Radiosurgery for
re-irradiation of brain metastasis: results in 54 patients.
Radiother Oncol. 2001;60(1):61–67.
26. Andrews DW, Scott CB, Sperduto PW, et al. Whole brain
radiation therapy with or without stereotactic radiosurgery
boost for patients with one to three brain metastases: phase III
results of the RTOG 9508 randomised trial. Lancet.
2004;363(9422):1665–1672.
27. Kondziolka D, Patel A, Lunsford LD, Kassam A, Flickinger JC.
Stereotactic radiosurgery plus whole brain radiotherapy versus
radiotherapy alone for patients with multiple brain metastases.
Int J Radiat Oncol Biol Phys. 1999;45(2):427–434.
28. Sanghavi SN, Miranpuri SS, Chappell R, et al. Radiosurgery
for patients with brain metastases: a multi-institutional
analysis, stratified by the RTOG recursive partitioning anal-
ysis method. Int J Radiat Oncol Biol Phys. 2001;51(2):
426–434.
29. Mueller RP, Soffietti R, Abacioglu MU, et al. Adjuvant
whole-brain radiotherapy versus observation after radiosur-
gery or surgical resection of 1–3 cerebral metastases: Results
of the EORTC 22952-26001 study. J Clin Oncol (Meeting
Abstracts). 2009;27(Suppl):abstr 2008.
30. Sawrie SM, Guthrie BL, Spencer SA, et al. Predictors
of distant brain recurrence for patients with newly
diagnosed brain metastases treated with stereotactic radio-
surgery alone. Int J Radiat Oncol Biol Phys. 2008;70(1):
181–186.
31. Barker FG, 2nd. Craniotomy for the resection of metastatic brain
tumors in the U.S., 1988–2000: decreasing mortality and the
effect of provider caseload. Cancer. 2004;100(5):999–1007.
32. Mintz AH, Kestle J, Rathbone MP, et al. A randomized trial
to assess the efficacy of surgery in addition to radiotherapy
in patients with a single cerebral metastasis. Cancer.
1996;78(7):1470–1476.
33. Vecht CJ, Haaxma-Reiche H, Noordijk EM, et al. Treatment
of single brain metastasis: radiotherapy alone or combined
with neurosurgery? Ann Neurol. 1993;33(6):583–590.
34. Paek SH, Audu PB, Sperling MR, Cho J, Andrews DW.
Reevaluation of surgery for the treatment of brain metastases:

73724_CH02_Printer.indd 61 03/29/09 2:40:00 PM


62 Chapter 2

review of 208 patients with single or multiple brain metastases


treated at one institution with modern neurosurgical
techniques. Neurosurgery. 2005;56(5):1021–1034.
35. Stark AM, Tscheslog H, Buhl R, Held-Feindt J, Mehdorn HM.
Surgical treatment for brain metastases: prognostic factors and
survival in 177 patients. Neurosurg Rev. 2005;28(2):115–119.
36. Bindal RK, Sawaya R, Leavens ME, Lee JJ. Surgical treat-
ment of multiple brain metastases. J Neurosurg.
1993;79(2):210–216.
37. Patchell RA, Tibbs PA, Regine WF, et al. Postoperative
radiotherapy in the treatment of single metastases to the
brain: a randomized trial. JAMA. 1998;280(17):1485–1489.
38. Soffietti R, Ruda R, Trevisan E. Brain metastases: current
management and new developments. Curr Opin Oncol.
2008;20(6):676–684.
39. Nguyen TD, DeAngelis LM. Brain metastases. Neurol Clin.
2007;25(4):1173–1192, x–xi.
40. Tosoni A, Franceschi E, Brandes AA. Chemotherapy in
breast cancer patients with brain metastases: have new
chemotherapic agents changed the clinical outcome? Crit
Rev Oncol Hematol. 2008;68(3):212–221.
41. Lee YT. Breast carcinoma: pattern of metastasis at autopsy.
J Surg Oncol. 1983;23(3):175–180.
42. Boogerd W. Central nervous system metastasis in breast
cancer. Radiother Oncol. 1996;40(1):5–22.
43. Boogerd W, Vos VW, Hart AA, Baris G. Brain metastases in
breast cancer; natural history, prognostic factors and out-
come. J Neurooncol. 1993;15(2):165–174.
44. Lin NU, Bellon JR, Winer EP. CNS metastases in breast
cancer. J Clin Oncol. 2004;22(17):3608–3617.
45. Tham YL, Sexton K, Kramer R, Hilsenbeck S, Elledge R.
Primary breast cancer phenotypes associated with propen-
sity for central nervous system metastases. Cancer. 2006;
107(4):696–704.
46. Bendell JC, Domchek SM, Burstein HJ, et al. Central ner-
vous system metastases in women who receive trastuzumab-
based therapy for metastatic breast carcinoma. Cancer.
2003;97(12):2972–2977.
47. Clayton AJ, Danson S, Jolly S, et al. Incidence of cerebral
metastases in patients treated with trastuzumab for meta-
static breast cancer. Br J Cancer. 2004;91(4):639–643.
48. Shmueli E, Wigler N, Inbar M. Central nervous system pro-
gression among patients with metastatic breast cancer

73724_CH02_Printer.indd 62 03/29/09 2:40:00 PM


Brain Metastases 63

responding to trastuzumab treatment. Eur J Cancer. 2004;


40(3):379–382.
49. Stemmler HJ, Kahlert S, Siekiera W, Untch M, Heinrich B,
Heinemann V. Characteristics of patients with brain metas-
tases receiving trastuzumab for HER2 overexpressing meta-
static breast cancer. Breast. 2006;15(2):219–225.
50. Barrett-Lee P, Bidard FC, Pierga JY. Contemporary issues
and the potential uses of capecitabine in metastatic breast
cancer. Cancer Treat Rev. 2009;35(7):582–589.
51. Stemmler HJ, Heinemann V. Central nervous system metas-
tases in HER-2-overexpressing metastatic breast cancer:
a treatment challenge. Oncologist. 2008;13(7):739–750.
52. Lin NU, Dieras V, Paul D, et al. Multicenter phase II
study of lapatinib in patients with brain metastases from
HER2-positive breast cancer. Clin Cancer Res. 2009;
15(4):1452–1459.
53. Adamo V, Franchina T, Adamo B, Scandurra G, Scimone A.
Brain metastases in patients with non-small cell lung can-
cer: focus on the role of chemotherapy. Ann Oncol. 2006;
17(Suppl 2):ii73–ii75.
54. Barton R. Inoperable brain metastases from non-small cell
lung cancer: what part does whole brain radiotherapy play in
standard treatment? Thorax. 2008;63(1):1–2.
55. Robnett TJ, Machtay M, Stevenson JP, Algazy KM, Hahn SM.
Factors affecting the risk of brain metastases after definitive
chemoradiation for locally advanced non-small-cell lung carci-
noma. J Clin Oncol. 2001;19(5):1344–1349.
56. Arrieta O, Saavedra-Perez D, Kuri R, et al. Brain metastasis
development and poor survival associated with carcinoem-
bryonic antigen (CEA) level in advanced non-small cell lung
cancer: a prospective analysis. BMC Cancer. 2009;9:119.
57. Shen KR, Meyers BF, Larner JM, Jones DR. Special treat-
ment issues in lung cancer: ACCP evidence-based clinical
practice guidelines (2nd edition). Chest. 2007;132(3 Suppl):
290S–305S.
58. Kvale PA, Selecky PA, Prakash UB. Palliative care in lung
cancer: ACCP evidence-based clinical practice guidelines
(2nd edition). Chest. 2007;132(3 Suppl):368S–403S.
59. Kleisbauer JP, Guerin JC, Arnaud A, Poirier R, Vesco D.
[Chemotherapy with high-dose cisplatin in brain metastasis
of lung cancers]. Bull Cancer. 1990;77(7):661–665.
60. Bernardo G, Cuzzoni Q, Strada MR, et al. First-line chemo-
therapy with vinorelbine, gemcitabine, and carboplatin in the

73724_CH02_Printer.indd 63 03/29/09 2:40:00 PM


64 Chapter 2

treatment of brain metastases from non-small-cell lung


cancer: a phase II study. Cancer Invest. 2002;20(3):293–302.
61. Cortes J, Rodriguez J, Aramendia JM, et al. Front-line pacli-
taxel/cisplatin-based chemotherapy in brain metastases from
non-small-cell lung cancer. Oncology. 2003;64(1):28–35.
62. Cotto C, Berille J, Souquet PJ, et al. A phase II trial of fote-
mustine and cisplatin in central nervous system metastases
from non-small cell lung cancer. Eur J Cancer. 1996;32A(1):
69–71.
63. Franciosi V, Cocconi G, Michiara M, et al. Front-line che-
motherapy with cisplatin and etoposide for patients with
brain metastases from breast carcinoma, nonsmall cell lung
carcinoma, or malignant melanoma: a prospective study.
Cancer. 1999;85(7):1599–1605.
64. Minotti V, Crino L, Meacci ML, et al. Chemotherapy with
cisplatin and teniposide for cerebral metastases in non-small
cell lung cancer. Lung Cancer. 1998;20(2):93–98.
65. Abrey LE, Olson JD, Raizer JJ, et al. A phase II trial of temo-
zolomide for patients with recurrent or progressive brain
metastases. J Neurooncol. 2001;53(3):259–265.
66. Christodoulou C, Bafaloukos D, Kosmidis P, et al. Phase II
study of temozolomide in heavily pretreated cancer patients
with brain metastases. Ann Oncol. 2001;12(2):249–254.
67. Dziadziuszko R, Ardizzoni A, Postmus PE, et al. Temo-
zolomide in patients with advanced non-small cell lung can-
cer with and without brain metastases. a phase II study of
the EORTC Lung Cancer Group (08965). Eur J Cancer.
2003;39(9):1271–1276.
68. Giorgio CG, Giuffrida D, Pappalardo A, et al. Oral temozo-
lomide in heavily pre-treated brain metastases from non-
small cell lung cancer: phase II study. Lung Cancer. 2005;
50(2):247–254.
69. Omuro AM, Raizer JJ, Demopoulos A, Malkin MG, Abrey LE.
Vinorelbine combined with a protracted course of temozolo-
mide for recurrent brain metastases: a phase I trial.
J Neurooncol. 2006;78(3):277–280.
70. Shimato S, Mitsudomi T, Kosaka T, et al. EGFR mutations
in patients with brain metastases from lung cancer: associa-
tion with the efficacy of gefitinib. Neuro Oncol. 2006;8(2):
137–144.
71. Amer MH, Al-Sarraf M, Baker LH, Vaitkevicius VK.
Malignant melanoma and central nervous system metasta-
ses: incidence, diagnosis, treatment and survival. Cancer.
1978;42(2):660–668.

73724_CH02_Printer.indd 64 03/29/09 2:40:00 PM


Brain Metastases 65

72. McLoughlin JM, Zager JS, Sondak VK, Berk LB. Treatment
options for limited or symptomatic metastatic melanoma.
Cancer Control. 2008;15(3):239–247.
73. Muacevic A, Siebels M, Tonn JC, Wowra B. Treatment of
brain metastases in renal cell carcinoma: radiotherapy, radio-
surgery, or surgery? World J Urol. 2005;23(3):180–184.
74. Klatte T, Lam JS, Shuch B, Belldegrun AS, Pantuck AJ.
Surveillance for renal cell carcinoma: why and how? When
and how often? Urol Oncol. 2008;26(5):550–554.
75. Harada Y, Nonomura N, Kondo M, et al. Clinical study of
brain metastasis of renal cell carcinoma. Eur Urol. 1999;
36(3):230–235.
76. Samlowski WE, Majer M, Boucher KM, et al. Multi-
disciplinary treatment of brain metastases derived from clear
cell renal cancer incorporating stereotactic radiosurgery.
Cancer. 2008;113(9):2539–2548.

73724_CH02_Printer.indd 65 03/29/09 2:40:00 PM


73724_CH02_Printer.indd 66 03/29/09 2:40:00 PM
C H A P T E R 3

Spinal Tumors

Introduction
■ Spinal tumors are uncommon but can cause significant
neurologic morbidity.
■ Early diagnosis and adequate treatment are critical for
functional outcomes and long-term prognosis.

Spinal Cord Anatomy (Figure 3-1)


■ Bone: the spinal cord is housed within a canal formed by

vertebral bodies and neural arches.


• The vertebral body lies on the ventral (anterior) side of the
spinal cord.
• The neural arch lies on the dorsal (posterior) side of the
spinal cord.
• The spinal ner ves pass through an opening between
the vertebrae called the inter vertebral foramen.

Figure 3-1: Spinal cord anatomy

67

73724_CH03_Printer.indd 67 03/29/09 2:36:00 PM


68 Chapter 3

■ Meninges: the spinal cord is covered by three membranes


known together as the meninges.
• Dura mater
• Arachnoid mater
• Pia mater
■ Potential spaces between meningeal layers
• Epidural space: between the bone and the dura mater,
contains fat and vertebral veins
• Subdural space: between the dura mater and the
arachnoid mater
• Subarachnoid space: between the arachnoid mater
and the pia mater, contains cerebrospinal fluid
■ Spinal cord tumors are classified according to their ana-
tomical location.
• Extradural: between bone and the dura mater
• Intradural: inside the dura mater
■ Extramedullary: between the dura mater and the

pia mater
■ Intramedullary: within the spinal cord parenchyma

Extradural Tumors
■ The extradural space is the most common site for spinal
tumors, with 60% of all spinal cord neoplasms located in
the extradural space and another 10% spanning the extra-
dural and intradural spaces.1
• Most tumors in the extradural space are metastatic, such
as epidural metastases (discussed later in this chapter).
• Some extradural tumors may arise in the vertebra,
such as osteosarcomas, hemangiomas, lymphoma, and
plasmacytoma.
■ Some extradural tumors may undermine the structural
stability of the vertebrae, leading to painful vertebral
fractures.

Metastatic Epidural Spinal Cord Compression (MESCC)


Epidemiology
■ MESCC is second only to brain metastasis as the cause

of neurological dysfunction caused by metastasis.2

73724_CH03_Printer.indd 68 03/29/09 2:36:00 PM


Spinal Tumors 69

■ In patients with cancer, approximately 2% to 5% will


develop MESCC.2
■ MESCC most frequently occurs in patients with known
cancer but can be the presenting manifestation in 20% of
patients.3
Pathophysiology
■ Compression of the dural sac and its contents (spinal

cord and/or cauda equina) is caused by an extradural


tumor mass.
■ Common causes of MESCC include prostate cancer,

breast cancer, lung cancer, non-Hodgkin lymphoma,


renal cell cancer, and multiple myeloma.2
■ The site(s) of compression in the spinal column is pro-

portional to the relative bone mass and blood flow for


each part of the spine.2
• 15% in the cervical spine
• 60% in the thoracic spine
• 25% in the lumbosacral spine
■ Methods of metastatic spread to the epidural space are as

follows:
• Hematogenous spread to the vertebral body followed
by extension into the epidural space
• Direct extension into the spinal canal through inter-
vertebral foramen, most commonly associated with
lymphoma and neuroblastomas4
■ Spinal cord damage is caused by direct spinal cord com-

pression and secondary vascular damage.


• Acute compression causes stenosis and occlusion of
the epidural venous plexus, resulting in breakdown of
the blood–spinal cord barrier and vasogenic edema.
• Prolonged compression causes arterial flow obstruc-
tion, resulting in spinal cord ischemia.
Clinical Features
■ Worsening back pain: often localized

■ Radicular pain: often worse at night, lying down, with

Valsalva
■ Weakness: may be upper motor neuron or lower motor

neuron, depending on location of compression

73724_CH03_Printer.indd 69 03/29/09 2:36:00 PM


70 Chapter 3

■ Sensory deficits: usually follow pain and weakness


■ Bladder or bowel dysfunction
■ Inability to walk

Diagnosis
■ A whole-spine MRI with gadolinium is the imaging

modality of choice to evaluate for MESCC.


• The minimum radiologic evidence for cord compres-
sion is indentation of the thecal sac at the level of
clinical features.5
• If patient is not able to undergo an MRI, the CT or
conventional myelogram is an alternative but subopti-
mal choice.
■ Definitive diagnosis is based on pathology, but a pre-

sumptive diagnosis may be made on imaging in a patient


with cancer.

Treatment
■ MESCC is a medical emergency causing paraplegia if

left untreated.
■ Early diagnosis and appropriate treatment can prevent or

reverse paraplegia in most patients.6


■ Corticosteroids

• Corticosteroids are first-line treatment to reduce


edema for all tumor types and to induce tumor lysis
for leukemias and lymphomas.
• The addition of steroids to radiation may improve
ambulatory outcomes.7
■ In patients with MESCC from solid tumors, treat-

ment with dexamethasone followed by radiation


improved ambulatory rates at 3 and 6 months com-
pared with radiation alone.
■ Dexamethasone was given as 96 mg IV followed by

96 mg/day PO 3 3 days followed by a 10-day taper.


• The optimal dosing regimen of corticosteroids is
unknown.
■ Randomized studies of high boluses of dexame-

thasone 96–100 mg versus moderate doses


10–16 mg show no differences in pain or ambula-
tion outcomes.8,9

73724_CH03_Printer.indd 70 03/29/09 2:36:00 PM


Spinal Tumors 71

■ Animal studies suggest that steroids have a


dose–response effect on spinal cord compres-
sion, and thus, some advocate the use of high-
dose dexamethasone for patients who cannot
walk at diagnosis or have rapidly progressive motor
symptoms.2
■ Based on a small phase II study of 20 patients with

MESCC but no neurologic deficits or massive inva-


sion of the spine, radiation therapy without steroids
is a feasible regimen.10
■ Radiation therapy
• There are no randomized studies comparing external
beam radiation to supportive care, but multiple retro-
spective studies demonstrate improvement or stabili-
zation in function with radiation.2
• This is most effective for radiosensitive tumors and for
patients who are ambulatory prior to treatment.
• Nonambulant patients have only an 18% to 29%
chance of regaining ambulation with radiotherapy.5
• In a randomized phase III study of MESCC patients
with short life expectancies, #6 months, or unfavorable
histologies (NSCLC, colorectal, kidney, gastric, head
and neck, liver, bladder, sarcoma, melanoma, uterine
carcinoma), there is no difference in ability to walk,
pain relief, survival, or toxicity between short-course
(16 Gy in two fractions) versus split-course (30 Gy in
eight fractions) radiotherapy.11
• The optimal dose, schedule, or technique for patients
with good prognosis are unknown.5
• Data on newer radiation techniques such as stereotac-
tic radiosurgery in MESCC are sparse.
■ Surgery2
• This may be beneficial in selected groups of patients.
■ Unknown primary tumor

■ Relapse after radiation therapy

■ Progression while on radiation therapy

■ Unstable spine or pathological features

■ Less radiosensitive primary tumors with single area

of cord compression, paraplegia ,48 hours, and a


predicted survival .3 months

73724_CH03_Printer.indd 71 03/29/09 2:36:00 PM


72 Chapter 3

• An anterior surgical approach is often preferred over


laminectomy because most epidural metastases extend
from the vertebral body.
■ An anterior approach allows direct decompression

and spine reconstruction.


■ Laminectomy does not remove the tumor and may

result in spine destabilization.


• The benefit from surgery was demonstrated in a ran-
domized study of patients with less radiosensitive
tumors, a single area of MESCC, paraplegia ,48 hours,
and a predicted survival .3 months comparing direct
decompressive surgery plus radiation therapy versus
radiation therapy alone.12
■ The study was terminated after interim analysis

because of positive results.


■ The surgical arm had higher ambulatory rates (84%

versus 57%) and retained ability to ambulate longer


(median 122 days vs. 13 days).
■ For those unable to walk prior to treatment, more

patients regained the ability to ambulate (10 versus


3 patients).
■ The median overall survival was longer in the surgi-

cal arm (126 days vs. 100 days).


• The benefit of direct decompressive surgery in patients
with good neurological function and stable spines is
unclear.5
■ Chemotherapy
• Chemotherapy is rarely used to treat MESCC even in
chemosensitive tumors because the response is too
slow and unpredictable.
• Chemotherapy may be used as salvage therapy for
patients who are no longer candidates for radiation
therapy or surgery.
■ Recurrent MESCC
• The recurrence rate after successful initial treatment
is 7% to 14%.2
• If the recurrence is within the original radiation field,
then reirradiation is not recommended because of
possible adverse events such as radiation-induced
myelopathy.

73724_CH03_Printer.indd 72 03/29/09 2:36:00 PM


Spinal Tumors 73

Prognosis
■ The median survival for patients with MESCC (based on
retrospective studies) is 3 to 6 months.2
■ Good prognostic factors associated with prolonged sur-

vival include the ability to ambulate before and after


treatment, radiosensitive tumors (multiple myeloma,
germ cell tumors, lymphomas, small cell carcinomas),
no visceral or brain metastases, and a single site of epi-
dural cord compression (as opposed to multiple
sites).2
■ Poor prognostic factors for treatment with radiation

include bony compression and spinal instability.5

Intradural Tumors
■ Thirty percent of all spinal cord tumors are contained
within the intradural space, with an additional 10% span-
ning the extradural and intradural spaces.1
■ Intradural tumors can be further subdivided into
extramedullary (outside the spinal cord parenchyma) and
intramedullary (within the spinal cord parenchyma).

Extramedullary Tumors
■ These account for more than 70% of intradural tumors in
adults.1

Nerve Sheath Tumors


■ Arise from nerve sheath cells covering the spinal nerve

roots
■ Most commonly arise in the lumbosacral region13

■ Histology

• Schwannomas
■ Typically round or oval, lobulated, encapsulated

tumors that contain Schwann cells without inva-


sion of nerve fibers
■ Generally considered benign

■ Most commonly seen in patients with neurofibro-

matosis type 2
■ Represent 25% to 30% of all intraspinal masses14

73724_CH03_Printer.indd 73 03/29/09 2:36:00 PM


74 Chapter 3

■ Intradural in 70% to 75%, extradural in 15%, com-


bined intradural–extradural (dumbbell) in 15%14
■ Commonly arises from the dorsal sensory roots of

the cervical and lumbar spine


• Neurofibromas
■ Contain Schwann cells, fibroblasts, and nerve

fibers
■ Generally considered benign

■ Rare intraspinal tumors except in patients with

neurofibromatosis type 2
• Malignant peripheral nerve-sheath tumors (MPNSTs)
■ Uncommon primary tumor of nerve sheath origin

■ More than 50% of MPNSTs associated with neuro-

fibromatosis type 115


■ Prognosis generally poor despite treatment

■ Diagnosis
• MRI with contrast is the imaging modality of choice.
■ Nerve sheath tumors are isointense on T1-weighted

images and hyperintense on T2-weighted images.


■ Schwannomas and neurofibromas are indistinguish-

able on MRIs.
■ Enhancement may be seen with benign or malig-

nant tumors.
■ They may contain both intradural and extradural com-

ponents, creating a dumbbell-shaped appearance.


■ Imaging features suggestive of MPNST include

larger size (.5 cm), heterogeneity, indistinct mar-


gins, and a lack of a pseudocapsule.14
• Definitive diagnosis is made on the basis of histology.
■ Treatment
• Observation
■ Benign, asymptomatic schwannomas, or neurofibro-

mas could be followed with serial imaging.


■ Definitive diagnosis requires pathology, however.

• Surgery
■ Total resection is the primary treatment for nerve

sheath tumors.
■ Total resection often requires removal of the

involved ventral or dorsal nerve root and is usually

73724_CH03_Printer.indd 74 03/29/09 2:36:00 PM


Spinal Tumors 75

not associated with pronounced postoperative


motor or sensory deficit.16
• Radiation and/or chemotherapy
■ These treatment modalities are used primarily for

MPNSTs.
• Radiation therapy may provide local control and
delay recurrence in MPNSTs but has little effect
on long-term survival.17
• The role of chemotherapy is controversial in
MPNSTs because they are traditionally consid-
ered chemotherapy insensitive.17
■ These treatment modalities are not indicated for

completely resected schwannomas or neurofibromas.


■ Partially resected or unresectable schwannomas or

neurofibromas
• The role of radiation is controversial.
• Because tumors are benign and slow growing,
tumors could be monitored with serial imaging
and undergo further resection if necessary.
• Patients with neurofibromatosis may be at
higher risk for secondary malignancies from
radiation.

Spinal Meningioma
■ These arise from meningeal cells along the spinal cord

surface.
■ Ninety percent of spinal meningiomas are intradural,

whereas the remaining 10% are extradural or dumbbell


tumors.18
■ Epidemiology is as follows:

• Most occur between the fifth and seventh decades of


life and are more common in women than men.18
• They account for 25% to 46% of all primary intraspinal
neoplasms.19
• They are most commonly located in the thoracic region,
lateral or posterior to the spinal cord.
• They are usually solitary lesions, although patients
with neurofibromatosis type 2 may have multiple
spinal meningiomas.

73724_CH03_Printer.indd 75 03/29/09 2:36:00 PM


76 Chapter 3

■ Clinical features
• The most common presenting symptom is focal pain.
• Myelopathy may be present.
• Neurologic deficits depend on tumor location.
■ Pathology
• Psammomatous meningiomas is the most common
histology, followed by meningothelial and transitional.18
• Atypical or anaplastic meningiomas are rare.
■ Diagnosis
• Spine MRI with contrast is the imaging modality of
choice.
■ Meningiomas are well-circumscribed lesions, are

hypointense to isointense on T1-weighted images,


are hyperintense on T2-weighted images, and dis-
play homogeneous enhancement.
■ Dural tail and calcification may be seen but less

commonly than with intracranial meningiomas.


• Presumptive diagnosis can often be made with imaging,
although definitive diagnosis is based on pathology.
■ Treatment
• Surgery
■ Gross total resection is the primary treatment and

is feasible as meningiomas are usually well circum-


scribed.
■ Recurrence rates with total or subtotal resection is

3% to 7% in WHO grade I meningomas.1


• Radiation therapy
■ Not indicated for completely resected low-grade

meningiomas
■ Indications

• Adjuvant therapy after subtotal resection


• Treatment after recurrence
My xopapillary Ependymoma
■ Typically benign

■ Account for 40% to 50% of spinal ependymomas1

■ Arise from the conus medullaris and filum terminalis

■ Clinical features: may present with features suggestive of

conus medullaris or cauda equina syndrome, including


radicular pain, lower extremity numbness or weakness,
bowel or bladder dysfunction

73724_CH03_Printer.indd 76 03/29/09 2:36:00 PM


Spinal Tumors 77

■ Imaging: circumscribed mass with homogeneous


enhancement, hypointense on T1-weighted images and
hyperintense on T2-weighted images
■ Treatment
• Gross total resection is the primary treatment, but
entrapment of nerve roots may limit resectability.
• Adjuvant radiation may be considered after subtotal
resection or for disseminated tumors.

Extramedullary Leptomeningeal Metastasis (see Chapter 4)

Intramedullary Tumors
■ Account for 20% to 30% of intradural tumors in adults1

■ Clinical features

• May present with slowly progressive myelopathy


• Midline back pain with or without radicular pain at
the level of the tumor
• Leg weakness and sensory changes
• Bowel/ bladder dysfunction are late symptoms

Ependymoma
■ They account for 40% to 60% of intramedullary spinal

cord tumors in adults.1


■ They can occur anywhere in the central nervous system,

but nearly 50% of all ependymomas arise within the spi-


nal canal.20
■ Most arise in the cervical or cervicothoracic regions.

■ Most are benign, slow growing, and tend to compress

rather than infiltrate normal adjacent tissue.21


■ The diagnosis is as follows:

• Spine MRI with contrast is the imaging modality of


choice.
■ Homogeneously enhancing mass
■ Hypointense on T1-weighted images and hyperin-

tense on T2-weighted images


• Definitive diagnosis requires pathology.
• All patients with spinal ependymomas should undergo
screening of the entire neuroaxis with brain MRI with
contrast, whole-spine MRI with contrast, and CSF
cytology.

73724_CH03_Printer.indd 77 03/29/09 2:36:00 PM


78 Chapter 3

■ Treatment
• Surgery
■ Gross total resection is the primary treatment and

is achieved in 50% to 65% of patients.21


■ The extent of resection is the strongest predictor of

survival.21
• Radiation therapy
■ Not indicated after gross total resection in low-

grade tumors
■ May increase tumor-free survival rates in patients

with high-grade ependymomas or after subtotal


resection1
• The role of chemotherapy is controversial.
• Five-year survival rates for low-grade intramedullary
ependymomas are 83% to 100%.1
Astrocytoma
■ These can occur anywhere in the CNS, with approxi-

mately 3% originating in the spinal cord.20


■ The most common locations are the cervical and cervico-

thoracic regions.
■ Astrocytomas usually extend multiple levels at the time

of diagnosis.
■ The pathology is as follows:

• They are graded according to the same WHO criteria


as intracranial astrocytomas.
• Almost two-thirds of intramedullary astrocytomas are
pilocytic astrocytoma or fibrillary astrocytoma.1
• Approximately 10% of intramedullary astrocytomas
are anaplastic astrocytoma and glioblastoma.1
• Except for pilocytic astrocytomas, most spinal cord
astrocytomas typically grow in a diffuse manner, infil-
trating surrounding normal neural tissue.
• Thirty percent to 60% of the tumors are associated with
rostral, caudal, or intratumoral cysts or syringomelias.1
■ Diagnosis

• Spine MRI with contrast is the imaging modality of


choice.

73724_CH03_Printer.indd 78 03/29/09 2:36:00 PM


Spinal Tumors 79

■ Low-grade tumors
• Focal fusiform expansions of the spinal cord,
hypointense to isointense on T1-weighted
images, hyperintense on T2-weighted images
• Little or no cord edema
• May enhance with contrast
• Calcifications are rare
■ High-grade tumors

• Appear heterogeneous because of intratumor


cysts, necrosis, and surrounding edema22
• Heterogeneous enhancement with contrast
• A definitive diagnosis requires pathology.
■ Treatment
• Treatment depends on tumor histology.
• Surgery
■ Gross total resection is feasible in pilocytic

astrocytomas, but may not be feasible in higher


grade tumors because of their infiltrative nature.
■ Electrophysiological monitoring of the spinal cord

during surgery may help with resection.


• Radiation
■ Recommended as adjuvant therapy for high-grade

astrocytomas
■ Could be considered for progressive disease or for

unresectable tumors
• Chemotherapy
■ The role in spinal cord astrocytomas is unknown.

■ Based on its role in intracranial glioblastomas,

temozolomide and other chemotherapies could


be considered in patients with intramedullary
glioblastomas.

Intramedullary Metastasis
■ These are extremely rare and suggest advanced meta-

static disease.
■ The most common tumors causing intramedullary metas-

tases are lung and breast cancer.20

73724_CH03_Printer.indd 79 03/29/09 2:36:00 PM


80 Chapter 3

References
1. Traul DE, Shaffrey ME, Schiff D. Part I: spinal-cord
neoplasms-intradural neoplasms. Lancet Oncol. 2007;8(1):
35–45.
2. Cole JS, Patchell RA. Metastatic epidural spinal cord com-
pression. Lancet Neurol. 2008;7(5):459–466.
3. Schiff D, O’Neill BP, Suman VJ. Spinal epidural metastasis
as the initial manifestation of malignancy: clinical features
and diagnostic approach. Neurology. 1997;49(2):452–456.
4. Sioutos PJ, Arbit E, Meshulam CF, Galicich JH. Spinal
metastases from solid tumors. Analysis of factors affecting
survival. Cancer. 1995;76(8):1453–1459.
5. George R, Jeba J, Ramkumar G, Chacko AG, Leng M,
Tharyan P. Interventions for the treatment of metastatic
extradural spinal cord compression in adults. Cochrane
Database Syst Rev. 2008(4):CD006716.
6. Abrahm JL, Banffy MB, Harris MB. Spinal cord compres-
sion in patients with advanced metastatic cancer: “all I care
about is walking and living my life.” JAMA. 2008;299(8):
937–946.
7. Sorensen S, Helweg-Larsen S, Mouridsen H, Hansen HH.
Effect of high-dose dexamethasone in carcinomatous meta-
static spinal cord compression treated with radiotherapy:
a randomised trial. Eur J Cancer. 1994;30A(1):22–27.
8. Graham PH, Capp A, Delaney G, et al. A pilot randomised
comparison of dexamethasone 96 mg vs 16 mg per day for
malignant spinal-cord compression treated by radiotherapy:
TROG 01.05 Superdex study. Clin Oncol (R Coll Radiol).
2006;18(1):70–76.
9. Vecht CJ, Haaxma-Reiche H, van Putten WL, de Visser M,
Vries EP, Twijnstra A. Initial bolus of conventional versus
high-dose dexamethasone in metastatic spinal cord com-
pression. Neurology. 1989;39(9):1255–1257.
10. Maranzano E, Latini P, Beneventi S, et al. Radiotherapy
without steroids in selected metastatic spinal cord compres-
sion patients. A phase II trial. Am J Clin Oncol. 1996;19(2):
179–183.
11. Maranzano E, Bellavita R, Rossi R, et al. Short-course ver-
sus split-course radiotherapy in metastatic spinal cord
compression: results of a phase III, randomized, multicenter
trial. J Clin Oncol. 2005;23(15):3358–3365.

73724_CH03_Printer.indd 80 03/29/09 2:36:00 PM


Spinal Tumors 81

12. Patchell RA, Tibbs PA, Regine WF, et al. Direct decompressive
surgical resection in the treatment of spinal cord compression
caused by metastatic cancer: a randomised trial. Lancet.
2005;366(9486):643–648.
13. Conti P, Pansini G, Mouchaty H, Capuano C, Conti R.
Spinal neurinomas: retrospective analysis and long-term out-
come of 179 consecutively operated cases and review of the
literature. Surg Neurol. 2004;61(1):34–43; discussion 44.
14. Parmar HA, Ibrahim M, Castillo M, Mukherji SK. Pictorial
essay: diverse imaging features of spinal schwannomas.
J Comput Assist Tomogr. 2007;31(3):329–334.
15. Acharya R, Bhalla S, Sehgal AD. Malignant peripheral nerve
sheath tumor of the cauda equina. Neurol Sci. 2001;22(3):
267–270.
16. Jinnai T, Koyama T. Clinical characteristics of spinal nerve
sheath tumors: analysis of 149 cases. Neurosurgery. 2005;
56(3):510–515.
17. Gupta G, Maniker A. Malignant peripheral nerve sheath
tumors. Neurosurg Focus. 2007;22(6):E12.
18. Saraceni C, Harrop JS. Spinal meningioma: chronicles of
contemporary neurosurgical diagnosis and management.
Clin Neurol Neurosurg. 2009;111(3):221–226.
19. Albanese V, Platania N. Spinal intradural extramedullary
tumors. Personal experience. J Neurosurg Sci. 2002;46(1):
18–24.
20. Parsa AT, Lee J, Parney IF, Weinstein P, McCormick PC,
Ames C. Spinal cord and intradural-extraparenchymal spi-
nal tumors: current best care practices and strategies.
J Neurooncol. 2004;69(1–3):291–318.
21. Ruda R, Gilbert M, Soffietti R. Ependymomas of the adult:
molecular biology and treatment. Curr Opin Neurol. 2008;
21(6):754–761.
22. Runge VM, Muroff LR, Jinkins JR. Central nervous system:
review of clinical use of contrast media. Top Magn Reson
Imaging. 2001;12(4):231–263.

73724_CH03_Printer.indd 81 03/29/09 2:36:00 PM


73724_CH03_Printer.indd 82 03/29/09 2:36:00 PM
C H A P T E R 4

Leptomeningeal
Metastases
Introduction
■ Neoplastic meningitis: spread of malignant cells to the
leptomeninges and subarachnoid space
■ Carcinomatous meningitis or leptomeningeal carcinoma-
tosis: neoplastic meningitis in patients with solid tumors
■ Leukemic meningitis: neoplastic meningitis in patients
with leukemia
■ Lymphomatous meningitis: neoplastic meningitis in
patients with lymphoma

Epidemiology
■ The number of patients diagnosed with neoplastic men-
ingitis varies according to the type of tumor.1,2
• Solid tumors: breast cancer 3%, small cell lung cancer
6%, non-small cell lung cancer 1%, and melanoma 1%
to 5%
• Leukemia 5% to 15%
• Diffuse high-grade non-Hodgkin lymphoma 5% to 15%
■ Risk factors include high serum LDH, low serum

albumin, age ,60, involvement of testis, breast,


bone marrow or more than two extranodal sites
• Non-Hodgkin lymphomas with aggressive clinical
course (e.g., Burkitt lymphoma, lymphoblastic lym-
phoma) have a .25% risk of meningeal relapse without
CNS-directed therapy.3
• Primary brain tumors 1% to 2%
■ Based on autopsy studies, 19% of cancer patients with
neurologic signs/symptoms have evidence of neoplastic
meningitis.4

83

73724_CH04_Printer.indd 83 03/29/09 2:35:00 PM


84 Chapter 4

■ Carcinomas of unknown primary account for 1% to 7% of


patients with neoplastic meningitis.5
■ Presentation may occur in the following circumstances6:
• Widely disseminated and progressive systemic cancer
(60% to 70%)5,7
• Following a disease-free interval (20%)
• First manifestation of cancer (5% to 10%)8
• Synchronous with intraparenchymal brain metastases
(11% to 31%)5,9

Pathogenesis
■ Routes of metastasis are as follows6:
• Hematogenous (arterial or venous plexus of Batson)
• Direct extension from a tumor adjacent to the CSF
space
• Migration along perineural or perivascular spaces
■ Dissemination occurs throughout the CSF to the entire
neuroaxis.
■ Tumor infiltration is most prominent at the skull base,
dorsal surface of spinal cord, and cauda equina.1,10

Clinical Features
■ Clinical features depend on the location of tumor depos-
its and may be multifocal.
• Cerebral hemispheres: headache, mental status changes,
seizures
• Skull base: cranial nerve deficits with diplopia (CN IV
more commonly affected than III, VI), sensory loss
(CN V), facial weakness (CN VII), cochlear dysfunc-
tion (CN VIII), and optic neuropathy (CN II)
• Spine: weakness (lower extremities more than upper
extremities), sensory loss in a dermatomal pattern, pain
in neck or back, radiculopathy, cauda equina syndrome
■ Nuchal rigidity is present in only 15% of cases.6
■ Raised intracranial pressure or hydrocephalus may occur
from obstruction of CSF outflow by tumor deposits.
■ Thirty percent to 40% of patients with leptomeningeal
disease may also have parenchymal brain metastases.6

73724_CH04_Printer.indd 84 03/29/09 2:35:00 PM


Leptomeningeal Metastases 85

Diagnosis
■ Diagnosis in early stages is important to prevent progressive
neurologic dysfunction, as fixed neurologic deficits rarely
respond to treatment.
■ Three methods are used to diagnose leptomeningeal
metastases and should be used in combination: clinical
presentation, cerebrospinal fluid (CSF) cytology, and
neuroimaging.
■ CSF studies
• Increased opening pressure (.200 mm H2O)
• Elevated WBC (.4 per mm3)
• Elevated protein (.50 mg/dL)
• Decreased glucose (,60 mg/dL)
• Positive cytology is diagnostic
■ A volume .10.5 mL is recommended.11

■ A normal CSF cytology does not exclude the diag-

nosis of leptomeningeal metastases.


■ If the first cytology is negative but suspicion for

neoplastic meningitis remains high, CSF studies


should be repeated.
• Up to 45% of patients eventually found to have
positive cytology had a negative cytology on first
examination.4
• Diagnostic yield increases to 80% with second
CSF cytology.5
• Up to 40% of patients with clinically suspected
leptomeningeal disease and negative CSF
cytology had pathologically proven disease at
autopsy.4
■ Factors associated with false negative cytology

include the following11:


• Not collecting CSF from a site of symptomatic
or radiographic involvement
• Small CSF volumes (,10.5 mL)
• Delayed processing of samples
• Obtaining fewer than two samples
• In leukemia and lymphoma, positive flow cytometry or
cytogenetic studies may be more sensitive than CSF
cytology.6

73724_CH04_Printer.indd 85 03/29/09 2:35:00 PM


86 Chapter 4

■ Meningeal biopsy may be considered if no primary cancer


is found and CSF studies are not diagnostic.
■ Radiographic imaging
• A brain and spine MRI with gadolinium is the radio-
graphic test of choice, but findings are nonspecific.
■ Enhancement of leptomeninges (occurring in the

gyri and superficial sulci), ventricles, cranial nerves,


or intradural extramedullary nodules may be seen.
■ Imaging should be performed prior to lumbar

puncture because the procedure can cause a men-


ingeal reaction resulting in dural-arachnoidal
enhancement.12
■ A normal MRI does not exclude diagnosis, as the

false-negative rate is $30%.6


• Radionuclide studies (111indium-diethylenetriamine
pentaacetic acid or 99tecnetium macroaggregated
albumin)
■ These may be used to evaluate CSF flow dynamics

but are not routinely done in many centers.


■ Patients with CSF flow obstruction (as demonstrated

on radionuclide studies) have decreased survival


times compared with patients with normal CSF
flow.13–15
■ Involved field radiation to sites of CSF flow obstruc-

tion restores flow in 30% of patients with spinal


disease and 50% of patients with intracranial
disease.16,17
■ Reestablishment of CSF flow with radiation 1

intrathecal chemotherapy is associated with longer


survival and lower rates of treatment-related mor-
bidity compared with patients with persistent CSF
flow obstruction.13,14

Prognosis
■ The median survival without treatment is 4 to 6 weeks;
death is usually related to neurologic dysfunction.5
■ Treatment may prolong survival to 4 to 6 months,18 but
responses depend on the primary tumor type.

73724_CH04_Printer.indd 86 03/29/09 2:35:00 PM


Leptomeningeal Metastases 87

• Breast cancer is the best responder among solid


tumors, with a median survival of 6 months and 1-year
survival rates of 11% to 25%.11,19
• Favorable responses are also seen in patients with
lymphoma.
• Less than 15% survive longer than 12 months and have
a complete treatment response.2
■ Treatment may improve or stabilize neurologic symp-
toms but generally does not improve fixed neurologic
deficits.
■ The following are poor prognostic factors for survival and
response to treatment6:
• Poor performance status
• Multiple fixed neurologic deficits
• Bulky CNS disease
• Coexistent carcinomatous encephalopathy
• CSF flow abnormalities on radionuclide ventricu-
lography
• Widely metastatic and aggressive cancers that do not
respond well to systemic therapies

Treatment
■ Important for palliation and may prolong survival
• There is no standard treatment.
• Most treatment studies are small, nonrandomized,
and retrospective.
• Conversion of a positive CSF cytology/flow into a
negative result suggests response and continuation of
therapy.
• More aggressive treatments are generally reserved for
patients with a life expectancy of more than 3 months
and a Karnofsky performance status score of more
than 60%.

Surgery
■ Placement of an intraventricular catheter with a subgaleal
reservoir (e.g., Ommaya reservoir) for intrathecal chemo-
therapy and CSF sampling

73724_CH04_Printer.indd 87 03/29/09 2:35:00 PM


88 Chapter 4

• Often preferred to repeated lumbar punctures, as


drug distribution in CSF is more uniform through a
reservoir20–22
• Complications include malposition, obstruction, and
infection
■ Ventriculoperitoneal (VP) shunt for symptomatic hydro-
cephalus
• Does not appear to result in peritoneal dissemination
of cancer23
• May include an in-line on/off valve to allow chemo-
therapy administration in the off position, although it
is unclear whether this improves outcomes

Radiation Therapy
■ Decreasing bulky disease because intrathecal chemo-

therapy is unable to penetrate further than 2 to 3 mm


into a tumor nodule6
■ Palliation of symptoms regardless of presence of bulky

disease or not, including symptoms related to cauda


equina or cranial nerve involvement
■ Restore CSF flow due to obstruction (see the previous

section on radionuclide imaging)


■ Radiation to the entire CNS axis not feasible because of

systemic toxicity

Chemotherapy
Intrathecal (IT) Chemotherapy
■ Agents include methotrexate, liposomal cytarabine, and

thiotepa.
■ This may be administered via lumbar puncture or ven-

tricular reservoir (Ommaya).


■ Complications include the following:

• CSF infection with Ommaya reservoir


■ This occurs in 2% to 13% of patients on IT chemo-

therapy.6
■ Signs/symptoms include headache, mental status

change, fever, and reservoir malfunction.


■ These are most frequently caused by Staphylococcus

epidermidis.

73724_CH04_Printer.indd 88 03/29/09 2:35:00 PM


Leptomeningeal Metastases 89

■ Treatment includes intravenous antibiotics 6 oral/


intraventricular antibiotics and possibly removal of
the ventricular reservoir.
• Myelosuppression
• Chemical aseptic meningitis
■ Occurs in almost 50% of patients on IT chemo-

therapy6
■ Usually caused by an inflammatory reaction

■ Signs/symptoms: fever, headache, nausea, vomiting,

meningismus, and photophobia


■ May be managed as an outpatient with antipyretics,

antiemetics, and corticosteroids


• Chemotherapy-related neurotoxicity, including sub-
acute leukoencephalopathy or myelopathy, is rare.
• The combination of radiation and chemotherapy
(especially methotrexate) may result in late leukoen-
cephalopathy.6

Systemic Chemotherapy
■ Some agents are able to penetrate the blood–brain bar-

rier, resulting in cytotoxic CSF levels, including high-dose


methotrexate, cytarabine, and thiotepa.
■ Some studies suggest that certain systemic chemothera-

pies are associated with better outcomes than IT


chemotherapies while sparing the complications of IT
treatments.24–26
■ In patients with methotrexate sensitive cancers (i.e.,

lymphoma and breast cancer), high-dose methotrexate


may be used to treat parenchymal and leptomeningeal
metastases.26
■ Hormone therapy is occasionally useful in breast

cancer.27,28

Supportive Care
■ Should be offered to all patients with neoplastic meningitis

regardless of treatment plan


■ Anticonvulsants for seizures

■ Analgesia and pain control

■ Corticosteroids for vasogenic edema from intraparen-

chymal or epidural metastases

73724_CH04_Printer.indd 89 03/29/09 2:35:00 PM


90 Chapter 4

■ Antiemetics for nausea and vomiting


■ Psychostimulants for decreased attention and somno-
lence

References
1. Chamberlain MC. Carcinomatous meningitis. Arch Neurol.
1997;54(1):16–17.
2. Gleissner B, Chamberlain MC. Neoplastic meningitis.
Lancet Neurol. 2006;5(5):443–452.
3. Gokbuget N, Hoelzer D. Meningeosis leukaemica in adult
acute lymphoblastic leukaemia. J Neurooncol. 1998;38
(2–3):167–180.
4. Glass JP, Melamed M, Chernik NL, Posner JB. Malignant
cells in cerebrospinal fluid (CSF): the meaning of a positive
CSF cytology. Neurology. 1979;29(10):1369–1375.
5. Wasserstrom WR, Glass JP, Posner JB. Diagnosis and
treatment of leptomeningeal metastases from solid tumors:
experience with 90 patients. Cancer. 1982;49(4):759–772.
6. Chamberlain MC. Neoplastic meningitis. Oncologist.
2008;13(9):967–977.
7. Balm M, Hammack J. Leptomeningeal carcinomatosis.
Presenting features and prognostic factors. Arch Neurol.
1996;53(7):626–632.
8. Yap HY, Yap BS, Tashima CK, DiStefano A, Blumenschein GR.
Meningeal carcinomatosis in breast cancer. Cancer.
1978;42(1):283–286.
9. Freilich RJ, Krol G, DeAngelis LM. Neuroimaging and cere-
brospinal fluid cytology in the diagnosis of leptomeningeal
metastasis. Ann Neurol. 1995;38(1):51–57.
10. Little JR, Dale AJ, Okazaki H. Meningeal carcinomatosis.
Clinical manifestations. Arch Neurol. 1974;30(2):138–143.
11. Glantz MJ, Cole BF, Glantz LK, et al. Cerebrospinal fluid
cytology in patients with cancer: minimizing false-negative
results. Cancer. 1998;82(4):733–739.
12. Mittl RL, Jr., Yousem DM. Frequency of unexplained men-
ingeal enhancement in the brain after lumbar puncture.
AJNR Am J Neuroradiol. 1994;15(4):633–638.
13. Chamberlain MC, Kormanik PA. Prognostic significance of
111
indium-DTPA CSF flow studies in leptomeningeal metas-
tases. Neurology. 1996;46(6):1674–1677.
14. Glantz MJ, Hall WA, Cole BF, et al. Diagnosis, management,
and survival of patients with leptomeningeal cancer based

73724_CH04_Printer.indd 90 03/29/09 2:35:00 PM


Leptomeningeal Metastases 91

on cerebrospinal fluid-flow status. Cancer. 1995;75(12):


2919–2931.
15. Mason WP, Yeh SD, DeAngelis LM. 111Indium-diethylene-
triamine pentaacetic acid cerebrospinal fluid flow studies
predict distribution of intrathecally administered chemo-
therapy and outcome in patients with leptomeningeal metas-
tases. Neurology. 1998;50(2):438–444.
16. Chamberlain MC, Corey-Bloom J. Leptomeningeal metas-
tases: 111indium-DTPA CSF flow studies. Neurology. 1991;
41(11):1765–1769.
17. Chamberlain MC, Kormanik P, Jaeckle KA, Glantz M.
111
Indium-diethylenetriamine pentaacetic acid CSF flow
studies predict distribution of intrathecally administered
chemotherapy and outcome in patients with leptomeningeal
metastases. Neurology. 1999;52(1):216–217.
18. Grossman SA, Krabak MJ. Leptomeningeal carcinomatosis.
Cancer Treat Rev. 1999;25(2):103–119.
19. Hildebrand J. Prophylaxis and treatment of leptomeningeal
carcinomatosis in solid tumors of adulthood. J Neurooncol.
1998;38(2–3):193–198.
20. Berweiler U, Krone A, Tonn JC. Reservoir systems for intra-
ventricular chemotherapy. J Neurooncol. 1998;38(2–3):
141–143.
21. Sandberg DI, Bilsky MH, Souweidane MM, Bzdil J, Gutin PH.
Ommaya reservoirs for the treatment of leptomeningeal
metastases. Neurosurgery. 2000;47(1):49–54; discussion
54–55.
22. Shapiro WR, Young DF, Mehta BM. Methotrexate: distribu-
tion in cerebrospinal fluid after intravenous, ventricular and
lumbar injections. N Engl J Med. 1975;293(4):161–166.
23. Omuro AM, Lallana EC, Bilsky MH, DeAngelis LM.
Ventriculoperitoneal shunt in patients with leptomeningeal
metastasis. Neurology. 2005;64(9):1625–1627.
24. Bokstein F, Lossos A, Siegal T. Leptomeningeal metastases
from solid tumors: a comparison of two prospective series
treated with and without intra-cerebrospinal fluid chemo-
therapy. Cancer. 1998;82(9):1756–1763.
25. Boogerd W, van den Bent MJ, Koehler PJ, et al. The rele-
vance of intraventricular chemotherapy for leptomeningeal
metastasis in breast cancer: a randomised study. Eur J
Cancer. 2004;40(18):2726–2733.
26. Glantz MJ, Cole BF, Recht L, et al. High-dose intravenous
methotrexate for patients with nonleukemic leptomeningeal

73724_CH04_Printer.indd 91 03/29/09 2:35:00 PM


92 Chapter 4

cancer: is intrathecal chemotherapy necessary? J Clin Oncol.


1998;16(4):1561–1567.
27. Boogerd W, Dorresteijn LD, van Der Sande JJ, de Gast GC,
Bruning PF. Response of leptomeningeal metastases from
breast cancer to hormonal therapy. Neurology. 2000;
55(1):117–119.
28. Ozdogan M, Samur M, Bozcuk HS, et al. Durable remission
of leptomeningeal metastasis of breast cancer with letrozole:
a case report and implications of biomarkers on treatment
selection. Jpn J Clin Oncol. 2003;33(5):229–231.

73724_CH04_Printer.indd 92 03/29/09 2:35:00 PM


C H A P T E R 5

Neurologic
Complications of
Radiation Therapy
Introduction
■ Ionizing radiation damages DNA to create permanent
cell injury or death but can also damage other intracellu-
lar molecules such as lipids or proteins.
■ Radiation is nonspecifically toxic and therefore can dam-
age surrounding normal neural tissue.

Description of Radiation Terms


■ Fractionation: administration of radiation therapy in

divided doses
• Equally efficacious and better tolerated than a single
dose
• Spares normal tissues by allowing time to repair and
repopulation of normal cells between fractions
■ Gray (Gy): absorption of 1 joule per kilogram

■ External beam therapy (EBRT): uses a focused beam of

high-energy rays generated outside the patient


■ Intensity-modulated radiation therapy (IMRT): assigns

nonuniform intensities to individual rays within each


beam to better control dose distributions
■ Stereotactic radiosurgery (SRS): technique used to pre-

cisely deliver a single, high-dose fraction of external beam


radiation to a small intracranial volume. Methods of
delivery include charged particle beams, modified linear
accelerators, and gamma knife units.
■ Stereotactic radiotherapy (SRT): delivery of multiple

fractions using the stereotactic process


■ Brachytherapy: a form of radiation therapy where a radio-

active source is placed inside or next to the tumor

93

73724_CH05_Printer.indd 93 03/29/09 2:35:00 PM


94 Chapter 5

■ Whole-brain radiation therapy (WBRT): a form of


external radiation that treats the entire brain
■ Prophylactic cranial irradiation (PCI): radiation ther-
apy provided to the head to reduce the risk of brain
metastases

Radiation Effects on the Central Nervous


System
■ Cranial irradiation is used as a treatment modality in
many primary brain tumors, metastatic disease to the
brain, CNS involvement of leukemia/lymphomas, and
head/neck cancers.
■ The radiation field may include healthy brain tissue,
resulting in radiation toxicity.

Timing of Radiation Toxicity


Acute Effects
■ Acute radiation toxicity occurs during or shortly after

radiation.
■ Symptoms include the following:

• The most common symptom is progressive fatigue,


which may persist for several weeks after the comple-
tion of radiation, but is generally reversible.
• A less common, more severe acute radiation enceph-
alopathy (dizziness, signs of increased intracranial
pressure, or focal neurologic deficits) may occur.
■ Symptoms may be secondary to edema and disruption of

the blood–brain barrier.


■ Steroid treatment results in clinical improvement of

progressive fatigue and other mild symptoms, but


increased intracranial pressure requires more aggressive
intervention.

Early-Delayed Effects
■ Early-delayed radiation toxicity occurs 6 to 12 weeks after
radiation.
■ Symptoms after cranial radiation include general-

ized weakness, fatigue, somnolence, and cognitive


dysfunction.

73724_CH05_Printer.indd 94 03/29/09 2:35:00 PM


Neurologic Complications of Radiation Therapy 95

■ Manifestations are mostly transient and reversible.


■ These are secondary to transient demyelination, edema,
and disruption of the blood–brain barrier.
Late-Delayed Effects
■ Late delayed radiation toxicity occurs months to years
after radiation.
■ Clinical manifestations may include

• Minor to severe neurocognitive dysfunction (discussed


further later in this chapter)
• Focal radiation necrosis of brain parenchyma (discussed
further later in this chapter)
• Diffuse radiation necrosis of brain parenchyma
• Chronic progressive myelopathy due to spinal cord
injury
• Endocrine dysfunction, usually due to hypothalamic
injury
• Radiation-induced brain tumors (discussed further
later in this chapter)
• Accelerated atherosclerosis affecting the carotid arter-
ies, after neck irradiation (see Chapter 8)
■ Manifestations are often irreversible.

■ Hippocampal dysfunction may account for some of the

neurocognitive changes.
■ The pathophysiology is not completely understood, but

the following have been implicated:


• Excessive generation of reactive oxygen species from
injured and/or proinflammatory cells
• Vascular endothelial damage
• Long-term damage to neural cell types

Risk Factors for Radiation Toxicity


■ Total radiation dose (.50 Gy)
■ Fraction size (.2 Gy)

■ Volume of tissue being irradiated

■ Administration of radiation with chemotherapy1

■ Novel radiation delivery technique (e.g., stereotactic

radiosurgery)
■ Young children, especially during developmental stages

of neural tissue

73724_CH05_Printer.indd 95 03/29/09 2:35:00 PM


96 Chapter 5

■ Old age (.60 years)


■ Vascular risk factors
■ Genetic instability syndromes (e.g., neurofibromatosis)
■ Close proximity to nervous system structures
■ Tumors compatible with long survival

Pathology
■ Pathological changes are not specific to radiation and
include the following:
• Parenchymal cell loss, including white matter
demyelination, encephalomalacia, gliosis, and neural
cell loss
• Vascular changes, including endothelial damage and
fibrinoid necrosis in vessels, resulting in vascular
occlusion and tissue necrosis
■ Brain damage can be focal or diffuse.

Imaging Abnormalities Following Radiation Therapy


■ White matter changes

• White matter abnormalities may be present in some


patients as early as 2 to 6 months after completion of
radiation therapy, likely representing early-delayed
radiation-induced injury.2
• Periventricular white matter changes occur 12 to
18 months after radiation therapy.2,3
• White matter changes are more severe in older
patients.4,5
• The data are conflicting on whether white matter
changes correlate with neurologic dysfunction.
■ Lacunar lesions

■ Cerebral atrophy

Neurocognitive Dysfunction
■ Radiation may have long-term detrimental effects on
neurocognitive function.
■ Radiation may also have short-term beneficial effects

on neurocognitive function because of improved tumor


control.

73724_CH05_Printer.indd 96 03/29/09 2:35:00 PM


Neurologic Complications of Radiation Therapy 97

■ It is often difficult to determine whether the decline in


cognitive function is attributable to therapy or another
etiology such as
• Disease progression
• Radiation
• Surgery
• Chemotherapy
• Medications (including corticosteroids)
• Paraneoplastic disorder

Clinical Features 6
■ Short-term memory deficits

■ Difficulty with spatial relations

■ Difficulty with visual motor processing

■ Difficulty with quantitative skills

■ Deficits in attention

■ Subcortical dementia with gait disturbance and inconti-

nence

Pathophysiology
■ Early memory impairment after WBRT may be related to

hippocampus damage.7–9

Clinical Studies
■ Radiation is an important cause of neurocognitive dys-
function based on data from retrospective studies10,11 and
nonrandomized prospective studies12 in brain metastases
and primary brain tumors.
■ Progression of cognitive deficits may only become appar-

ent after several years.


• In a longitudinal study of patients with low-grade
glioma at a mean 12 years after diagnosis, patients
who received radiation therapy had more deficits in
attentional functioning, executive functioning, and
information processing speed than patients who did
not have radiation therapy (no differences seen
between groups at a mean 6 years after diagnosis).13
• Data on late cognitive effects in patients with brain metas-
tases are sparse because of death in all studies.12

73724_CH05_Printer.indd 97 03/29/09 2:35:00 PM


98 Chapter 5

■ Studies on neurocognitive function, however, may also be


confounded by other factors such as tumor progression
and baseline cognitive dysfunction.
• Several prospective studies report cognitive impairment
in cancer patients even before radiation therapy.14,15
• Results from other prospective studies suggest that
tumor control is strongly associated with neurocogni-
tive stability.15
■ Some data suggest that radiation does not significantly
affect cognition in patients with brain metastases based
on short-term (1–2 years) follow-up.
• A prospective study of 208 patients with brain metas-
tases treated with WBRT showed stable neurocogni-
tive function on formal testing in long-term survivors
at 15 months compared with baseline.16
• Randomized studies of prophylactic cranial irradiation
(PCI) in patients with small cell lung cancer suggest
that patients have substantial levels of impairments at
randomization (even before PCI)17 and show no differ-
ences in neuropsychological testing between the PCI
group and the no PCI group.17,18
■ One should consider the likelihood of radiation-induced
neurocognitive decline versus tumor progression when
considering whether to provide or delay brain radiation.

Treatment
• Donepezil may improve cognition and quality of life in
irradiated brain tumor patients.19
• VP shunting is an option for radiation-induced hydro-
cephalus.
• Other interventions used in the treatment of cognitive
impairment (such as cognitive rehabilitation) may also
be considered but have not been tested specifically in
the irradiated brain tumor population.

Focal Radiation Necrosis


■ This is a severe form of radiation-induced injury, mostly

affecting white matter, although lesions may extend into


cortex or deep gray matter.

73724_CH05_Printer.indd 98 03/29/09 2:35:00 PM


Neurologic Complications of Radiation Therapy 99

Epidemiology
■ The incidence after conventional therapy is unknown
because most studies were performed prior to modern
imaging, but ranges from 3% to 24% have been reported.1

Clinical Features
■ Best characterized following conventional external beam
radiation, although may be relatively more common after
stereotactic radiosurgery or brachytherapy
■ Rarely occurs with cumulative doses of standard fraction-

ated radiation less than 50–60 Gy to the brain or 45 Gy


to the spinal cord20
■ Uncommon in patients who receive WBRT for brain

metastases, although radiation necrosis after WBRT has


been reported
■ Generally occurs as a late complication of radiation, as

early as 2 months and as late as 47 months after radiation


therapy1
• The mean interval from the end of radiation therapy is
approximately 12 months.
• The latency period is approximately five times shorter
in patients with glioma than other types of tumors.
■ Nonspecific presenting signs and symptoms such as focal

neurologic deficits, seizures, confusion, or headaches

Pathophysiology
■ Poorly understood
■ Pathologically characterized by necrosis with hypocellu-

lar edges and dystrophic calcifications, vasculopathy


(telangiectatic, hyalinized, angionecrotic blood vessels),
hemorrhage, edema, and gliosis
■ Potential causes21

• Vascular damage: radiation-induced endothelial dam-


age leading to microvasculopathy, vascular insuffi-
ciency, and infarction followed by gray and/or white
matter necrosis
• Glial damage: radiation-induced glial damage leading to
ablation of glial precursors and demyelinative necrosis

73724_CH05_Printer.indd 99 03/29/09 2:35:00 PM


100 Chapter 5

Diagnosis
■ Radiation necrosis is difficult to distinguish from tumor
recurrence.
■ Specific patterns of enhancement on MRI (“soap bub-

bles” or “Swiss cheese”) have been associated with radia-


tion necrosis as opposed to tumor recurrence.22
■ Radiation necrosis is typically hypometabolic on PET,

whereas tumor recurrence may be hypermetabolic.


■ A definitive diagnosis requires pathology.

Treatment
■ Surgery is not always necessary but may be an option

for symptomatic control in the setting of raised intrac-


ranial pressure or progression despite conservative
management.
■ Resolution of radionecrosis is reported in cases or case

series with the following treatments, but efficacy has not


been established.
• Corticosteroids1
• Bevacizumab
■ A retrospective review of six glioma patients with

biopsy-confirmed radiation necrosis demonstrated


radiographic response in six of six patients and clin-
ical improvements in three of six patients.23
■ A retrospective review of eight glioma patients (two

with biopsy-confirmed radiation necrosis) demon-


strated radiographic improvements in postcontrast
and FLAIR images.24
• Anticoagulation
■ In a small study of eight patients with biopsy-

confirmed radiation necrosis treated with heparin


followed by warfarin for 3–6 months, clinical improve-
ment were noted in five of eight patients, but symp-
toms reemerged after stopping anticoagulation.25
• Hyperbaric oxygen therapy
■ Typical regimen: oxygen was delivered at 20–24

atmospheres for 20–30 sessions, with each session


lasting 90–120 minutes.
■ In a study of 10 patients (8 with biopsy-confirmed

radiation necrosis), clinical improvements were

73724_CH05_Printer.indd 100 03/29/09 2:35:00 PM


Neurologic Complications of Radiation Therapy 101

noted in 6 of 10 patients, with three documented


radiographic responses, but many of these patients
also received high-dose steroids.26

Radiation-Induced Brain Tumors


■ The most common secondary tumors in the CNS are

meningiomas, nerve sheath tumors, pituitary adenomas,


gliomas, sarcomas, and embryonal neoplasms.
■ Radiation-induced brain tumors tend to be more aggres-

sive and higher grade.

Epidemiology
■ Based on a retrospective review of 10,080 children treated

with low-dose scalp irradiation (mean 1.4–1.8 Gy) for


tinea capitis, relative risks as compared with matched
nonirradiated controls for meningiomas were 9.5 and for
gliomas were 2.6.27
■ In a study of 9,720 children treated for prophylactic or

therapeutic CNS irradiation for ALL, these patients suf-


fered a 22-fold excess of neoplasms, mostly gliomas and
primitive neuroectodermal tumors (PNET).28
■ The latency period after radiation varies but typically

presents years to decades after therapy.29

Risk Factors
■ Radiation during childhood
■ Higher radiation doses

Treatment
■ Treatment options are similar to analogous non–

radiation-induced tumors but are rarely successful.30

Radiation Effects on the Peripheral Nervous


System
■ Most effects on the peripheral nervous system occur in a
late-delayed fashion.

Postradiation Optic Neuropathy


■ This is caused by injury to the optic nerve and apparatus.

73724_CH05_Printer.indd 101 03/29/09 2:35:00 PM


102 Chapter 5

■ It is most often associated with radiation to the optic


pathways but can occur with radiation to tumors in close
proximity to the optic pathways.
■ It occurs 3 months to 8 years after radiation therapy.31
■ It presents with painless, progressive, monocular vision
loss and constriction of visual fields.30
■ It is usually irreversible.
■ Optimal management is unknown.31

Radiation Plexopathy
■ It may occur in the brachial plexus after radiation for lung

or breast cancer or the lumbosacral plexus following radi-


ation for pelvic or lower abdominal tumors.
Clinical Features
■ Most occur 5 to 30 months after radiation therapy.
■ Predominating symptoms are gradually progressive weak-

ness and paresthesias.


■ Ipsilateral lymphedema is common in radiation-induced

brachial plexopathy.
■ Bilateral plexus involvement (although asymmetric) is

common in radiation-induced lumbosacral plexopathy.


■ Pain is uncommon, which distinguishes radiation plex-

opathy from tumor infiltration or compression.


Diagnosis
■ It is based on clinical diagnosis.
■ Electromyography may demonstrate myokymias, which is

seen only in radiation plexopathy.22


■ MRI of the plexus may be used to exclude tumor infiltra-

tion or compression.
Treatment
■ Pain can be managed with NSAIDs or opioids.

■ For severe plexopathy, surgical exploration may be con-

sidered to release neural elements from fibrotic tissues.32

Radiation-Induced Peripheral Nerve Tumors


■ Includes schwannomas, neurofibromas, and malignant

peripheral nerve sheath tumors (MPNSTs)

73724_CH05_Printer.indd 102 03/29/09 2:35:00 PM


Neurologic Complications of Radiation Therapy 103

Epidemiology
■ This is a rare complication from radiation, but the true
incidence is unknown.
■ The association between radiation and peripheral nerve

tumors has been demonstrated in several large retrospec-


tive studies.33
■ Based on a retrospective review of 10,080 children

treated with low-dose scalp irradiation for tinea capitis,


the relative risk of developing cranial nerve schwanno-
mas compared with matched nonirradiated controls
was 18.8.27

Risk Factors
■ Clinical risk factors for development of radiation-induced
neurofibromas include radiation at a young age, heavy
use of radiation, and long-term survival (owing to the long
latency between radiation and diagnosis of a peripheral
nerve sheath tumor).34
■ Patients with neurofibromatosis are at elevated risk

of developing peripheral nerve tumors after radiation


therapy.35

References
1. Ruben JD, Dally M, Bailey M, Smith R, McLean CA,
Fedele P. Cerebral radiation necrosis: incidence, outcomes,
and risk factors with emphasis on radiation parameters and
chemotherapy. Int J Radiat Oncol Biol Phys. 2006;65(2):
499–508.
2. Constine LS, Konski A, Ekholm S, McDonald S, Rubin P.
Adverse effects of brain irradiation correlated with MR
and CT imaging. Int J Radiat Oncol Biol Phys. 1988;15(2):
319–330.
3. Packer RJ, Zimmerman RA, Bilaniuk LT. Magnetic reso-
nance imaging in the evaluation of treatment-related central
nervous system damage. Cancer. 1986;58(3):635–640.
4. Johannesen TB, Lien HH, Hole KH, Lote K. Radiological
and clinical assessment of long-term brain tumour survivors
after radiotherapy. Radiother Oncol. 2003;69(2):169–176.
5. Tsuruda JS, Kortman KE, Bradley WG, Wheeler DC, Van
Dalsem W, Bradley TP. Radiation effects on cerebral white

73724_CH05_Printer.indd 103 03/29/09 2:35:00 PM


104 Chapter 5

matter: MR evaluation. AJR Am J Roentgenol. 1987;149(1):


165–171.
6. Akyurek S, Senturk V, Oncu B, Ozyigit G, Yilmaz S, Gokce SC.
The effect of tianeptine in the prevention of radiation-induced
neurocognitive impairment. Med Hypotheses. 2008;71(6):
930–932.
7. Mizumatsu S, Monje ML, Morhardt DR, Rola R, Palmer TD,
Fike JR. Extreme sensitivity of adult neurogenesis to low
doses of X-irradiation. Cancer Res. 2003;63(14):4021–4027.
8. Monje ML, Mizumatsu S, Fike JR, Palmer TD. Irradiation
induces neural precursor-cell dysfunction. Nat Med.
2002;8(9):955–962.
9. Monje ML, Palmer T. Radiation injury and neurogenesis.
Curr Opin Neurol. 2003;16(2):129–134.
10. DeAngelis LM, Delattre JY, Posner JB. Radiation-induced
dementia in patients cured of brain metastases. Neurology.
1989;39(6):789–796.
11. Frytak S, Shaw JN, O’Neill BP, et al. Leukoencephalopathy
in small cell lung cancer patients receiving prophylactic
cranial irradiation. Am J Clin Oncol. 1989;12(1):27–33.
12. Ricard D, Taillia H, Renard JL. Brain damage from antican-
cer treatments in adults. Curr Opin Oncol. 2009.
13. Douw L, Klein M, Fagel SS, et al. Cognitive and radiological
effects of radiotherapy in patients with low-grade glioma:
long-term follow-up. Lancet Neurol. 2009;8(9):810–818.
14. Welzel G, Fleckenstein K, Schaefer J, et al. Memory func-
tion before and after whole brain radiotherapy in patients
with and without brain metastases. Int J Radiat Oncol Biol
Phys. 2008;72(5):1311–1318.
15. Brown PD, Kee AY, Eshleman JS, Fiveash JB. Adjuvant
whole brain radiotherapy: strong emotions decide but ratio-
nale studies are needed: in regard to Brown et al. (Int
J Radiat Oncol Biol Phys 2008;70:1305–1309). In reply to
Drs. Larson and Sahgal. Int J Radiat Oncol Biol Phys. 2009;
75(1):316–317.
16. Li J, Bentzen SM, Renschler M, Mehta MP. Regression
after whole-brain radiation therapy for brain metastases cor-
relates with survival and improved neurocognitive function.
J Clin Oncol. 2007;25(10):1260–1266.
17. Gregor A, Cull A, Stephens RJ, et al. Prophylactic cranial
irradiation is indicated following complete response to
induction therapy in small cell lung cancer: results of a
multicentre randomised trial. United Kingdom Coor-
dinating Committee for Cancer Research (UKCCCR) and

73724_CH05_Printer.indd 104 03/29/09 2:35:00 PM


Neurologic Complications of Radiation Therapy 105

the European Organization for Research and Treatment


of Cancer (EORTC). Eur J Cancer. 1997; 33(11):
1752–1758.
18. Arriagada R, Le Chevalier T, Borie F, et al. Prophylactic cra-
nial irradiation for patients with small-cell lung cancer
in complete remission. J Natl Cancer Inst. 1995;87(3):
183–190.
19. Shaw EG, Rosdhal R, D’Agostino RB, Jr., et al. Phase II
study of donepezil in irradiated brain tumor patients: effect
on cognitive function, mood, and quality of life. J Clin
Oncol. 2006;24(9):1415–1420.
20. Schultheiss TE, Kun LE, Ang KK, Stephens LC. Radiation
response of the central nervous system. Int J Radiat Oncol
Biol Phys. 1995;31(5):1093–1112.
21. Yoshii Y. Pathological review of late cerebral radionecrosis.
Brain Tumor Pathol. 2008;25(2):51–58.
22. Kumar AJ, Leeds NE, Fuller GN, et al. Malignant gliomas: MR
imaging spectrum of radiation therapy- and chemotherapy-
induced necrosis of the brain after treatment. Radiology.
2000;217(2):377–384.
23. Torcuator R, Zuniga R, Mohan YS, et al. Initial experience
with bevacizumab treatment for biopsy confirmed cerebral
radiation necrosis. J Neurooncol. 2009;94(1):63–68.
24. Gonzalez J, Kumar AJ, Conrad CA, Levin VA. Effect of bev-
acizumab on radiation necrosis of the brain. Int J Radiat
Oncol Biol Phys. 2007;67(2):323–326.
25. Glantz MJ, Burger PC, Friedman AH, Radtke RA, Massey
EW, Schold SC, Jr. Treatment of radiation-induced nervous
system injury with heparin and warfarin. Neurology.
1994;44(11):2020–2027.
26. Chuba PJ, Aronin P, Bhambhani K, et al. Hyperbaric oxygen
therapy for radiation-induced brain injury in children.
Cancer. 1997;80(10):2005–2012.
27. Ron E, Modan B, Boice JD, Jr., et al. Tumors of the brain
and nervous system after radiotherapy in childhood. N Engl
J Med. 1988;319(16):1033–1039.
28. Neglia JP, Meadows AT, Robison LL, et al. Second neo-
plasms after acute lymphoblastic leukemia in childhood.
N Engl J Med. 1991;325(19):1330–1336.
29. Perry A, Schmidt RE. Cancer therapy-associated CNS neu-
ropathology: an update and review of the literature. Acta
Neuropathol. 2006;111(3):197–212.
30. Cross NE, Glantz MJ. Neurologic complications of radia-
tion therapy. Neurol Clin. 2003;21(1):249–277.

73724_CH05_Printer.indd 105 03/29/09 2:35:00 PM


106 Chapter 5

31. Durkin SR, Roos D, Higgs B, Casson RJ, Selva D.


Ophthalmic and adnexal complications of radiotherapy.
Acta Ophthalmol Scand. 2007;85(3):240–250.
32. Gosk J, Rutowski R, Reichert P, Rabczynski J. Radiation-
induced brachial plexus neuropathy-aetiopathogenesis, risk
factors, differential diagnostics, symptoms and treatment.
Folia Neuropathol. 2007;45(1):26–30.
33. Zadeh G, Buckle C, Shannon P, Massicotte EM, Wong S,
Guha A. Radiation induced peripheral nerve tumors: case
series and review of the literature. J Neurooncol. 2007;83(2):
205–212.
34. Donohue WL, Jaffe FA, Rewcastle NB. Radiation induced
neurofibromata. Cancer. 1967;20(4):589–595.
35. Sznajder L, Abrahams C, Parry DM, Gierlowski TC,
Shore-Freedman E, Schneider AB. Multiple schwannomas
and meningiomas associated with irradiation in childhood.
Arch Intern Med. 1996;156(16):1873–1878.

73724_CH05_Printer.indd 106 03/29/09 2:35:00 PM


C H A P T E R 6

Neurologic
Complications of
Chemotherapy
Introduction
■ Chemotherapy: the use of chemical agents in the treatment
or control of disease such as cancer

Common Chemotherapy Agents (Table 6-1)


Table 6-1: Common Chemotherapy Agents

Mechanism of
Class of Agent Action Examples

Alkylating Add alkyl groups to Cisplatin


agents DNA, resulting in Carboplatin
cross-linking,
Oxaliplatin
miscoding, or DNA
strand breakage Busulfan
Carmustine (BCNU)
Lomustine (CCNU)
Temozolomide
Cyclophosphamide

Antimetabolites Inhibits DNA synthesis 5-Fluorouracil (5-FU)


Capecitabine
Cytarabine
Gemcitabine
Fludarabine

(Continues)

107

73724_CH06_Printer.indd 107 03/29/09 2:38:00 PM


108 Chapter 6

Table 6-1: Common Chemotherapy Agents (Continued)

Mechanism of
Class of Agent Action Examples

Antifolates Inhibits multiple Methotrexate


enzymatic sites
leading to the
depletion of
intracellular folates
and direct blockage
of purine and
pyrimidine
biosynthesis

Vinca alkaloids Bind to tubulin and Vincristine


inhibit the assembly Vinblastine
of tubulin into
Vinorelbine
microtubules,
thereby preventing
mitosis and cell
division

Taxanes Stabilize Paclitaxel


microtubules, Docetaxel
resulting in inhibition
of mitosis and cell
division

Topoisomerase Inhibit topoisomerase Irinotecan


inhibitors I or II, enzymes which Topotecan
promote DNA strand
Etoposide
unwinding essential
for DNA synthesis and Teniposide
repair Doxorubicin
Epirubicin

Thalidomide and Immunomodulatory, Thalidomide


its analogues anti-inflammatory and Revlimid
antiangiogenic prop-
erties due to down-
regulation of certain
signaling molecules

(Continues)

73724_CH06_Printer.indd 108 03/29/09 2:38:00 PM


Neurologic Complications of Chemotherapy 109

Table 6-1: Common Chemotherapy Agents (Continued)

Mechanism of
Class of Agent Action Examples

Molecular Mechanism varies, Trastuzumab


targeted agents target specific (monoclonal antibody
molecules associated of HER2/neu)
with tumorigenesis, Rituximab (monoclonal
tumor growth and/or antibody of CD20)
progression
Bevacizumab
(monoclonal antibody
of VEGF)
Imatinib (tyrosine
kinase inhibitor of
BCR-Abl)
Erlonitib (ATP pocket
binding inhibitor
of EGFR)
Bortezomib
(proteosome inhibitor)

Antiestrogens Bind the estrogen Tamoxifen


receptor and exert
either estrogenic or
antiestrogenic effects
depending on the
specific organ

Data are from Chabner BA, Lynch TJ, Longo DL, eds. Harrison’s Manual of
Oncology. New York: McGraw Hill Companies; 2008.

Treatment Schemes
Treatment Modalities
■ Neoadjuvant: chemotherapy prior to surgery or radiother-

apy designed to shrink the tumor


■ Concomitant: chemotherapy given during radiation

■ Adjuvant: chemotherapy after surgery or radiotherapy

Palliative Chemotherapy
■ Chemotherapy is given without curative intent.
■ The goal is to decrease tumor burden, provide symptom-

atic relief, and increase life expectancy.

73724_CH06_Printer.indd 109 03/29/09 2:38:00 PM


110 Chapter 6

Central Nervous System


Chemotherapy-Related Cognitive Impairment
(“Chemobrain”)
■ The nature and extent of chemotherapy-related cognitive

impairment is unknown.
■ Studies of cancer-related cognitive dysfunction (prior to

chemotherapy administration) demonstrate baseline dys-


function in several cancer types.1

Clinical Features
■ Symptoms may be subtle but adversely affect quality

of life.
■ They may arise shortly after starting chemotherapy and

persist after the completion of chemotherapy.


■ The neuropsychological profile suggests disruption of

frontal–subcortical networks, including problems with


• Short-term memory
• Executive function
• Working memory
• Sustained attention

Risk Factors1
■ Higher doses of chemotherapy (either through high-dose

regimens or exposure to higher doses caused by impaired


systemic clearance)
■ Multiagent chemotherapy

■ Intrathecal chemotherapy

Pathophysiology
■ Unknown but postulated hypotheses include2

• Chemotherapeutic agents crossing the blood–brain


barrier
• DNA damage and shortened telomere length
• Cytokine dysregulation resulting in higher levels of
proinflammatory cytokines
• Problems with neural repair
• Decreased neurotransmitter activity

73724_CH06_Printer.indd 110 03/29/09 2:38:00 PM


Neurologic Complications of Chemotherapy 111

Treatment
■ There are no standard treatments because of a lack of
studies.
■ Empirical treatments include psychostimulants and cog-

nitive therapy.

Reversible Posterior Leukoencephalopathy


Syndrome (RPLE)
■ Also known as posterior reversible encephalopathy syn-

drome (PRES)
■ Etiologies

• Uncontrolled hypertension
• Eclampsia
• Immunosuppressants such as cyclosporine and
FK-506
• Described in association with several antineoplastic
agents, including asparaginase, bevacizumab, capecit-
abine, carboplatin, cisplatin, cyclophosphamide, cytara-
bine, doxorubicin, etanercept, fluorouracil, gemcitabine,
interferon alpha, irinotecan, melphalan, methotrexate,
oxaliplatin, paclitaxel, prednisone, sorafenib, tacrolimus,
thalidomide, and vincristine3
• No single antineoplastic agent consistently associated
with RPLE
• More frequently associated with multidrug chemo-
therapy than single-agent chemotherapy3
■ Characterized by bilateral, reversible, symmetric abnor-

malities predominantly involving white matter in the


cerebral hemisphere
■ Clinical features

• Typically subacute at onset


• Characterized by headache and altered mental status
with or without seizures
• Occipital involvement may present with visual symp-
toms such as cortical blindness
■ MRI demonstrates bilateral T2-hyperintensities caused

by vasogenic edema typically involving the subcortical


white matter, although additional areas of the brain can
be involved.

73724_CH06_Printer.indd 111 03/29/09 2:38:00 PM


112 Chapter 6

■ Management3
• Early diagnosis important
• Discontinuation of offending agent
• Correction of high blood pressure, renal dysfunction,
and low magnesium

CNS Neurotoxicity of Specific Agents


Cytarabine
■ Also known as cytosine arabinoside and ara-C

■ Used in acute leukemias, chronic myelogenous leukemia,

non-Hodgkin lymphoma, and neoplastic meningitis


■ High-dose cytarabine associated with an acute cerebellar

syndrome4
• Clinical features
■ The predominant clinical feature is cerebellar dys-

function, including dysarthria, dysdiadochokinesia,


dysmetria, and ataxia.
■ Many patients also develop a concomitant cerebral

dysfunction characterized by somnolence, altered


mentation, headache, or seizures.
■ Symptoms are usually noted between 3 and 8 days

after initiation of therapy.


• The outcome is variable, but neurologic dysfunction
resolves within 5 days of discontinuing agent in the
majority of cases.4
■ Intrathecal liposomal cytarabine is associated with aseptic

meningitis.
• Signs and symptoms include headache, nuchal rigidity,
nausea, vomiting, and fever.
• The incidence is higher without dexamethasone pro-
phylaxis.5
• It may resolve spontaneously or respond to steroid
therapy.

5-Fluorouracil (5-FU)
■ Used in colorectal cancer, gastric cancer, breast cancer,
basal cell cancer, head and neck cancer, and bladder cancer
■ Associated with acute and delayed neurotoxicities, although

both are uncommon6

73724_CH06_Printer.indd 112 03/29/09 2:38:00 PM


Neurologic Complications of Chemotherapy 113

• Acute cerebellar toxicity


■ Characterized by incoordination, ataxia, dysarthria,

and nystagmus
■ Often reversible with drug discontinuation

• Acute encephalopathy
■ Characterized by confusion, cognitive disturbances,

and altered mentation


■ Associated with hyperammonemia and lactic acidosis

• Subacute multifocal leukoencephalopathy


■ Reported in cases of fluorouracil and levamisole as

combination therapy in patients with stage C colon


cancer
■ Develops 3 to 5 months after beginning chemo-

therapy
■ Characterized by cognitive abnormalities, altered

consciousness, dysarthria, focal extremity weak-


ness, and ataxia
■ MRI that demonstrates multifocal enhancing

white-matter lesions
■ Clinical improvement with discontinuation of fluo-

rouracil and levamisole as well as administration of


corticosteroids

Fludarabine
■ Used in chronic lymphocytic leukemia and low-grade

lymphoma
■ Associated with a severe neurotoxicity syndrome7

• Described at doses greater than 40 mg/m2/day


• Signs and symptoms that include blindness, encephal-
opathy, coma
• Caused by a diffuse, necrotizing leukoencephalopathy,
most severe in the occipital lobes

Ifosfamide
■ Used in lung cancer, breast cancer, gastric cancer,
Hodgkin lymphoma, non-Hodgkin lymphoma, and soft-
tissue sarcoma
■ Neurotoxicity often the dose-limiting toxicity when

appropriate preventative measures are used to reduce


urotoxicity8

73724_CH06_Printer.indd 113 03/29/09 2:38:00 PM


114 Chapter 6

■ Associated with encephalopathy7


• Seen in 10% to 16% of patients treated with ifosfamide
• Signs and symptoms that range from agitation to sei-
zures to coma
• May improve with discontinuation of ifosfamide and
administration of methylene blue

Interferon-a
■ Uses include hairy cell leukemia, Kaposi sarcoma, mela-
noma, chronic myelogenous leukemia, follicular non-
Hodgkin lymphoma, multiple myeloma, kidney cancer,
and bladder cancer
■ Associated with neuropsychiatric symptoms

• Predominantly depression, which is often reversible


• Rarely associated with cognitive dysfunction

Methotrexate
■ Uses include acute lymphoblastic leukemia, acute myel-
ogenous leukemia, neoplastic meningitis, trophoblastic
tumors, breast cancer, lung cancer, head and neck can-
cers, Burkitt lymphoma, osteosarcoma, and primary CNS
lymphoma
■ CNS toxicity may be associated to homocysteine levels,

folate levels, and genetic variants of methionine metabolism


■ Associated with delayed leukoencephalopathy

• Ranging from asymptomatic white matter changes on


imaging to severe CNS demyelination
• Rarely associated with a disseminated necrotizing
leukoencephalopathy9,10
■ May occur with intrathecal or intravenous metho-

trexate alone but more commonly associated with


the combination of methotrexate and radiation
■ Presenting signs and symptoms that include per-

sonality changes, confusion, altered consciousness,


seizures, and coma
■ MRI that demonstrates multifocal T2-weighted

hyperintensities in the deep white matter with


patchy or diffuse contrast enhancement
■ Often fatal

73724_CH06_Printer.indd 114 03/29/09 2:38:00 PM


Neurologic Complications of Chemotherapy 115

■ Intrathecal methotrexate is associated with aseptic


meningitis.
• Signs and symptoms include headache, nuchal rigid-
ity, nausea, vomiting, and fever occurring 2 to 4 hours
after IT methotrexate and lasting for 12 to 72 hours.
• CSF studies show pleocytosis with negative cultures.
• This may resolve spontaneously or respond to steroid
therapy.

Peripheral Nervous System


Chemotherapy-Induced Peripheral Neuropathy (CIPN)
■ CIPN is a common dose-limiting toxicity for many older
chemotherapeutic agents.
■ CIPN may lead to chemotherapeutic dose reduction,

treatment delay, or treatment discontinuation even if the


patient is responding to the agent.
■ There is no standard of care for the prevention or treat-

ment of CIPN.
Common Chemotherapeutic Agents That Cause Peripheral
Neuropathy (Table 6-2)
■ Vincristine, paclitaxel, cisplatin, and thalidomide are the

most neurotoxic.
Risk Factors11
■ Patient age

■ Dose intensity, cumulative dose, therapy duration

■ Coadministration of other neurotoxic agents

■ Preexisting conditions associated with neuropathy

(i.e., diabetes, alcohol abuse)


Clinical Signs and Symptoms
■ The pattern depends on drug class (Table 6-2).
• Vinca alkaloids, taxanes, and bortezomib are associ-
ated with length-dependent neuropathy predomi-
nantly affecting the small fibers.
• Cisplatin is associated with ataxic neuropathy involv-
ing all four limbs.
• Vincristine and taxanes are also associated with auto-
nomic neuropathy.

73724_CH06_Printer.indd 115 03/29/09 2:38:00 PM


Table 6-2: Chemotherapy-Induced Peripheral Neuropathy
Cumulative

73724_CH06_Printer.indd 116
Drugs Pathophysiology Toxic Dose Neuropathy Installation Evolution Electrophysiology
116 Chapter 6

Vinca Inhibition of .30–50 mg Sensory more Acute Recovery or Axonal distal


alkaloids microtubule than motor Chronic improvement neuropathy
Vincristine assembly painful length- After effect
Axonal transport dependent
Vinblastine
neuropathy
Vinorelbine
Cranial nerves
(oculomotor, facial,
trigeminal,
recurrent)
Optic neuropathy
Autonomic
neuropathy
(especially paralytic
ileus)

(Continues)

03/29/09 2:38:00 PM
Table 6-2: Chemotherapy-Induced Peripheral Neuropathy (Continued)
Cumulative
Drugs Pathophysiology Toxic Dose Neuropathy Installation Evolution Electrophysiology

73724_CH06_Printer.indd 117
Taxanes Promotion of .175–200 mg/m2 Sensory . Acute Recovery or Axonal distal
Paclitaxel microtubule motor, improvement neuropathy
assembly painful, length-
Docetaxel
Axonal dependent
transport neuropathy
Facial nerve palsy
Autonomic
dysfunction

Platinum Inhibition of .300 mg/m2 Sensory Acute Improvement Axonal sensory


analogues protein synthesis for cisplatin and neuronopathy Subacute After effect neuronopathy
Cisplatin Dorsal root oxaliplatin (with all) (Sensory evoked
ganglion apoptosis 400 mg/m2 for Acute transient potentials show a
Carboplatin
carboplatin sensory ganglionopathy)
Oxaliplatin Axonal transport
neuropathy and
Nerve
neuromyotonia
hyperexcitability
(with oxaliplatin)
Antiangiogenesis
Neurologic Complications of Chemotherapy 117

(Continues)

03/29/09 2:38:00 PM
Table 6-2: Chemotherapy-Induced Peripheral Neuropathy (Continued)

Cumulative
Drugs Pathophysiology Toxic Dose Neuropathy Installation Evolution Electrophysiology

73724_CH06_Printer.indd 118
Suramin Inhibition of Guillain-Barré Improvement Demyelinating
118 Chapter 6

. 350 µg/mL
growth factors max plasma syndrome-like After effect polyneuropathy with
Glycolipid level neuropathy conduction blocks
Lysosomal Sensorimotor Axonal distal
length- neuropathy
Inclusion in
dependent
dorsal root
neuropathy
Ganglion

Thalidomide Inhibition of nerve 50 mg/day Sensory painful Chronic Recovery or Axonal sensory
growth factor length-dependent improvement neuropathy
transcription factors neuropathy (sensory evoked
Immunomodulation Sensory potentials show a
neuronopathy ganglionopathy)
antiangiogenesis
Rare sensorimotor
neuropathy

(Continues)

03/29/09 2:38:00 PM
Table 6-2: Chemotherapy-Induced Peripheral Neuropathy (Continued)
Cumulative
Drugs Pathophysiology Toxic Dose Neuropathy Installation Evolution Electrophysiology

73724_CH06_Printer.indd 119
Bortezomib Proteasome Unknown Sensory length- Subacute Recovery or Mainly axonal
inhibition dependent improvement sensory/motor
neuropathy neuropathy
Sensorimotor
length-dependent
neuropathy
Mononeuropathy
multiplex
Demyelinating
neuropathy

Epothilones Promotion of Unknown Sensory


microtubule neuropathy
assembly

From Antoine JC, Camdessanche JP. Peripheral nervous system involvement. Lancet Neurol. 2007;6(1):75–86.
Neurologic Complications of Chemotherapy 119

03/29/09 2:38:00 PM
120 Chapter 6

■ Paresthesias and dysesthesias are commonly seen in toes


and fingers.
■ Symptoms often begin distally and spread proximally to
affect lower and upper extremities in a stocking-glove
distribution.

Prevention
■ Calcium (Ca21) and magnesium (Mg21) infusions may

reduce the severity of oxaliplatin-induced chronic periph-


eral sensory neurotoxicity without reducing response
rates.
• This is based on preliminary results from three ran-
domized, placebo-controlled, double-blinded studies
of intravenous Ca21–Mg21 in oxaliplatin-based treat-
ment regimens for colon or colorectal cancer: the com-
bined oxaliplatin neurotoxicity prevention trial
(CONcePT),12,13 the North Central Cancer Treatment
Group (NCCTG) N04C7 trial,14 and the Neuroxa
trial.15,16
• Based on patient reported outcome analysis in the
NCCTG trial, Ca 21–Mg 21 significantly reduced
muscle cramps and chronic cumulative sensory neu-
rotoxicity, but no effect was noted on phenomena
associated with acute sensory neurotoxicity.
■ Vitamin E may reduce the incidence and/or severity of

chemotherapy-induced peripheral neurotoxicity, but its


effect on chemotherapy responses is unknown.
• The benefit of vitamin E was shown in several trials.
■ Pilot open-label study of patients randomized to

vitamin E 300 mg per day versus no intervention in


patients receiving cisplatin-based regimens17
■ Pilot open-label study of patients randomized to

vitamin E 300 mg twice per day versus no interven-


tion in patients receiving cisplatin and/or paclitaxel
regimens18,19
■ Preliminary results from prospective, randomized,

placebo-controlled, double-blinded study of vitamin E


400 mg per day in patients receiving cisplatin-based
regimens20

73724_CH06_Printer.indd 120 03/29/09 2:38:00 PM


Neurologic Complications of Chemotherapy 121

■ Preliminary results from a prospective, randomized,


placebo-controlled, double-blinded study of vita-
min E 300 mg twice per day in patients receiving a
variety of neurotoxic chemotherapies21
• None of these trials determined whether vitamin E
had adverse effects on responses to treatment.
■ Glutathione may prevent cisplatin- or oxaliplatin-induced
peripheral neurotoxicity without reducing response
rates.
• A randomized, double-blinded study of glutathione in
patients receiving a cisplatin-based regimen for ovar-
ian cancer showed that glutathione reduced neurotox-
icity and improved quality of life.22
• Randomized, placebo-controlled, double-blinded
studies of glutathione in patients receiving a cisplatin-
based regimen for advanced gastric cancer23 or an
oxaliplatin-based regimen for advanced colorectal
cancer24 showed that glutathione reduced neurotoxic-
ity without affecting response rates.
■ Xaliproden may reduce the severity of oxaliplatin-induced
peripheral neurotoxicity without reducing response
rates.
• Xaliproden is an orally administered nonpeptide neu-
rotrophic agent.
• Preliminary results from a phase III randomized,
placebo-controlled, double-blinded study suggest that
xaliproden reduces the risk of higher grade oxaliplatin-
induced peripheral sensory neuropathy without
impacting antitumor activity.25
■ Other agents may protect against CIPN but have not
been tested in large trials.
• Glutamate: earlier small studies performed in patients
receiving paclitaxel suggested a benefit with glutamate
prophylaxis, but in a recent small randomized,
placebo-controlled, double-blinded study with gluta-
mate 500 mg three times per day in ovarian cancer
patients receiving a paclitaxel-based regimen, glutamate
failed to protect against peripheral neurotoxicity.26
• Glutamine for high-dose paclitaxel or oxaliplatin-
induced peripheral neurotoxicity.27

73724_CH06_Printer.indd 121 03/29/09 2:38:00 PM


122 Chapter 6

• Oxcarbazepine: in a small randomized, open-label


study of oxcarbazepine 600 mg twice daily in patients
receiving an oxaliplatin-based regimen for colon can-
cer, the incidence of peripheral neuropathy was
reduced in patients receiving oxcarbazepine.28
■ Agents that may not be effective as prophylaxis include
• Amifostine for the prevention of platinum- or paclitaxel-
associated neuropathy: several randomized controlled
trials have demonstrated inconsistent results29
• Nimodipine for the prevention of cisplatin-associated
neuropathy30
• Org 2766 (an adrenocorticotrophic hormone ana-
logue) for the prevention of vincristine-31 or cisplatin-
associated32 neuropathy
• rhuLIF (a recombinant human leukemia inhibitory
factor) for the prevention of carboplatin/paclitaxel-
associated neuropathy33

Treatment
■ Treatments for diabetic neuropathies are not necessarily

helpful in CIPN.
■ Topical baclofen, amitriptyline, and ketamine (BAK):

preliminary results from a randomized, placebo-con-


trolled, double-blinded study of BAK in patients receiv-
ing a variety of neurotoxic agents suggest that BAK may
improve tingling and functioning in the hands.34
■ Other agents that have been tested in small randomized,

placebo-controlled trials but demonstrated no benefit in


improving neuropathic symptoms over placebo are11
• Tricyclic antidepressants, including amitriptyline and
nortriptyline
• Gabapentin
• Lamotrigine

Prognosis
■ Variable depending on the agent (may or may not be

reversible)

73724_CH06_Printer.indd 122 03/29/09 2:38:00 PM


Neurologic Complications of Chemotherapy 123

References
1. Wefel JS, Witgert ME, Meyers CA. Neuropsychological
sequelae of non-central nervous system cancer and cancer
therapy. Neuropsychol Rev. 2008;18(2):121–131.
2. Ahles TA, Saykin AJ. Candidate mechanisms for chemo-
therapy-induced cognitive changes. Nat Rev Cancer. 2007;
7(3):192–201.
3. Bhatt A, Farooq MU, Majid A, Kassab M. Chemotherapy-
related posterior reversible leukoencephalopathy syndrome.
Nat Clin Pract Neurol. 2009;5(3):163–169.
4. Baker WJ, Royer GL, Jr., Weiss RB. Cytarabine and neuro-
logic toxicity. J Clin Oncol. 1991;9(4):679–693.
5. Glantz MJ, Jaeckle KA, Chamberlain MC, et al. A random-
ized controlled trial comparing intrathecal sustained-release
cytarabine (DepoCyt) to intrathecal methotrexate in patients
with neoplastic meningitis from solid tumors. Clin Cancer
Res. 1999;5(11):3394–3402.
6. Pirzada NA, Ali, II, Dafer RM. Fluorouracil-induced neuro-
toxicity. Ann Pharmacother. 2000;34(1):35–38.
7. Sioka C, Kyritsis AP. Central and peripheral nervous sys-
tem toxicity of common chemotherapeutic agents. Cancer
Chemother Pharmacol. 2009;63(5):761–767.
8. Fleming RA. An overview of cyclophosphamide and ifosf-
amide pharmacology. Pharmacotherapy. 1997;17(5 Pt 2):
146S–154S.
9. Pande AR, Ando K, Ishikura R, et al. Disseminated necro-
tizing leukoencephalopathy following chemoradiation ther-
apy for acute lymphoblastic leukemia. Radiat Med. 2006;
24(7):515–519.
10. Matsubayashi J, Tsuchiya K, Matsunaga T, Mukai K.
Methotrexate-related leukoencephalopathy without radia-
tion therapy: distribution of brain lesions and pathological
heterogeneity on two autopsy cases. Neuropathology. 2009;
29(2):105–115.
11. Wolf S, Barton D, Kottschade L, Grothey A, Loprinzi C.
Chemotherapy-induced peripheral neuropathy: prevention and
treatment strategies. Eur J Cancer. 2008;44(11):1507–1515.
12. Hochster H, Grothey A, Shpilsky A, Childs B. Effect of
intravenous (IV) calcium and magnesium (Ca/Mg) versus
placebo on response to FOLFOX 1 bevacizumab (BEV)
in the CONcePT trial (abstract 280). Paper presented

73724_CH06_Printer.indd 123 03/29/09 2:38:00 PM


124 Chapter 6

at 2008 Gastrointestinal Cancers Symposium; January


25–27, 2008; Orlando, FL.
13. Grothey A, Hart LL, Rowland KM, et al. Intermittent oxali-
platin (oxali) administration and time-to-treatment-failure
(TTF) in metastatic colorectal cancer (mCRC): Final results
of the phase III CONcePT trial. J Clin Oncol (Meeting
Abstracts). 2008;26(Suppl):abstr 4010.
14. Grothey A, Nikcevich DA, Sloan JA, et al. Evaluation of the
effect of intravenous calcium and magnesium (CaMg) on
chronic and acute neurotoxicity associated with oxaliplatin:
Results from a placebo-controlled phase III trial. J Clin
Oncol (Meeting Abstracts). 2009;27(Suppl):abstr 4025.
15. Gamelin L, Boisdron-Celle M, Morel A, et al. Oxaliplatin-
related neurotoxicity: interest of calcium-magnesium
infusion and no impact on its efficacy. J Clin Oncol. 2008;
26(7):1188–1189; author reply 1189–1190.
16. Gamelin L, Boisdron-Celle M, Poirier A, Berger V, Morel A,
Gamelin E. Prevention of oxaliplatin-induced neurotoxicity
with Ca21/Mg 21 infusions: preliminary results of the
NEUROXA randomized trial in patients with colorectal can-
cer (CRC) receiving the FOLFOX4 regimen (abstract 602).
Gastrointest Cancer Res. 2007;1(6):259.
17. Pace A, Savarese A, Picardo M, et al. Neuroprotective effect
of vitamin E supplementation in patients treated with cispl-
atin chemotherapy. J Clin Oncol. 2003;21(5):927–931.
18. Argyriou AA, Chroni E, Koutras A, et al. Vitamin E for
prophylaxis against chemotherapy-induced neuropathy: a
randomized controlled trial. Neurology. 2005;64(1):26–31.
19. Argyriou AA, Chroni E, Koutras A, et al. A randomized con-
trolled trial evaluating the efficacy and safety of vitamin E
supplementation for protection against cisplatin-induced
peripheral neuropathy: final results. Support Care Cancer.
2006;14(11):1134–1140.
20. Pace A, Carpano S, Galie E, et al. Vitamin E in the
neuroprotection of cisplatin induced peripheral neurotoxicity
and ototoxicity. J Clin Oncol (Meeting Abstracts). 2007;
25(Suppl):abstr 9114.
21. Kottschade LA, Sloan JA, Mazurczak MA, et al. The use of
vitamin E for prevention of chemotherapy-induced periph-
eral neuropathy: a phase III double-blind, placebo controlled
study—N05C31. J Clin Oncol (Meeting Abstracts). 2009;
27(Suppl):abstr 9532.
22. Smyth JF, Bowman A, Perren T, et al. Glutathione reduces the
toxicity and improves quality of life of women diagnosed with

73724_CH06_Printer.indd 124 03/29/09 2:38:00 PM


Neurologic Complications of Chemotherapy 125

ovarian cancer treated with cisplatin: results of a double-blind,


randomised trial. Ann Oncol. 1997;8(6):569–573.
23. Cascinu S, Cordella L, Del Ferro E, Fronzoni M, Catalano G.
Neuroprotective effect of reduced glutathione on cisplatin-
based chemotherapy in advanced gastric cancer: a random-
ized double-blind placebo-controlled trial. J Clin Oncol.
1995;13(1):26–32.
24. Cascinu S, Catalano V, Cordella L, et al. Neuroprotective
effect of reduced glutathione on oxaliplatin-based chemo-
therapy in advanced colorectal cancer: a randomized,
double-blind, placebo-controlled trial. J Clin Oncol. 2002;
20(16):3478–3483.
25. Cassidy J, Bjarnason GA, Hickish T, et al. Randomized dou-
ble blind (DB) placebo (Plcb) controlled phase III study
assessing the efficacy of xaliproden (X) in reducing the cumu-
lative peripheral sensory neuropathy (PSN) induced by the
oxaliplatin (Ox) and 5-FU/LV combination (FOLFOX4) in
first-line treatment of patients (pts) with metastatic colorec-
tal cancer (MCRC). J Clin Oncol (Meeting Abstracts).
2006;24(Suppl):abstr 3507.
26. Loven D, Levavi H, Sabach G, et al. Long-term glutamate
supplementation failed to protect against peripheral neuro-
toxicity of paclitaxel. Eur J Cancer Care (Engl). 2009;18(1):
78–83.
27. Amara S. Oral glutamine for the prevention of chemothera-
py-induced peripheral neuropathy. Ann Pharmacother.
2008;42(10):1481–1485.
28. Argyriou AA, Chroni E, Polychronopoulos P, et al. Efficacy
of oxcarbazepine for prophylaxis against cumulative
oxaliplatin-induced neuropathy. Neurology. 2006;67(12):
2253–2255.
29. Hensley ML, Hagerty KL, Kewalramani T, et al. American
Society of Clinical Oncology 2008 clinical practice guide-
line update: use of chemotherapy and radiation therapy
protectants. J Clin Oncol. 2009;27(1):127–145.
30. Cassidy J, Paul J, Soukop M, et al. Clinical trials of nimo-
dipine as a potential neuroprotector in ovarian cancer
patients treated with cisplatin. Cancer Chemother Pharmacol.
1998;41(2):161–166.
31. Koeppen S, Verstappen CC, Korte R, et al. Lack of neuro-
protection by an ACTH (4–9) analogue: a randomized trial
in patients treated with vincristine for Hodgkin’s or non-
Hodgkin’s lymphoma. J Cancer Res Clin Oncol. 2004;
130(3):153–160.

73724_CH06_Printer.indd 125 03/29/09 2:38:00 PM


126 Chapter 6

32. Roberts JA, Jenison EL, Kim K, Clarke-Pearson D,


Langleben A. A randomized, multicenter, double-blind,
placebo-controlled, dose-finding study of ORG 2766 in the
prevention or delay of cisplatin-induced neuropathies in
women with ovarian cancer. Gynecol Oncol. 1997;67(2):
172–177.
33. Davis ID, Kiers L, MacGregor L, et al. A randomized,
double-blinded, placebo-controlled phase II trial of recom-
binant human leukemia inhibitory factor (rhuLIF, emfiler-
min, AM424) to prevent chemotherapy-induced peripheral
neuropathy. Clin Cancer Res. 2005;11(5):1890–1898.
34. Barton DL, Wos E, Qin R, et al. A randomized controlled
trial evaluating a topical treatment for chemotherapy-
induced neuropathy: NCCTG trial N06CA. J Clin Oncol
(Meeting Abstracts). 2009;27(Suppl):abstr 9531.

73724_CH06_Printer.indd 126 03/29/09 2:38:00 PM


C H A P T E R 7

Paraneoplastic
Disorders
Introduction
■ Paraneoplastic disorders (PND) are an extensive group of
syndromes that can affect any part of the nervous system
by mechanisms that are immune mediated.
■ Symptoms may develop prior to discovery of a tumor.
■ The diagnosis is complex.
• Symptoms can mimic other neurological complica-
tions of cancer or its treatments.
• Not all patients with PND have detectable or recog-
nized paraneoplastic antibodies.
• Not all patients with paraneoplastic antibodies have
PND, although titers are usually much higher in
patients with PND than those without PND.1,2
• There are patients in whom no tumor is ever detected
despite presenting with classic PND and high titers of
well-characterized onconeural antibody.3
■ Tumor treatment and immunotherapy may stabilize or
improve symptoms.

Epidemiology
■ The true prevalence is unknown.
• Based on serological screening of patients with sus-
pected PND (generally unselected population), 553 of
60,000 consecutive cases over 4 years (0.9%) were
positive for antibodies associated with PND.4
• Based on serological screening of preselected popula-
tion using clinical criteria, 163 of 649 consecutive cases
over 23 months (25%) were positive for antibodies
associated with PND.5
■ The incidence varies depending on type of cancer: SCLC
(3% develop a paraneoplastic disorder), thymoma (30%),

127

73724_CH07_Printer.indd 127 03/29/09 2:40:00 PM


128 Chapter 7

plasma cell dyscrasias associated with malignant mono-


clonal gammopathy (5% to 15%), and other solid tumors
(including breast cancer or ovarian cancer) (,1%).6
■ Central nervous system disorders are more frequently
found in the following tumors:
• Tumors that express neuroendocrine proteins (e.g.,
SCLC, neuroblastoma)
• Tumors that affect organs with immunoregulatory
properties (e.g., thymoma)
• Tumors that contain mature or immature neuronal tis-
sue (e.g., teratomas)
■ Peripheral nervous system disorders are more frequently
found in tumors that derive from cells that produce
immunoglobulins (e.g., plasma cell dyscrasias, B-cell
lymphomas).7

Pathogenesis
■ The pathogenesis is not entirely clear.
■ Most PNDs are probably caused by immunological
responses against intraneuronal antigens expressed by
the underlying cancer.
• Tumor cells express antigens found in only the ner-
vous system.8,9
• Tumor antigens are identified as foreign, resulting in
an immune response.
• The antitumor immune response cross-reacts with the
normal nervous system.

Antibody-Mediated PND
■ The direct pathogenic role of antibodies has been dem-

onstrated in a few disorders (i.e., development of charac-


teristic syndrome after passive transfer of antibodies in
mice or rats).
• Myasthenia gravis (MG): antibodies to the acetylcho-
line receptor of the neuromuscular junction
• Lambert-Eaton myasthenic syndrome (LEMS):
antibodies to voltage-gated calcium channels
• Stiff-person syndrome (SPS): antibodies to amphy-
physin10

73724_CH07_Printer.indd 128 03/29/09 2:40:00 PM


Paraneoplastic Disorders 129

• Single case report of two patients in remission from


Hodgkin lymphoma who developed cerebellar ataxia
found to have antibodies against mGluR111
■ Antibodies usually react with cell surface antigens, but
antibodies to intracellular antigens are also described
(e.g., antibodies against amphiphysin in paraneoplastic
stiff-person syndrome).
■ Associated disorders can occur with or without cancer.12
• The likelihood of an underlying tumor with LEMS is
50%, most commonly SCLC.13
• The likelihood of an underlying tumor with MG is
10%, most commonly thymoma.13
■ The presence of antibodies does not confer disease
because some paraneoplastic antibodies are commonly
present in cancer patients without a PND.12,14

T-Cell–Mediated PND
■ Some PNDs may be mediated by T-cell immune

responses against target antigens of accompanying anti-


bodies, although the evidence is mostly circumstantial.5
• Yo- or Hu-specific T-cells identified in blood or CSF
of patients with anti-Yo or anti-Hu antibodies
• Difficulty in treating these T-cell–mediated disorders
with strategies directed at humoral immune response
• Extensive T-cell infiltrates in CNS

Clinical Features
■ There is a rapid development of symptoms and signs of
inflammation in the CSF,5 including moderate lympho-
cytic pleocytosis (30–40 cells/mL), an elevated protein
concentration (50–100 mg/dL), a high IgG index, and
CSF-specific oligoclonal bands15
■ Neurologic symptoms are the first manifestation of tumor
in 70% of patients with PND; 70% to 80% of these
patients are found to have cancer on initial screening.5
■ Symptoms are progressive over weeks to months followed
by stabilization.
■ In 80% of patients with no known cancer history who
develop a PND, cancer is usually diagnosed within
months to years.16

73724_CH07_Printer.indd 129 03/29/09 2:40:00 PM


130 Chapter 7

Diagnosis
Diagnostic Criteria (Figure 7-1)17
Definite PND
■ Classic syndrome with cancer diagnosed within 5 years

of neurologic symptom development


■ Nonclassic syndrome that resolves or significantly

improves after cancer treatment without concomitant


immunotherapy, provided that the syndrome is not sus-
ceptible to spontaneous remission
■ Nonclassic syndrome with cancer diagnosed within

5 years of neurologic symptom development and positive


neuronal antibodies
■ Neurologic syndrome (classic or not) without cancer and

with well-characterized antineuronal antibodies (Hu, Yo,


CV2/CRMP5, Ri, Ma2, or amphiphysin)
Possible PND
■ Classic syndrome with high risk of cancer, without anti-

neuronal antibodies
■ Neurologic syndrome (classic or not) without cancer and

with partly characterized antineuronal antibodies

Figure 7-1: Paraneoplastic diagnosis flowchart. Graus F, et al.


Recommended diagnostic criteria for paraneoplastic neurological
syndromes. J Neurol Neurosurg Psychiatry. 2004;75(8):1135–1140.

73724_CH07_Printer.indd 130 03/29/09 2:40:00 PM


Paraneoplastic Disorders 131

■ Nonclassic syndrome with cancer diagnosed within


2 years of neurologic symptom development, without
neuronal antibodies

Diagnostic Workup18
■ If PND is suspected, the initial workup should include

MRI, neurophysiologic studies, CSF studies, including


paraneoplastic antibody testing, and serum paraneoplas-
tic antibody testing.
■ If paraneoplastic antibodies are present or clinical

suspicion is high, the workup should also include cancer


screening.
• In general, a full-body CT scan and an FDG-PET scan
are recommended.
• For breast cancer-associated syndromes, a careful
breast examination and mammography are also
recommended.
• For gynecologic-associated syndromes (i.e., anti-Yo–
associated paraneoplastic cerebellar degeneration), a
careful pelvic examination and pelvic ultrasound are
also recommended.
• For testicular-associated syndromes (i.e., anti-Ma2–
associated limbic encephalitis in young men), a
testicular ultrasound is also recommended.
• If an ovarian or testicular malignancy is strongly sus-
pected but not found on screening, some advocate
elective oophorectomy or orchidectomy.19
• For neuroblastoma-associated opsoclonus-myoclonus,
when conventional imaging is negative, metaiodoben-
zylguanidine whole-body scintigraphy may identify
occult cases.20
■ If a tumor is not initially detected, continued surveillance

at regular intervals is necessary.18

Common Paraneoplastic Disorders (Table 7-1)


Paraneoplastic Cerebellar Degeneration
■ Commonly associated tumors: SCLC, gynecologic

cancers, breast cancers, Hodgkin lymphoma5


■ Clinical features

73724_CH07_Printer.indd 131 03/29/09 2:40:00 PM


Table 7-1: Antibodies, Paraneoplastic Syndromes, and Associated Tumors

Antibody Syndrome Associated Tumors

73724_CH07_Printer.indd 132
Well-characterized paraneoplastic antibodies (identified by several investigators)
132 Chapter 7

Anti-Hu (ANNA-1) Paraneoplastic encephalomyelitis, including Small cell lung cancer


cortical, limbic, brainstem encephalitis Others
Paraneoplastic cerebellar degeneration
Myelitis
Paraneoplastic sensory neuronopathy
Autonomic dysfunction

Anti-Yo (PCA-1) Paraneoplastic cerebellar degeneration Gynecologic cancers


Breast cancer

Anti-Ri (ANNA-2) Paraneoplastic cerebellar degeneration Breast cancer


Brainstem encephalitis Gynecologic cancers
Opsoclonus-myoclonus Small cell lung cancer

(Continues)

03/29/09 2:40:00 PM
Table 7-1: Antibodies, Paraneoplastic Syndromes, and Associated Tumors (Continued)

Antibody Syndrome Associated Tumors

Anti-CV2/CRMP5 Paraneoplastic encephalomyelitis Small cell lung cancer

73724_CH07_Printer.indd 133
Paraneoplastic cerebellar degeneration Thymoma
Chorea Others
Uveitis
Optic neuritis
Peripheral neuropathy

Anti-Ma proteins Limbic, hypothalamic, brainstem encephalitis Germ cell tumors of testis
(Ma2, Ma1) Infrequently paraneoplastic cerebellar Nonsmall cell lung cancer
degeneration Other solid tumors

Antiamphiphysin Stiff-person syndrome Small cell lung cancer


Paraneoplastic encephalomyelitis Breast cancer
Limbic encephalitis
Myelopathy
Paraneoplastic Disorders 133

(Continues)

03/29/09 2:40:00 PM
Table 7-1: Antibodies, Paraneoplastic Syndromes, and Associated Tumors (Continued)

Antibody Syndrome Associated Tumors

Partly characterized paraneoplastic antibodies

73724_CH07_Printer.indd 134
Anti-Tr Paraneoplastic cerebellar degeneration Hodgkin lymphoma
134 Chapter 7

Anti-Zic 4 Paraneoplastic cerebellar degeneration Small cell lung cancer

mGluR1 Paraneoplastic cerebellar degeneration Hodgkin lymphoma

ANNA3 Various paraneoplastic disorders of the CNS Small cell lung cancer

PCA2 Various paraneoplastic disorders of the CNS Small cell lung cancer

Antibodies that occur with and without cancer association

Anti-NR1/NR2 of Characteristic encephalitis with prominent Teratoma (usually in the ovary)


NMDA receptor psychiatric symptoms, memory loss,
decreased consciousness with frequent
hypoventilation, autonomic instability,
dyskinesias

(Continues)

03/29/09 2:40:00 PM
Table 7-1: Antibodies, Paraneoplastic Syndromes, and Associated Tumors (Continued)

Antibody Syndrome Associated Tumors

Anti-VGKC Limbic encephalitis Thymoma

73724_CH07_Printer.indd 135
Peripheral nerve hyperexcitability Small cell lung cancer
(neuromyotonia) Others
Other

Anti-VGCC Lambert-Eaton myasthenic syndrome Small cell lung cancer


Paraneoplastic cerebellar degeneration

Anti-AChR Myasthenia gravis Thymoma

Anti-nAChR Subacute pandysautonomia Small cell lung cancer


Others

Anti-GAD Stiff-person syndrome Thymoma


Cerebellar ataxia Others
Limbic encephalitis
Others
Paraneoplastic Disorders 135

Modified from Dalmau J, Rosenfeld MR. Paraneoplastic syndromes of the CNS. Lancet Neurol. 2008;7(4):327–340.

03/29/09 2:40:00 PM
136 Chapter 7

• Gait unsteadiness rapidly progresses into truncal and


appendicular ataxia, nystagmus, diplopia, dysarthria,
dysphagia
• Occasionally prodromal symptoms (viral-like illness,
dizziness, nausea, or vomiting) before the onset of
neurologic signs that might be attributed to a vestibu-
lar process21
• Neurologic symptoms that begin abruptly, progress
over weeks to months, and then stabilize but usually
leave the patient significantly impaired
• Occasionally blurry vision, oscillopsia, and transient
opsoclonus5
• Anti-Yo, anti-Hu, anti-CRMP5 associated with more
severe neurologic deficits5
■ CSF studies: may show signs of inflammation with ele-
vated protein, lymphocytic pleocytosis, and oligoclonal
banding
■ Imaging
• Early stages
■ The MRI is initially normal in most patients,

although some may have transient diffuse cerebel-


lar hemispheric enlargement or cortical-meningeal
enhancement.22
■ A fluorodeoxyglucose PET may show cerebellar

hypermetabolism.23
• Later stages
■ An MRI shows cerebellar atrophy.

■ A PET shows hypometabolism.

■ Differential diagnosis includes


• Alcohol-related cerebellar degeneration
• Infectious or postinfectious cerebellitis
• Cerebellar metastasis
• Vitamin deficiency (thiamine, vitamin E)
• Toxins (antiepileptic agents, chemotherapy-related
agents such as fluorouracil or cytarabine)
• Creutzfeldt-Jakob disease (12% to 15% of patients
with CNS PND have 14-3-3 protein in CSF24)
• Glutamic acid decarboxylase (GAD)-associated cerebel-
lar degeneration: slower progression than paraneoplastic

73724_CH07_Printer.indd 136 03/29/09 2:40:00 PM


Paraneoplastic Disorders 137

cerebellar degeneration, milder and asymmetric limb


ataxia (mainly affecting gait), associated with endocrine
dysfunction (insulin-dependent diabetes, thyroiditis,
pernicious anemia),25 may be associated with transient
or self-limited muscle spasms26
■ Pathology: extensive loss of Purkinje cells
■ Serologic testing
• Gynecologic cancer or breast cancer is often associ-
ated with anti-Yo (cdr2).5
■ Anti-Yo has high syndrome specificity.

■ Patients with anti-Yo usually have a significant long-

term disability because of irreversible Purkinje cell


destruction.27,28
• Hodgkin lymphoma is often associated with anti-Tr.29
■ Anti-Tr has high syndrome specificity.

■ It is more commonly present in young men.

• SCLC is associated with one or multiple antibodies:


41% develop antibodies against voltage-gated calcium
channels with or without associated LEMS, 23%
develop anti-Hu, and the minority develop antibodies
against other targets (CRMP or CV2, amphiphysin,
PCA2, ANNA3).5
■ Treatment
• There is no standard of care.
• The treatment of the underlying tumor is necessary
for stabilization or symptom improvement.
• Immunotherapy (corticosteroids, plasma exchange, intra-
venous immunoglobulin, cyclophosphamide, tacrolimus)
does not substantially modify the neurologic outcome in
patients whose tumors are successfully treated.5
• There are case reports where immunotherapy
benefitted.30,31
■ Patients with anti-Yo who improved were treated

within 1 month of symptom onset and usually


received concurrent cancer therapy.31
■ Prognosis
• Antibodies associated with more severe neurological
deficits (Yo, Hu, CRMP5) are the most refractory to
treatment.5

73724_CH07_Printer.indd 137 03/29/09 2:40:00 PM


138 Chapter 7

• The median survival from time of PND diagnosis


(defined as first positive antineuronal antibody result)28
is as follows:
■ Anti-Yo 13 months

■ Anti-Hu 7 months

■ Anti-Tr .113 months

■ Anti-Ri .69 months

• Patients who received tumor treatment lived longer


than those who did not, regardless of whether they
also received immunotherapy or not.28

Paraneoplastic Opsoclonus-Myoclonus
■ Commonly associated tumors: SCLC, breast cancer,

ovarian cancer in adults; neuroblastoma in children


■ Clinical features

• Opsoclonus: involuntary, arrhythmic, chaotic, multidi-


rectional saccades with horizontal, vertical and tor-
sional components—may be constant, even during
sleep
• Myoclonic jerks involving limbs and trunk
• Cerebellar ataxia, most often in children
• Tremor
• Encephalopathy
■ CSF Studies: may show mild pleocytosis

■ Imaging: brain MRI may be normal

■ Differential diagnosis: opsoclonus-myoclonus associated

with infectious, toxic-metabolic disorders


■ Pathology: disinhibition of fastigial nucleus of cerebellum

may be involved32,33
■ Laboratory abnormalities

• Most patients are antibody negative.


• Anti-Ri
■ Found in a small subset of adults (usually with

breast or ovarian cancer) with paraneoplastic brain-


stem and cerebellar dysfunction34
■ Opsoclonus often present but not always35,36

• In neuroblastoma-associated opsoclonus-myoclonus,
no consistent specific autoantibodies have been
isolated, although antibodies to surface proteins in
cerebellar granular neurons have been reported.37,38

73724_CH07_Printer.indd 138 03/29/09 2:40:00 PM


Paraneoplastic Disorders 139

■ Treatment
• Pediatric cases
■ Opsoclonus often responds to immunotherapies

(corticosteroids, adrenocorticotropic hormone,


intravenous immunoglobulin, plasma exchange,
cyclophosphamide, rituximab).39
■ There is a high frequency of residual motor, speech,

behavioral, and sleep disorders.40


■ Symptoms can relapse during illnesses.

• Adult cases
■ There is improvement with tumor control.41

■ Improvement with immunotherapy is only mild or

not sustained unless the tumor is controlled.42


■ Prognosis
• In children, neurologic symptoms may resolve sponta-
neously or with treatment, but may relapse.43
• In adults, partial or complete neurologic recovery may
be seen with treatment of the underlying tumor.41

Paraneoplastic Limbic Encephalitis


■ Commonly associated tumors: SCLC, testicular germ-cell

neoplasms, thymoma, Hodgkin lymphoma, and ovarian


teratoma44
■ Clinical features

• Similar to nonparaneoplastic limbic encephalitis


• Mood disturbances
• Sleep disturbances
• Seizures
• Hallucinations
• Short-term memory loss that can progress to
dementia44
■ CSF studies: may show signs of inflammation with

elevated protein, lymphocytic pleocytosis, oligoclonal


banding
■ Imaging and other studies

• An EEG typically demonstrates foci of epileptic activ-


ity in one or both temporal lobes or focal or general-
ized slow activity.45
• A brain MRI typically demonstrates FLAIR or T2
hyperintensity in one or both mesial temporal lobes,44,45

73724_CH07_Printer.indd 139 03/29/09 2:40:00 PM


140 Chapter 7

although these areas rarely demonstrate gadolinium


enhancement.
• A PET may demonstrate hypermetabolism in one or
both temporal lobes that can precede MRI changes or
clinical symptoms.46
■ The differential diagnosis includes
• Viral encephalitis, including herpes simplex virus: the
course is more rapid and severe than paraneoplastic
limbic encephalitis
• Encephalitis associated with rheumatic disease such as
systemic lupus erythematous and Sjogren syndrome.
• Primary autoimmune limbic encephalitis: some may
be associated with antibodies to voltage-gated potas-
sium channels without an underlying tumor.47,48
■ Laboratory abnormalities
• Anti-Hu
■ Patients develop extensive or multifocal encephalo-

myelitis, which may begin as limbic encephalitis or


cerebellar degeneration.5
■ SCLC is the most commonly associated tumor, but

only 50% of patients with SCLC and limbic


encephalitis have anti-Hu antibodies.
• Anti-CRMP5 (anti-CV2)
■ The disease is rarely confined to the limbic system

because the antibody is also associated with enceph-


alomyelitis, sensorimotor neuropathy, cerebellar
ataxia, chorea, uveitis, and optic neuritis.5
■ SCLC and thymoma are the most commonly asso-

ciated tumors.
• Anti-Ma2 (Ta)
■ This typically affects the limbic system, hypothala-

mus, and brainstem, resulting in limbic dysfunction


as well as excessive daytime sleepiness, narcolepsy,
cataplexy, REM-sleep abnormalities, hyperphagia,
hypothalamic–pituitary hormone deficits, and
supranuclear gaze palsy.5
■ This may resemble CNS Whipple disease.49

■ The commonly associated tumors are as follows:

• In men less than 50 years old, anti-Ma2 enceph-


alitis is almost always associated with testicular

73724_CH07_Printer.indd 140 03/29/09 2:40:00 PM


Paraneoplastic Disorders 141

germ-cell tumors, which can be microscopic and


difficult to diagnose.50
• In older men and women, NCLC and breast
cancer are more common.5
• Anti-VGKC (voltage-gated potassium channels)
■ The two main CNS syndromes are as follows:

• Limbic encephalitis
• More diffuse encephalitis with psychiatric
symptoms, hallucinations, peripheral nerve
hyperexcitability, autonomic dysfunction (e.g.,
hyperhydrosis)5
■ REM sleep disturbances and hyponatremia are

common.51
■ Approximately 30% with anti-VGKC have tumors.

■ CSF shows less pleocytosis, lower protein concen-

tration, less intrathecal synthesis of IgG compared


with other paraneoplastic disorders, or immune-
mediated limbic encephalitis.5
■ Anti-SOX antibodies may help differentiate para-

neoplastic limbic encephalitis from the nonpara-


neoplastic form.
• Anti-NMDA receptors
■ This typically affects young women.

■ Patients first develop prodromal syndromes

(including headache, fever, or viral-like illness) and


then progress to develop severe psychiatric
symptoms or memory loss, seizures, decreased
consciousness associated with dyskinesias, hypoven-
tilation, or autonomic instability.5
■ Approximately 65% have an underlying tumor,

usually a cystic ovarian teratoma (mature or


immature).
■ Treatment
• For anti-Hu associated limbic encephalitis, early
treatment can result in substantial and prolonged
recovery.52
• For anti-CRMP5, treatment response is generally
poor.53
• For anti-Ma2–associated limbic encephalitis in men
with a testicular germ-cell tumor, treatment includes

73724_CH07_Printer.indd 141 03/29/09 2:40:00 PM


142 Chapter 7

orchiectomy and immunotherapy (corticosteroids and


intravenous immunoglobulin).
• For anti-VGKC–associated limbic encephalitis, treat-
ment includes corticosteroids, plasma exchange, or
intravenous immunoglobulin. About 80% respond to
treatment.5
• For patients with limbic encephalitis caused by ovar-
ian teratoma and antibodies against NMDA receptors,
an unstable clinical condition may hinder teratoma
removal.
■ Immunotherapy (corticosteroids, plasma exchange,

intravenous immunoglobulin, rituximab or cyclo-


phosphamide) often results in sufficient improve-
ment to enable tumor removal.54
■ Patients who do not respond to one form of immu-

notherapy may respond to another.


■ Improvement may be slow and take several

months.
■ Tumor removal speeds up recovery and decreases

the relapse rate.


■ Prognosis
• In patients with SCLC, the prognosis is worse in
patients without anti-Hu antibodies than patients with
anti-Hu antibodies.55
• For anti-Hu-associated limbic encephalitis, early
treatment can result in substantial and prolonged
recovery.52
• For anti-Ma2-associated limbic encephalitis, 35%
have neurologic responses to treatment.5
• For anti-VGKC-associated limbic encephalitis, the
prognosis is worse in patients with lung cancer than
those without lung cancer.56
• For patients with limbic encephalitis caused by ovar-
ian teratoma and antibodies against NMDA receptors,
65% have near or full recovery.57

Stiff-Person Syndrome
■ This syndrome may be idiopathic or paraneoplastic.

■ The commonly associated tumors are breast cancer,

SCLC, and Hodgkin lymphoma.

73724_CH07_Printer.indd 142 03/29/09 2:40:00 PM


Paraneoplastic Disorders 143

■ The nonparaneoplastic variant is also autoimmune, asso-


ciated with glutamic acid decarboxylase antibodies, and
usually affects patients with other autoimmune diseases,
especially diabetes mellitus type I.58
■ Clinical features are as follows:
• Gradual onset of stiffness and rigidity, initially in axial
muscles and progressing to proximal limb muscles,
primarily involving the legs
• EMG that demonstrates continuous motor unit activ-
ity in affected muscles
• Paraspinal muscle rigidity that may lead to lumbar
hyperlordosis, difficulty ambulating, and frequent falls
• Continuously contracting antagonist muscles that
cause a rock-hard or board-like sensation to palpation
• Sudden, painful muscle spasms usually precipitated
by touch or involuntary movement, sudden loud noise,
or emotional stress59
■ CSF studies are usually normal.60
■ MRI studies are usually normal.60
■ The differential diagnosis includes tetanus, hyperekplexia,
and myelopathy.
■ Laboratory abnormalities are as follows:
• Amphiphysin antibodies are most commonly detected
in the paraneoplastic form.
• There are reports of anti-GAD in paraneoplastic stiff-
person syndrome, but this antibody is most commonly
found in the nonparaneoplastic form.
■ Treatment
• Intravenous immunoglobulin proven effective in two
placebo-controlled studies in nonparaneoplastic SPS
and reported effective in paraneoplastic SPS in a few
case reports61
• Dramatic response to diazepam in relieving stiffness62

Paraneoplastic Visual Syndromes


Cancer-Associated Retinopathy (CAR)
■ CAR presents with progressive, painless visual loss, pho-

tosensitivity, peripheral and ring scotomata, and flickering,


light-induced glare.

73724_CH07_Printer.indd 143 03/29/09 2:40:00 PM


144 Chapter 7

■ An electroretinogram demonstrates evidence of cone-


and rod-mediated abnormalities.
■ A funduscopic examination may demonstrate arteriolar
narrowing.63
■ Antirecoverin is the most commonly associated antibody,
usually in patients with SCLC.64

Melanoma-Associated Retinopathy (MAR)


■ MAR presents as an abrupt onset night blindness and

flickering light phenomena with normal visual acuity.63


■ Symptoms generally develop long after a diagnosis of

melanoma, usually in the setting of tumor progression.


■ The exact target antigen is unknown.

Paraneoplastic Optic Neuropathy


■ This typically presents with unilateral painless vision loss

progressing to involve both eyes.63


■ It is usually associated with encephalomyelopathy.

■ Anti-CMRP5 is the most commonly associated antibody,

usually in patients with SCLC.65

Paraneoplastic Motor Neuron Disease


■ It is usually idiopathic.

■ There are rare case reports of association with solid

tumors and motor neuron disease.18


■ There is a slightly higher incidence of lymphoproliferative

disorders (including Hodgkin lymphoma, non-Hodgkin


lymphoma, Waldenström macroglobulinemia, multiple
myeloma, chronic lymphocytic leukemia) in patients with
motor neuron disease.18
■ For both solid tumors and lymphoproliferative disorders,

patients rarely recover despite treatment of the underly-


ing tumor/disorder.

Peripheral Neuropathy
■ Causes in cancer patients

• More commonly caused by toxic-metabolic disorders


(i.e., chemotherapy, diabetes, malnutrition)
• Paraneoplastic peripheral neuropathy (i.e., subacute
sensory neuronopathy)

73724_CH07_Printer.indd 144 03/29/09 2:40:00 PM


Paraneoplastic Disorders 145

• Leptomeningeal disease
• Direct tumor invasion
Subacute Sensory Neuronopathy 66
■ Classic paraneoplastic peripheral neuropathy
■ Commonly associated tumor: SCLC (70% to 80% of
cases67), Hodgkin lymphoma
■ Clinical features68
• Subacute onset and rapidly progressive, although an
indolent course has been reported69
• Asymmetric, multifocal pain and paresthesias, ini-
tially involving arms but progressing to include all
extremities
• Sensory ataxia and pseudoathetoid movements of the
hands
• Neuropathy isolated in only 24% of patients with
anti-Hu, others have various combinations of central
nervous system (e.g., encephalomyelitis, limbic enceph-
alitis) and peripheral nervous system involvement70
■ CSF studies: elevated protein, pleocytosis, oligoclonal
bands67
■ EMG/NCS: small or absent sensory nerve action
potentials18
■ Pathophysiology: destruction of dorsal root ganglia by
cytotoxic T-lymphocytes71
■ Laboratory testing: anti-Hu 99% specific and 82% sensi-
tive for cancer diagnosis in patients with subacute sensory
neuropathy66
■ Treatment
• Tumor therapy
• No clear benefit to immunosuppressive treatment
■ Prognosis
• Poor despite immunosuppressive treatment18
• Possible stabilization if tumor therapy initiated early
Paraneoplastic Sensory or Sensorimotor Neuropathy
■ Commonly associated tumors: SCLC or thymoma

■ Clinical features

• Often with CNS dysfunction (i.e., cerebellar ataxia,


limbic encephalitis, or ocular involvement)
• Predominates in lower limbs

73724_CH07_Printer.indd 145 03/29/09 2:40:00 PM


146 Chapter 7

■ EMG/NCS: axonal or mixed axonal/demyelinating


pattern
■ Laboratory testing: anti-CV2 (peripheral neuropathy
occurs in 57% of patients with anti-CV2 antibodies67)
Paraproteinemic Neuropathy
■ Not included in diagnostic criteria but often considered

paraneoplastic17
■ Commonly associated with plasma cell dyscrasias,

including
• Monoclonal gammopathy of undetermined signifi-
cance (MGUS): associated with a polyneuropathy that
may improve with treatment of MGUS72
• Multiple myeloma: neuropathy that may be associated
with amyloidosis (painful sensory or sensorimotor
neuropathy mainly affecting small fibers73) or type 1
cryoglobulinemia
• POEMS syndrome: polyneuropathy, organomegaly,
endocrinopathy, monoclonal protein, and skin
changes
■ Associated with osteosclerotic myeloma

■ Primarily demyelinating and sensorimotor neuropa-

thy similar to chronic inflammatory demyelinating


polyneuropathy (CIDP)74
• Waldenström macroglobulinemia (lymphoplasmacytic
lymphoma)
■ Monoclonal IgM antibodies against myelin-associ-

ated glycoprotein (anti-MAG) associated with


chronic, distal, symmetric, large and small fiber,
predominantly demyelinating polyneuropathy
■ Monoclonal IgM antibodies against sulfatide asso-

ciated with more profound symptoms, sensory or


sensorimotor neuropathy67
■ Monoclonal IgM antibodies against disialosyl gan-

gliosides associated with predominantly sensory


neuropathy and ophthalmoplegia75
■ Monoclonal IgM antibodies against GM1 associated

with multifocal motor neuropathy67

73724_CH07_Printer.indd 146 03/29/09 2:40:00 PM


Paraneoplastic Disorders 147

Autonomic Neuropathy
■ Rarely occurs in isolation, most commonly with limbic
encephalitis and subacute sensory neuronopathy in
patients with SCLC or with LEMS in patients with
voltage-gated calcium channel antibodies74
■ Clinical features

• Chronic gastrointestinal pseudo-obstruction caused


by destruction of autonomic neurons in myenteric
plexuses
• Orthostatic hypotension
• Bladder dysfunction
• Impotence
• Sudomotor alterations
• Absence of heart rate variability

Neuromyotonia
■ Idiopathic or paraneoplastic

■ Commonly associated tumors: thymoma, SCLC, rarely

Hodgkin lymphoma18
■ Clinical features

• Muscle twitching and myokymia (continuous,


undulating, ripple of muscles described as a bag of
worms)
• Stiffness
• Painful cramps worse with muscle contraction
• Hyperhidrosis
• Muscle hypertrophy
• Pseudomyotonia (slow relaxation of muscle after con-
traction without percussion myotonia)
■ EMG/NCS: spontaneous, continuous, high-frequency

(150–300 Hz) doublet, triplet or multiple single motor


unit discharges76
■ Laboratory abnormalities

• Antibodies to voltage-gated potassium channels


(VGKC) often present in autoimmune and paraneo-
plastic neuromyotonia
• Anti-Hu antibodies in SCLC reported77

73724_CH07_Printer.indd 147 03/29/09 2:40:00 PM


148 Chapter 7

■ Treatment
• Responds well to plasma exchange more than intrave-
nous immunoglobulin78
• Symptomatic treatment with anticonvulsants such as
phenytoin and carbamazepine
■ Prognosis: may remit after tumor treatment

Lambert-Eaton Myasthenic Syndrome


■ Idiopathic or paraneoplastic, underlying malignancy in
60% of cases79
■ Commonly associated tumor: SCLC (LEMS affects 3%

of patients with SCLC)80


■ Clinical features

• Proximal muscle weakness (usually legs worse than


arms)
• Generalized fatigue
• Depressed tendon reflexes
• Autonomic dysfunction
■ Dry mouth

■ Eye dryness, blurred vision

■ Impotence

■ Constipation

■ Impaired sweating

■ Orthostatic hypotension

• Improved strength and tendon reflexes with exercise


• Very rarely ocular and bulbar muscle involvement
• Respiratory muscle weakness as a late complication
but rare
■ EMG/NCS

• Decreased CMAP amplitudes with repetitive nerve


stimulation between 1 to 5 Hz
• More than 100% increment in CMAP amplitudes
after repetitive nerve stimulation at 20 Hz or greater or
after brief maximal effort
• Normal motor and sensory latencies and conduction
velocities
• Jitter on single-fiber EMG
■ Pathology: antibodies to presynaptic P/Q voltage-gated

calcium channels preventing calcium influx into the cell


and acetylcholine release

73724_CH07_Printer.indd 148 03/29/09 2:40:00 PM


Paraneoplastic Disorders 149

■ Laboratory abnormalities
• P/Q voltage-gated calcium channel (VGCC) antibodies
present in almost 100% of patients with cancer and
90% without cancer81
• Anti-SOX antibodies found in 65% of patients with
cancer-associated LEMS and may help differentiate
paraneoplastic from nonparaneoplastic LEMS82
■ Treatment
• Treatment of the underlying malignancy is important.
• Immunotherapy without treatment of underlying
tumor usually produces little or no improvement in
strength.83
• 3,4-Diaminopyridine improves strength regardless of
etiology.84
■ Prognosis: LEMS may remit with tumor therapy, good
response to treatment.

Myasthenia Gravis
■ Idiopathic or paraneoplastic (10% to 15%)18
■ Commonly associated tumor: thymoma (up to 40% of

patients with thymoma will develop MG85)


■ Clinical features

• Weakness: ocular and bulbar symptoms predominate,


may progress to generalized weakness
• Normal deep tendon reflexes
• No associated autonomic features
• Muscle fatigability, noticeable to patients later in the
day or with prolonged use, which may be elicited on
examination through repetitive confrontational
strength testing or sustained upgaze (extraocular
muscles)
■ EMG/NCS

• Greater than 10% CMAP amplitude decrement


after exercise or with repetitive nerve stimulation at
2 to 5 Hz76
• Jitter on single-fiber EMG
■ Pathology: antibodies against postsynaptic acetylcholine

receptor, resulting in activation of complement, acceler-


ated receptor degradation, or blocking of acetylcholine
receptor binding86

73724_CH07_Printer.indd 149 03/29/09 2:40:00 PM


150 Chapter 7

■ Laboratory abnormalities18
• Anti-acetylcholine receptor (AChR) antibodies are
found in 80% to 90% of patients with MG and almost
all patients with thymoma-associated MG.
• Anti-titin antibodies and anti-ryanodine antibodies are
also associated with thymoma-associated MG.
■ Treatment
• All patients with MG should have CT or MRI of the
anterior mediastinum to look for thymoma.
• A select population should have a thymectomy.
• Short-term symptomatic treatment should include
acetylcholinesterase inhibitors.
• Long-term symptomatic control may be achieved with
immunosuppression.
• Exacerbations or myasthenic crisis may be managed
with intravenous immunoglobulin and/or plasma
exchange.
■ Prognosis
• The presence of thymoma is a poor prognostic factor
in MG.
• Long-term neurologic outcome is similar to patients
with nonthymomatous MG with early thymectomy.87

Inflammatory Myopathies
Polymyositis
■ Idiopathic or paraneoplastic, increased risk of malignancy

■ Commonly associated tumors: non-Hodgkin lymphoma,

lung cancer, bladder cancer88


■ Clinical features: symmetrical proximal muscle

weakness
■ EMG/NCS: small polyphasic motor unit potentials and

abnormal spontaneous activity


■ Pathophysiology: T-cell mediated

■ Laboratory abnormalities

• Elevated creatine kinase


• Autoantibodies found in idiopathic polymyositis (anti-
Jo-1, anti-OJ, anti-Mi-2) less commonly found in
paraneoplastic polymyositis18

73724_CH07_Printer.indd 150 03/29/09 2:40:00 PM


Paraneoplastic Disorders 151

■ Treatment: same as idiopathic polymyositis. Immunosup-


pression with corticosteroids, intravenous immunoglobulin,
plasma exchange, immunomodulatory drugs (methotrexate,
azathioprine, cyclosporine, mycophenolate mofetil), TNF-a
inhibitors (infliximab, etanercept)
■ Prognosis: good response to treatment

Dermatomyositis
■ Idiopathic or paraneoplastic, increased risk of malignancy

(higher than polymyositis)


■ Commonly associated tumors: ovarian, lung, gastric, col-

orectal, pancreatic cancers, non-Hodgkin lymphoma88


■ Clinical features: symmetrical proximal muscle weakness

with characteristic skin changes (purplish heliotrope rash


of the eyelids, Grotton’s sign, photosensitive erythema-
tous rash of chest and shoulders)
■ EMG/NCS: small polyphasic motor unit potentials and

abnormal spontaneous activity


■ Pathophysiology: complement-mediated intramuscular

microangiopathy leading to ischemia, muscular fiber


necrosis, and perifascicular atrophy89
■ Laboratory abnormalities

• Creatine kinase may be elevated.


• Patients with interstitial lung disease may have
anti-Jo 1.68
■ Treatment: same as idiopathic polymyositis. Immunosup-

pression with corticosteroids, intravenous immunoglobu-


lin, plasma exchange, immunomodulatory drugs
(methotrexate, azathioprine, cyclosporine, mycopheno-
late mofetil), TNF-a inhibitors (infliximab, etanercept)
■ Prognosis: good response to treatment

Inclusion Body Myositis


■ Idiopathic or paraneoplastic

■ Clinical features: distal weakness

■ EMG/NCS: small polyphasic motor unit potentials and

abnormal spontaneous activity


■ Pathology: rimmed vacuoles, congophilic inclusions

73724_CH07_Printer.indd 151 03/29/09 2:40:00 PM


152 Chapter 7

■ Laboratory abnormalities: mildly elevated or normal


creatine kinase
■ Treatment: generally unresponsive to corticosteroids or
immunosuppressive drugs. Intravenous immunoglobulin
may be beneficial in a small number of cases.
■ Prognosis: generally resistant to therapies.

References
1. Dalmau J, Furneaux HM, Gralla RJ, Kris MG, Posner JB.
Detection of the anti-Hu antibody in the serum of patients
with small cell lung cancer: a quantitative western blot anal-
ysis. Ann Neurol. 1990;27(5):544–552.
2. Roberts WK, Darnell RB. Neuroimmunology of the parane-
oplastic neurological degenerations. Curr Opin Immunol.
2004;16(5):616–622.
3. Greenlee JE. Recommended diagnostic criteria for paraneo-
plastic neurological syndromes. J Neurol Neurosurg
Psychiatry. 2004;75(8):1090.
4. Pittock SJ, Kryzer TJ, Lennon VA. Paraneoplastic antibodies
coexist and predict cancer, not neurological syndrome. Ann
Neurol. 2004;56(5):715–719.
5. Dalmau J, Rosenfeld MR. Paraneoplastic syndromes of the
CNS. Lancet Neurol. 2008;7(4):327–340.
6. Bataller L, Dalmau J. Paraneoplastic disorders of the ner-
vous system. Continuum. 2005;11(5):69–92.
7. Viala K, Behin A, Maisonobe T, et al. Neuropathy in lym-
phoma: a relationship between the pattern of neuropathy,
type of lymphoma and prognosis? J Neurol Neurosurg
Psychiatry. 2008;79(7):778–782.
8. Dalmau J, Gultekin HS, Posner JB. Paraneoplastic neuro-
logic syndromes: pathogenesis and physiopathology. Brain
Pathol. 1999;9(2):275–284.
9. Darnell RB, Posner JB. Paraneoplastic syndromes affecting
the nervous system. Semin Oncol. 2006;33(3):270–298.
10. Sommer C, Weishaupt A, Brinkhoff J, et al. Paraneoplastic
stiff-person syndrome: passive transfer to rats by means of
IgG antibodies to amphiphysin. Lancet. 2005;365(9468):
1406–1411.

73724_CH07_Printer.indd 152 03/29/09 2:40:00 PM


Paraneoplastic Disorders 153

11. Sillevis Smitt P, Kinoshita A, De Leeuw B, et al. Paraneoplastic


cerebellar ataxia due to autoantibodies against a glutamate
receptor. N Engl J Med. 2000;342(1):21–27.
12. Buckley C, Vincent A. Autoimmune channelopathies. Nat
Clin Pract Neurol. 2005;1(1):22–33.
13. O’Neill JH, Murray NM, Newsom-Davis J. The Lambert-
Eaton myasthenic syndrome: a review of 50 cases. Brain.
1988;111(Pt 3):577–596.
14. Graus F, Lang B, Pozo-Rosich P, Saiz A, Casamitjana R,
Vincent A. P/Q type calcium-channel antibodies in paraneo-
plastic cerebellar degeneration with lung cancer. Neurology.
2002;59(5):764–766.
15. Darnell RB, Posner JB. Paraneoplastic syndromes involving
the nervous system. N Engl J Med. 2003;349(16):1543–1554.
16. Honnorat J, Antoine JC. Paraneoplastic neurological syn-
dromes. Orphanet J Rare Dis. 2007;2:22.
17. Graus F, Delattre JY, Antoine JC, et al. Recommended diag-
nostic criteria for paraneoplastic neurological syndromes.
J Neurol Neurosurg Psychiatry. 2004;75(8):1135–1140.
18. Toothaker TB, Rubin M. Paraneoplastic neurological syn-
dromes: a review. Neurologist. 2009;15(1):21–33.
19. Vedeler CA, Antoine JC, Giometto B, et al. Management of
paraneoplastic neurological syndromes: report of an EFNS
Task Force. Eur J Neurol. 2006;13(7):682–690.
20. Rutherford GC, Dineen RA, O’Connor A. Imaging in the
investigation of paraneoplastic syndromes. Clin Radiol.
2007;62(11):1021–1035.
21. Dalmau J, Gonzalez RG, Lerwill MF. Case records of the
Massachusetts General Hospital: Case 4-2007: a 56-year-
old woman with rapidly progressive vertigo and ataxia.
N Engl J Med. 2007;356(6):612–620.
22. de Andres C, Esquivel A, de Villoria JG, Graus F, Sanchez-
Ramon S. Unusual magnetic resonance imaging and
cerebrospinal fluid findings in paraneoplastic cerebellar
degeneration: a sequential study. J Neurol Neurosurg
Psychiatry. 2006;77(4):562–563.
23. Choi KD, Kim JS, Park SH, Kim YK, Kim SE, Smitt PS.
Cerebellar hypermetabolism in paraneoplastic cerebellar
degeneration. J Neurol Neurosurg Psychiatry. 2006;77(4):
525–528.

73724_CH07_Printer.indd 153 03/29/09 2:40:00 PM


154 Chapter 7

24. Saiz A, Graus F, Dalmau J, Pifarre A, Marin C, Tolosa E.


Detection of 14-3-3 brain protein in the cerebrospinal fluid
of patients with paraneoplastic neurological disorders. Ann
Neurol. 1999;46(5):774–777.
25. Honnorat J, Saiz A, Giometto B, et al. Cerebellar ataxia with
anti-glutamic acid decarboxylase antibodies: study of
14 patients. Arch Neurol. 2001;58(2):225–230.
26. Rakocevic G, Raju R, Semino-Mora C, Dalakas MC. Stiff
person syndrome with cerebellar disease and high-titer anti-
GAD antibodies. Neurology. 2006;67(6):1068–1070.
27. Peterson K, Rosenblum MK, Kotanides H, Posner JB.
Paraneoplastic cerebellar degeneration. I. A clinical analysis
of 55 anti-Yo antibody-positive patients. Neurology.
1992;42(10):1931–1937.
28. Shams’ili S, Grefkens J, de Leeuw B, et al. Paraneoplastic
cerebellar degeneration associated with antineuronal anti-
bodies: analysis of 50 patients. Brain. 2003;126(Pt 6):
1409–1418.
29. Phuphanich S, Brock C. Neurologic improvement after
high-dose intravenous immunoglobulin therapy in patients
with paraneoplastic cerebellar degeneration associated with
anti-Purkinje cell antibody. J Neurooncol. 2007;81(1):
67–69.
30. David YB, Warner E, Levitan M, Sutton DM, Malkin MG,
Dalmau JO. Autoimmune paraneoplastic cerebellar degen-
eration in ovarian carcinoma patients treated with plasma-
pheresis and immunoglobulin: a case report. Cancer.
1996;78(10):2153–2156.
31. Widdess-Walsh P, Tavee JO, Schuele S, Stevens GH.
Response to intravenous immunoglobulin in anti-Yo associ-
ated paraneoplastic cerebellar degeneration: case report and
review of the literature. J Neurooncol. 2003;63(2):187–190.
32. Helmchen C, Rambold H, Sprenger A, Erdmann C,
Binkofski F. Cerebellar activation in opsoclonus: an fMRI
study. Neurology. 2003;61(3):412–415.
33. Wong CY, Yu CB, Fan DS, Ng JS, Chow PC, Lam DS.
Topical anesthesia for strabismus surgery. J Pediatr
Ophthalmol Strabismus. 2001;38(2):61.
34. Luque FA, Furneaux HM, Ferziger R, et al. Anti-Ri: an anti-
body associated with paraneoplastic opsoclonus and breast
cancer. Ann Neurol. 1991;29(3):241–251.
35. Pittock SJ, Lucchinetti CF, Lennon VA. Anti-neuronal
nuclear autoantibody type 2: paraneoplastic accompani-
ments. Ann Neurol. 2003;53(5):580–587.

73724_CH07_Printer.indd 154 03/29/09 2:40:00 PM


Paraneoplastic Disorders 155

36. Sutton IJ, Barnett MH, Watson JD, Ell JJ, Dalmau J.
Paraneoplastic brainstem encephalitis and anti-Ri anti-
bodies. J Neurol. 2002;249(11):1597–1598.
37. Blaes F, Fuhlhuber V, Korfei M, et al. Surface-binding
autoantibodies to cerebellar neurons in opsoclonus syn-
drome. Ann Neurol. 2005;58(2):313–317.
38. Korfei M, Fuhlhuber V, Schmidt-Woll T, Kaps M, Preissner
KT, Blaes F. Functional characterisation of autoantibodies
from patients with pediatric opsoclonus-myoclonus-
syndrome. J Neuroimmunol. 2005;170(1–2):150–157.
39. Pranzatelli MR, Tate ED, Travelstead AL, et al. Rituximab
(anti-CD20) adjunctive therapy for opsoclonus-myoclonus
syndrome. J Pediatr Hematol Oncol. 2006;28(9):585–593.
40. Mitchell WG, Davalos-Gonzalez Y, Brumm VL, et al.
Opsoclonus-ataxia caused by childhood neuroblastoma:
developmental and neurologic sequelae. Pediatrics. 2002;
109(1):86–98.
41. Bataller L, Graus F, Saiz A, Vilchez JJ. Clinical outcome
in adult onset idiopathic or paraneoplastic opsoclonus-
myoclonus. Brain. 2001;124(Pt 2):437–443.
42. Erlich R, Morrison C, Kim B, Gilbert MR, Alrajab S. ANNA-2:
an antibody associated with paraneoplastic opsoclonus in a
patient with large-cell carcinoma of the lung with neuroendo-
crine features—correlation of clinical improvement with
tumor response. Cancer Invest. 2004;22(2):257–261.
43. Wong A. An update on opsoclonus. Curr Opin Neurol.
2007;20(1):25–31.
44. Gultekin SH, Rosenfeld MR, Voltz R, Eichen J, Posner JB,
Dalmau J. Paraneoplastic limbic encephalitis: neurological
symptoms, immunological findings and tumour association
in 50 patients. Brain. 2000;123(Pt 7):1481–1494.
45. Lawn ND, Westmoreland BF, Kiely MJ, Lennon VA,
Vernino S. Clinical, magnetic resonance imaging, and elec-
troencephalographic findings in paraneoplastic limbic
encephalitis. Mayo Clin Proc. 2003;78(11):1363–1368.
46. Scheid R, Lincke T, Voltz R, von Cramon DY, Sabri O.
Serial 18F-fluoro-2-deoxy-D-glucose positron emission
tomography and magnetic resonance imaging of paraneo-
plastic limbic encephalitis. Arch Neurol. 2004;61(11):
1785–1789.
47. Thieben MJ, Lennon VA, Boeve BF, Aksamit AJ, Keegan M,
Vernino S. Potentially reversible autoimmune limbic enceph-
alitis with neuronal potassium channel antibody. Neurology.
2004;62(7):1177–1182.

73724_CH07_Printer.indd 155 03/29/09 2:40:00 PM


156 Chapter 7

48. Vincent A, Buckley C, Schott JM, et al. Potassium channel


antibody-associated encephalopathy: a potentially immuno-
therapy-responsive form of limbic encephalitis. Brain.
2004;127(Pt 3):701–712.
49. Castle J, Sakonju A, Dalmau J, Newman-Toker DE. Anti-
Ma2-associated encephalitis with normal FDG-PET: a case
of pseudo-Whipple’s disease. Nat Clin Pract Neurol.
2006;2(10):566–572; quiz 573.
50. Mathew RM, Vandenberghe R, Garcia-Merino A, et al.
Orchiectomy for suspected microscopic tumor in patients
with anti-Ma2-associated encephalitis. Neurology. 2007;
68(12):900–905.
51. Iranzo A, Molinuevo JL, Santamaria J, et al. Rapid-eye-
movement sleep behaviour disorder as an early marker for a
neurodegenerative disorder: a descriptive study. Lancet
Neurol. 2006;5(7):572–577.
52. Fadul CE, Stommel EW, Dragnev KH, Eskey CJ, Dalmau JO.
Focal paraneoplastic limbic encephalitis presenting as
orgasmic epilepsy. J Neurooncol. 2005;72(2):195–198.
53. Tuzun E, Dalmau J. Limbic encephalitis and variants: clas-
sification, diagnosis and treatment. Neurologist. 2007;
13(5):261–271.
54. Shimazaki H, Ando Y, Nakano I, Dalmau J. Reversible lim-
bic encephalitis with antibodies against the membranes of
neurones of the hippocampus. J Neurol Neurosurg Psychiatry.
2007;78(3):324–325.
55. Alamowitch S, Graus F, Uchuya M, Rene R, Bescansa E,
Delattre JY. Limbic encephalitis and small cell lung cancer:
clinical and immunological features. Brain. 1997; 120
(Pt 6):923–928.
56. Pozo-Rosich P, Clover L, Saiz A, Vincent A, Graus F. Voltage-
gated potassium channel antibodies in limbic encephalitis.
Ann Neurol. 2003;54(4):530–533.
57. Novillo-Lopez ME, Rossi JE, Dalmau J, Masjuan J.
Treatment-responsive subacute limbic encephalitis and
NMDA receptor antibodies in a man. Neurology. 2008;
70(9):728–729.
58. Solimena M, Folli F, Denis-Donini S, et al. Autoantibodies
to glutamic acid decarboxylase in a patient with stiff-man
syndrome, epilepsy, and type I diabetes mellitus. N Engl
J Med. 1988;318(16):1012–1020.

73724_CH07_Printer.indd 156 03/29/09 2:40:00 PM


Paraneoplastic Disorders 157

59. Lockman J, Burns TM. Stiff-person syndrome. Curr Treat


Options Neurol. 2007;9(3):234–240.
60. Murinson BB. Stiff-person syndrome. Neurologist. 2004;
10(3):131–137.
61. Pittock SJ, Lucchinetti CF, Parisi JE, et al. Amphiphysin
autoimmunity: paraneoplastic accompaniments. Ann
Neurol. 2005;58(1):96–107.
62. Lorish TR, Thorsteinsson G, Howard FM, Jr. Stiff-man syn-
drome updated. Mayo Clin Proc. 1989;64(6):629–636.
63. Damek DM. Paraneoplastic retinopathy/optic neuropathy.
Curr Treat Options Neurol. 2005;7(1):57–67.
64. Bataller L, Dalmau J. Neuro-ophthalmology and paraneo-
plastic syndromes. Curr Opin Neurol. 2004;17(1):3–8.
65. Cross SA, Salomao DR, Parisi JE, et al. Paraneoplastic auto-
immune optic neuritis with retinitis defined by CRMP-5-
IgG. Ann Neurol. 2003;54(1):38–50.
66. Molinuevo JL, Graus F, Serrano C, Rene R, Guerrero A,
Illa I. Utility of anti-Hu antibodies in the diagnosis of para-
neoplastic sensory neuropathy. Ann Neurol. 1998;44(6):
976–980.
67. Antoine JC, Camdessanche JP. Peripheral nervous system
involvement in patients with cancer. Lancet Neurol.
2007;6(1):75–86.
68. Rudnicki SA, Dalmau J. Paraneoplastic syndromes of the
peripheral nerves. Curr Opin Neurol. 2005;18(5):598–603.
69. Graus F, Bonaventura I, Uchuya M, et al. Indolent anti-Hu-
associated paraneoplastic sensory neuropathy. Neurology.
1994;44(12):2258–2261.
70. Graus F, Keime-Guibert F, Rene R, et al. Anti-Hu-associated
paraneoplastic encephalomyelitis: analysis of 200 patients.
Brain. 2001;124(Pt 6):1138–1148.
71. Kuntzer T, Antoine JC, Steck AJ. Clinical features and
pathophysiological basis of sensory neuronopathies (gangli-
onopathies). Muscle Nerve. 2004;30(3):255–268.
72. Eurelings M, Lokhorst HM, Kalmijn S, Wokke JH,
Notermans NC. Malignant transformation in polyneuropa-
thy associated with monoclonal gammopathy. Neurology.
2005;64(12):2079–2084.
73. Kelly JJ, Jr., Kyle RA, O’Brien PC, Dyck PJ. The natural
history of peripheral neuropathy in primary systemic amyloi-
dosis. Ann Neurol. 1979;6(1):1–7.

73724_CH07_Printer.indd 157 03/29/09 2:40:00 PM


158 Chapter 7

74. Dispenzieri A, Kyle RA, Lacy MQ, et al. POEMS syndrome:


definitions and long-term outcome. Blood. 2003;101(7):
2496–2506.
75. Kobayashi M, Kato K, Funakoshi K, Watanabe S, Toyoshima I.
Neuropathology of paraneoplastic neuropathy with anti-disia-
losyl antibody. Muscle Nerve. 2005;32(2):216–222.
76. American Association of Electrodiagnostic Medicine Quality
Assurance Committee. Practice parameter for repetitive
nerve stimulation and single fiber EMG evaluation of adults
with suspected myasthenia gravis or Lambert-Eaton myas-
thenic syndrome: summary statement. Muscle Nerve.
2001;24(9):1236–1238.
77. Toepfer M, Schroeder M, Unger JW, Lochmuller H,
Pongratz D, Muller-Felber W. Neuromyotonia, myocloni,
sensory neuropathy and cerebellar symptoms in a patient
with antibodies to neuronal nucleoproteins (anti-Hu-
antibodies). Clin Neurol Neurosurg. 1999;101(3):207–209.
78. van den Berg JS, van Engelen BG, Boerman RH, de Baets
MH. Acquired neuromyotonia: superiority of plasma
exchange over high-dose intravenous human immunoglobu-
lin. J Neurol. 1999;246(7):623–625.
79. Wirtz PW, Smallegange TM, Wintzen AR, Verschuuren JJ.
Differences in clinical features between the Lambert-Eaton
myasthenic syndrome with and without cancer: an analysis
of 227 published cases. Clin Neurol Neurosurg. 2002;
104(4):359–363.
80. Elrington GM, Murray NM, Spiro SG, Newsom-Davis J.
Neurological paraneoplastic syndromes in patients with
small cell lung cancer: a prospective survey of 150 patients.
J Neurol Neurosurg Psychiatry. 1991;54(9):764–767.
81. Lennon VA. Serologic profile of myasthenia gravis and dis-
tinction from the Lambert-Eaton myasthenic syndrome.
Neurology. 1997;48(Suppl 5):S23–S27.
82. Sabater L, Titulaer M, Saiz A, Verschuuren J, Gure AO,
Graus F. SOX1 antibodies are markers of paraneoplastic
Lambert-Eaton myasthenic syndrome. Neurology. 2008;
70(12):924–928.
83. Sanders DB. Lambert-Eaton myasthenic syndrome: diagno-
sis and treatment. Ann N Y Acad Sci. 2003;998:500–508.

73724_CH07_Printer.indd 158 03/29/09 2:40:00 PM


Paraneoplastic Disorders 159

84. Sanders DB, Massey JM, Sanders LL, Edwards LJ.


A randomized trial of 3,4-diaminopyridine in Lambert-Eaton
myasthenic syndrome. Neurology. 2000;54(3):603–607.
85. Vernino S, Lennon VA. Autoantibody profiles and neurologi-
cal correlations of thymoma. Clin Cancer Res. 2004;10(21):
7270–7275.
86. Conti-Fine BM, Milani M, Kaminski HJ. Myasthenia gravis:
past, present, and future. J Clin Invest. 2006;116(11):
2843–2854.
87. Kim HK, Park MS, Choi YS, et al. Neurologic outcomes of
thymectomy in myasthenia gravis: comparative analysis of
the effect of thymoma. J Thorac Cardiovasc Surg. 2007;
134(3):601–607.
88. Hill CL, Zhang Y, Sigurgeirsson B, et al. Frequency of
specific cancer types in dermatomyositis and polymyositis:
a population-based study. Lancet. 2001;357(9250):96–100.
89. Dalakas MC. Advances in the immunobiology and treat-
ment of inflammatory myopathies. Curr Rheumatol Rep.
2007;9(4):291–297.

73724_CH07_Printer.indd 159 03/29/09 2:40:00 PM


73724_CH07_Printer.indd 160 03/29/09 2:40:00 PM
C H A P T E R 8

Cerebrovascular
Complications
Introduction
■ Conflicting data exist between autopsy and clinical series
on whether cerebrovascular disease is elevated in cancer
patients.
• Based on a large autopsy series, cerebrovascular com-
plications, including hemorrhages and infarctions,
were the second most common CNS lesion in patients
with cancer.1
• Based on large modern clinical series, the frequency
of ischemic stroke was not elevated in cancer patients.2,3
• The risk of cerebral hemorrhages appears to be ele-
vated in hematological malignancies.4
■ Although some of the causes of cerebrovascular disease
in cancer patients may be similar to noncancer patients,
there are several cancer-specific and treatment-related
causes to be considered.

Ischemic Strokes
Risk Factors in Cancer Patients
■ Similar risk factors as the noncancer population, includ-

ing hypertension, diabetes mellitus, hyperlipidemia, atrial


fibrillation, smoking, and carotid artery disease
■ Additional risk factors in cancer patients that are treat-

ment and/or disease related discussed later in this chapter

Management of Ischemic Strokes in Cancer Patients


■ Guidelines for stroke management in cancer patients

should generally follow recommendations for noncancer


patients with special consideration of cancer-specific
causes of stroke.

161

73724_CH08_Printer.indd 161 03/29/09 2:40:00 PM


162 Chapter 8

• The American Stroke Association and American Heart


Association have published guidelines for early man-
agement of adults with ischemic stroke.5
• The American Stroke Association and American Heart
Association have published guidelines for secondary
prevention of ischemic stroke.6
■ Patients with acute stroke symptoms should be urgently
referred to the hospital for early management, including
diagnosis, imaging, treatment of acute complications,
and consideration of thrombolysis.
■ After emergency department management, additional
management may include the following:
• Risk factor management includes hypertension, hyper-
lipidemia, and diabetes mellitus.
• Noninvasive vessel imaging should be performed to
look for vascular stenoses, dissections, and so forth.
■ Carotid duplex ultrasound (although does not pro-

vide information about intracranial vasculature)


■ CT angiogram

■ MR angiogram

• A cardiac ultrasound should be performed to look for


valvular abnormalities, left ventricular thrombus, and
so forth.
■ A transthoracic echocardiogram may be sufficient,

but if the suspicion for endocarditis is high, then a


transesophageal echocardiogram may be more sen-
sitive in detecting valvular vegetations.
■ If strokes appear embolic in origin and there is no

left-sided source, then consider performing a bub-


ble study with the cardiac ultrasound to look for a
cardiac shunt.
• If strokes appear embolic in origin and the patient has
a patent foramen ovale, then venous studies should be
considered, including lower-extremity Doppler ultra-
sound and pelvic MR venography (as pelvic veins may
be an important source of paradoxical emboli).

73724_CH08_Printer.indd 162 03/29/09 2:40:00 PM


Cerebrovascular Complications 163

Treatment Considerations in Cancer Patients


■ The American Stroke Association and American Heart
Association have published guidelines for secondary pre-
vention of stroke.6
■ Additional considerations regarding thrombolysis in cancer

patients include the following:


• Further investigations are needed to determine the
role of thrombolysis in cancer patients, as large stud-
ies of recombinant tissue plasminogen activator (rTPA)
excluded patients with malignancy.
• Case series have reported the use of thrombolysis in
cancer patients.3
• Neither brain tumor nor cancer is an established
exclusion criteria for thrombolysis.
• Established exclusion criteria that may apply to cancer
patients include thrombocytopenia (platelet count
,100,000), major surgery within 14 days, and a prior
history of intracranial hemorrhage.5

Embolism
■ Signs and symptoms may not be limited to the CNS, as
emboli may spread to multiple organs or extremities.
■ The most common source of emboli in the general popu-
lation is the heart (usually caused by atrial fibrillation).
■ Other cancer-related causes of embolism are discussed
later in this chapter.

Tumor Embolism
■ Rare cause of stroke in cancer patients

■ Common sources of tumor emboli include

• Atrial myxomas and other cardiac tumors


• Lung cancer: most often occurs within 48 hours of sur-
gical manipulation of lung tissue but may also result
form tumor invasion of pulmonary veins or the left
atrium of the heart7

73724_CH08_Printer.indd 163 03/29/09 2:40:00 PM


164 Chapter 8

Nonbacterial Thrombotic Endocarditis (NBTE)


■ Formerly known as marantic endocarditis
■ Defined as sterile vegetations on cardiac valves in the

absence of bacteremia

Clinical Features
■ Cases have been reported in several malignancies, includ-

ing lung (especially adenocarcinoma), pancreatic, gastric,


breast, prostate, colorectal, ovarian, and thyroid.8
■ Although more common in cancer patients, NBTE

may also occur in patients with nonneoplastic hyperco-


agulable disorders such as antiphospholipid antibody
syndrome.
■ NBTE may occur in association with disseminated intra-

vascular coagulopathy (DIC).


■ Systemic emboli occur in nearly 50% of patients with

NBTE.8
■ Presenting signs and symptoms may depend on the site(s)

of embolization.
• Central nervous system infarcts may present as focal
or diffuse neurologic symptoms depending on the loca-
tion and number of emboli.
• Renal infarcts may present as hematuria.
• Splenic infarcts may present as left upper quadrant
pain.
• Peripheral artery emboli may present as a cold, cyan-
otic, or pulseless limb.
■ Cardiac murmurs are infrequently noted.

Characteristics of Vegetations8
■ The most commonly affected valves are the aortic and

mitral valves.
■ Vegetations generally do not alter or impede valve

function.
■ Vegetations contain degenerating platelets interwoven

with strands of fibrin.


■ Vegetations observed in NBTE tend to detach and

cause extensive infarction more readily than vegetations


observed in infective endocarditis.

73724_CH08_Printer.indd 164 03/29/09 2:40:00 PM


Cerebrovascular Complications 165

Diagnosis
■ An MRI is more sensitive and specific for diagnosing
embolic strokes acutely than CT.
■ A two-dimensional echocardiogram is useful, but a

transesophageal echocardiogram may be more sensitive


in detecting valvular vegetations than a transthoracic
echocardiogram.
■ Blood cultures are negative in NBTE.

Treatment
■ Optimal treatment is not well established.
■ Anticoagulation has been reported to prevent recurrent

embolism.8
• Several reports show successful management with
unfractionated heparin.
• Low molecular weight heparin may also be effective,
although the experience is limited.
• Recurrent thromboembolic events may occur on war-
farin and therefore warfarin is not recommended.
■ Anticoagulation may need to be continued indefinitely.

■ Cardiac surgery may be indicated for severe valvular dys-

function and recurrent emboli despite anticoagulation.9

Septic Embolism
■ Immunocompromised patients and leukemic patients are

susceptible to septic infection, infectious endocarditis,


and infectious vasculitis.
■ Causes include Aspergillus and Candida.

■ Infarctions may be multiple and/or hemorrhagic and may

evolve into cerebral abscesses.

Paradoxical Embolism
■ Embolism of venous origin through cardiac shunt (e.g.,
patent foramen ovale [PFO], atrial septal defect [ASD],
ventricular septal defect [VSD])
■ Strokes in patients with PFOs cannot be assumed due to

paradoxical embolism, as PFOs are common (detected in


20% to 35% of the general population at autopsy10).

73724_CH08_Printer.indd 165 03/29/09 2:40:00 PM


166 Chapter 8

■ Diagnostic criteria for the diagnosis of paradoxical


embolism11
• Cerebral or systemic embolism without a left-sided
source
• Presence of a venous thrombus or pulmonary embolism
• Demonstration of a right-to-left shunt
• Elevated right heart pressure, either constant eleva-
tion (e.g., pulmonary hypertension) or transient eleva-
tion (e.g., cough, Valsalva maneuver)

Thrombotic Disease (Including Small, Medium,


and Large Vessel Disease)
Postradiation Vasculopathy
■ Can affect intracranial and extracranial vessels

■ Typically involves medium and large vessels

Internal Carotid Artery (ICA) Stenosis


■ The frequency of ICA stenosis after external neck radia-
tion ranges from 12% to 60%.12
■ In a meta-analysis based on case series and retrospective

studies, the odds of a cerebrovascular event after neck


irradiation were 9.0 compared with nonirradiated
patients.13
■ Neck irradiation is associated with intimal damage with

mural thrombosis formation in 5 years, fibrotic occlusion


in 10 years, and atheroma formation with periarterial
fibrosis in 20 years or more.14
■ Other vascular pathologies after radiation have been

described, including vascular malformations, aneurysm


formation, and accelerated atherosclerosis.15

Postoperative Complications
■ Neurosurgical patients with brain tumors have an

increased risk of thromboembolic disease.16,17


■ Adjacent ischemic lesions are found on MRI in about

70% of high-grade glioma patients after craniotomy.18

73724_CH08_Printer.indd 166 03/29/09 2:40:00 PM


Cerebrovascular Complications 167

Intravascular Lymphoma
■ Also known as angiotropic large-cell lymphoma or malig-
nant angioendotheliomatosis
■ Rare type of non-Hodgkin lymphoma, typically a large

B-cell lymphoma, characterized by selective growth of


neoplastic cells within the lumina of small- and medium-
sized blood vessels

Clinical Features
■ Typically disseminated, although clinical presentations

often different depending on geographical location of


patient
■ In Western countries, propensity to manifest more fre-

quently in the central nervous system and skin19


• The central nervous system is affected in almost two
thirds of cases in Western patients with IVL.20
• Neurologic manifestations depend on location of
involvement but may include multifocal cerebrovascu-
lar events, dementia, subacute encephalopathy, sei-
zures, and myelopathy.
• Neurologic signs are caused by obliteration of vessels
and may transiently respond to corticosteroid
treatment.
■ In Asian countries, often presents as a hemophagocytic

syndrome (bone marrow involvement, fever, hepatosple-


nomegaly, thrombocytopenia)21

Diagnosis
■ Suggested workup based on international consensus
meeting22
• Physical examination with emphasis on nervous
system and skin (suspicious skin lesions should be
referred for biopsy)
• Routine blood studies, including hepatic, pulmonary,
renal, and thyroid function tests (with any abnormal
results further investigated by imaging or biopsy)
• Peripheral blood smear

73724_CH08_Printer.indd 167 03/29/09 2:40:00 PM


168 Chapter 8

• Body CT with contrast


• Brain MRI with contrast
• Lumbar puncture for CSF examination and cytology
• Bone marrow biopsy
• Possibly PET study
■ Laboratory findings are not specific and may include
anemia, increased serum lactate dehydrogenase, and
increased b2-microglobulin levels.
■ CSF may demonstrate elevated protein and pleocytosis,
with lymphona cells ancedotally reported in cases.
■ Neuroimaging and body CT are associated with a rela-
tively high proportion of false negatives.19
■ A brain MRI may demonstrate multiple infarct-like
lesions in the white matter, focal parenchymal enhance-
ment, dural and arachnoid enhancement, and vasculitis-
like lesions.23

Treatment
■ Limited data on best treatment options, although inter-
national consensus guidelines have been published22
■ Anthracycline-based chemotherapy recommended by

consensus panel, although a more intensive regimen with


greater CNS penetration is needed in patients with CNS
involvement

Prognosis
■ Commonly fatal disease characterized by an aggressive
course and short outcome with few long-term survivors
reported19
■ Aggressive chemotherapy early in the disease process

increases survival24 and therefore early diagnosis important

Hyperviscous Obstruction
■ Hyperviscosity may be caused by an increase in serum

proteins or an increase in cells.


■ Symptoms from hyperviscosity usually occur at a serum

viscosity greater than 5 centipoises (normal serum viscosity


is between 1.4 and 1.8 centipoises).25
■ Symptoms of hyperviscosity include

73724_CH08_Printer.indd 168 03/29/09 2:40:00 PM


Cerebrovascular Complications 169

• Spontaneous bleeding from mucous membranes


• Visual disturbances caused by retinopathy
• Neurologic symptoms, including headaches, vertigo,
seizures, altered consciousnesses, and cerebral
infarction
■ Cerebral infarction caused by hyperviscous obstruction is
rarely seen with polycythemia vera, acute myelogenous
leukemia (cell count .100,000 mm3), chronic lymphatic
leukemia (cell count .250,000 mm3), and multiple
myeloma.4

Intracranial Hemorrhages
Types of Intracranial Hemorrhages
Intraparenchymal Hemorrhage
■ This occurs more frequently in hematological malignancies,

especially acute myelogenous leukemia, and in patients


with coagulation disorders.4
■ This may occur as a result of venous infarction (see

cerebral venous thrombosis section later in this chapter).


■ Other causes include hypertension and cerebral amyloid

angiopathy.

Intratumoral Hemorrhage
■ Hemorrhage can be the presenting sign of a brain mass.

• Can obscure the tumor’s appearance on imaging


studies
• Neuroimaging clues to an underlying tumor include
■ More perifocal edema than expected for the

hemorrhage
■ Hemorrhage in unusual locations (e.g., close to the

subarachnoid space)
■ Pattern of contrast enhancement not expected for

the age of the hemorrhage


■ CNS metastases

• The risk of bleeding into CNS metastases varies accor-


ding to histology.
• Brain metastases from lung cancer, especially broncho-
genic carcinoma, are the most common hemorrhagic

73724_CH08_Printer.indd 169 03/29/09 2:40:00 PM


170 Chapter 8

lesions because they occur in greater numbers;


however, fewer than 1% of brain metastases are from
lung cancer bleed.26
• The risk of bleeding into CNS metastases is high in
thyroid cancer, melanoma (40% to 50%), renal cell
carcinoma (70%), and choriocarcinoma.26
■ Primary brain tumors
• The reported rates in primary brain tumors range from
0.6% to 14.6% with the majority of cases involving
high-grade astrocytomas and oligodendrogliomas.27
• Intracranial hemorrhage rates in patients with menin-
giomas range from 1.3% to 2.4%28,29 and may present as
subarachnoid, intratumoral, or subdural hemorrhage.

Intraventricular Hemorrhage
■ Rare in cancer patients
■ May be seen in primary brain tumors in or near the ven-

tricular space

Subarachnoid Hemorrhage
■ Neoplastic aneurysms (discussed further later in this

chapter)
■ Infectious aneurysms caused by fungal (e.g., Aspergillus,

Candida) or bacterial infections


■ Other causes include trauma, coagulation disorders

Subdural Hemorrhage
■ Tumors with propensity to invade or metastasize to the
subdural space may include leukemia, lymphoma, pros-
tate cancer, and breast cancer.
■ MRI with contrast may demonstrate thickened dura and

subdural hemorrhage.
■ Other causes include trauma, coagulation disorders, and

thrombocytopenia.

Epidural Hemorrhage
■ May occur in association with a primary brain tumor or
skull metastasis

73724_CH08_Printer.indd 170 03/29/09 2:40:00 PM


Cerebrovascular Complications 171

Clinical Features
■ Presentation depends on the size and location of hemor-
rhage and is similar to hemorrhages from noncancer
causes.
■ Presenting signs and symptoms may include acute onset

neurologic deficits, headache, and altered consciousness.

Diagnosis
■ A noncontrast head CT is the first-line diagnostic

approach.
■ MRI with gradient echo can also detect hyperacute

hemorrhages and may be more accurate for detecting


microhemorrhages.30
■ Angiography (CT or MR) may help identify aneurysms,

arteriovenous malformations, and vasculitis.

Management
■ This is based on management principles of intracranial

hemorrhages in the noncancer population.


■ Urgent referral to the hospital is recommended for diag-

nosis by imaging, correction of any potentially reversible


causes of hemorrhage (e.g., thrombocytopenia, antico-
agulation), and management of any adverse events (e.g.,
neurologic deterioration, cardiovascular instability).

Cancer-Related Causes of Intracranial Hemorrhage


Coagulation Disorders
■ This may result in intraparenchymal or subdural hemor-

rhages.
■ Examples of coagulopathies seen in cancer patients

include chemotherapy-induced thrombocytopenia,


disseminated intravascular coagulopathy, vitamin K defi-
ciency (due to poor diet), clotting factor deficiencies
caused by liver damage, immune-mediated platelet
destruction, and thrombotic thrombocytopenia purpura.

Neoplastic Aneurysms
■ These are aneurysms caused by neoplastic infiltration of
arteries.

73724_CH08_Printer.indd 171 03/29/09 2:40:00 PM


172 Chapter 8

■ Rupture may result in subarachnoid or intraparenchymal


hemorrhages.
■ Neoplastic aneurysms are usually small and located in
distal artery branches.
■ They are typically caused by metastasis of cardiac myxoma
or choriocarcinoma, although cases of lung cancer metas-
tasis have been reported.

Pituitary Tumor Apoplexy


■ Refers to hemorrhage or infarction of a preexisting pitu-

itary adenoma
■ Uncommon syndrome

■ Often spontaneous

■ Signs/symptoms caused by a sudden increase in sellar

contents compressing surrounding structures and portal


vessels
• Sudden, severe headache
• Visual disturbances
• Impairment in pituitary function
■ Management

• Supportive therapy with intravenous fluids and


corticosteroids
• Clinically unstable patients may require urgent surgi-
cal decompression
• Long-term follow-up for management of tumor and
pituitary dysfunction

Vascular Endothelial Growth Factor (VEGF) Pathway Inhibitors


■ Includes monoclonal antibodies against VEGF (e.g., bev-

acizumab) and tyrosine kinase inhibitors targeting the


VEGF receptor (e.g., sorafenib, sunitinib)
■ Increased risk of bleeding noted in early clinical studies

of bevacizumab ranging from minor mucocutaneous


bleeding (e.g., epistaxis) to major life-threatening bleed-
ing (e.g., hematemesis, hemoptysis)26
■ CNS metastases

• Until recently, patients with brain metastases were


often excluded from clinical trials of VEGF and
VEGFR inhibitors.26,31

73724_CH08_Printer.indd 172 03/29/09 2:40:00 PM


Cerebrovascular Complications 173

• Based on a review of clinical trials of VEGF and


VEGFR therapies in systemic cancer, rates of intrac-
ranial hemorrhage are negligible.26 In patients with
known brain metastases treated with anti-VEGFR
therapy, there were no episodes of intracranial
hemorrhage.
• Preliminary results from retrospective review of all
patients treated with bevacizumab in clinical trials (undi-
agnosed brain metastases at time of enrollment or
developed brain metastases during trial) are as follows31:
■ Data set A (13 completed randomized controlled

phase II and III trials in which 187 patients with


CNS metastases identified): low incidence of hem-
orrhage, 3/91 (3.29%) in bevacizumab arm, and
1/96 (1.04%) in nonbevacizumab arm
■ Data set B (2 open-label, single-arm safety studies

in which 321 patients with CNS metastases identi-


fied): low incidence of cerebral hemorrhage 3/321
(0.93%)
■ Data set C (2 ongoing trials in which 131 patients

with NSCLC and pretreated CNS metastases were


included): low incidence of cerebral hemorrhage,
1/131 (0.8%)
■ High-grade gliomas
• Based on phase II clinical trials of bevacizumab in
recurrent high-grade gliomas, rates of major intracra-
nial hemorrhage have ranged from 0 to 2.9%.32–37 Of
note, many of these trials excluded patients with
evidence of intracranial hemorrhage on imaging prior
to the initiation of therapy.
• In a small retrospective study of high-grade glioma
patients, the addition of anticoagulation to bevaci-
zumab treatment was not associated with major intrac-
erebral hemorrhage causing neurologic deficits or
death.38

Cerebral Venous Thrombosis (CVT)


■ Nonseptic thrombosis of the intracranial veins and/or
sinuses

73724_CH08_Printer.indd 173 03/29/09 2:40:00 PM


174 Chapter 8

Risk Factors
■ Similar risk factors as deep venous thrombosis, including
coagulation disorders, cancer, and pregnancy
■ May also be caused by compression or invasion of cere-

bral sinuses from dural-based lesions such as menin-


giomas, brain metastases, or lymphoma

Clinical Features
■ Clinical presentation is often nonspecific.

■ Most common signs and symptoms are headache, sei-

zures, focal neurologic deficits, altered consciousness,


and papilledema.
■ The onset of symptoms may be acute ( ,48 hours),

subacute (48 hours to 30 days), or chronic (.30 days).


■ Main patterns of presentation include39

• Isolated intracranial hypertension (headache and


papilledema)
• Focal syndrome (focal deficit, seizure)
• Cavernous sinus syndrome including orbital pain,
chemosis, proptosis, oculomotor palsies
• Subacute encephalopathy

Diagnosis
■ Imaging of the venous system demonstrating occlusion is

most helpful. Options include MR venography or CT


venography.
■ MRI and MR venography have been the noninvasive

imaging techniques of choice, although CT venography is


emerging as a competing technique.40
• Thrombus may manifest as absence of flow void, best
seen on FLAIR images and T2-weighted spin-echo
images.
• Imaging may demonstrate indirect signs of CVT,
including
■ Diffuse cerebral edema

■ Venous infarction

• Suspicion for a venous infarction should be raised if


the infarction does not conform to an arterial vascular
territory.

73724_CH08_Printer.indd 174 03/29/09 2:40:00 PM


Cerebrovascular Complications 175

• The lesion may be hemorrhagic.


• Deep venous thrombosis is typically associated with
bilateral or unilateral infarction in the thalami, basal
ganglia, and internal capsules.
■ A head CT may show direct or indirect signs of CVT,
although it may be negative in 10% to 30% of cases
of CVT.40
• Noncontrast head CT may demonstrate a dense clot
sign from thrombosis of the dural sinus or a cord sign
from thrombosis of a cortical vein.
• Contrast-enhanced head CT may demonstrate an
empty delta sign from thrombosis of the dural sinus.
• A head CT may also demonstrate indirect signs of CVT,
as seen in MRI (discussed previously in this chapter).

Treatment
■ Therapeutic anticoagulation

• Acute treatment of CVT with intravenous or subcuta-


neous heparin has been demonstrated on the basis of
randomized trials, a meta-analysis, and case series.39
• The length of treatment is unclear, although anticoag-
ulation with warfarin should be continued for at least
6 to 12 months, possibly longer depending on the risk
of further thromboses.
■ The utility of thrombolysis for CVT in cancer and non-

cancer patients is unclear.


■ The management of elevated intracranial pressure is the

same as the management of elevated intracranial pres-


sure from other causes.

Prognosis39
■ The prognosis in all patients with acute CVT (not just

cancer patients) is a 15% overall death and dependency


rate.
■ Long-term predictors of poor prognosis include any type

of cancer, CNS infection, deep venous system thrombosis,


intracranial hemorrhage, and age greater than 37 years.
■ Predictors of death at 30-days include depressed con-

sciousness, deep venous system thrombosis, right hemi-


spheric hemorrhage, and posterior fossa lesions.

73724_CH08_Printer.indd 175 03/29/09 2:40:00 PM


176 Chapter 8

■ The main causes of acute death include transtentorial


herniation caused by large hemorrhage, multiple lesions,
or diffuse brain edema.

References
1. Graus F, Rogers LR, Posner JB. Cerebrovascular complica-
tions in patients with cancer. Medicine (Baltimore). 1985;
64(1):16–35.
2. Cestari DM, Weine DM, Panageas KS, Segal AZ, DeAngelis
LM. Stroke in patients with cancer: incidence and etiology.
Neurology. 2004;62(11):2025–2030.
3. Oberndorfer S, Nussgruber V, Berger O, Lahrmann H,
Grisold W. Stroke in cancer patients: a risk factor analysis.
J Neurooncol. 2009;94(2):227.
4. Grisold W, Oberndorfer S, Struhal W. Stroke and cancer:
a review. Acta Neurol Scand. 2009;119(1):1–16.
5. Adams HP, Jr., del Zoppo G, Alberts MJ, et al. Guidelines
for the early management of adults with ischemic stroke:
a guideline from the American Heart Association/American
Stroke Association Stroke Council, Clinical Cardiology
Council, Cardiovascular Radiology and Intervention
Council, and the Atherosclerotic Peripheral Vascular Disease
and Quality of Care Outcomes in Research Interdisciplinary
Working Groups: the American Academy of Neurology
affirms the value of this guideline as an educational tool for
neurologists. Stroke. 2007;38(5):1655–1711.
6. Sacco RL, Adams R, Albers G, et al. Guidelines for preven-
tion of stroke in patients with ischemic stroke or transient
ischemic attack: a statement for healthcare professionals
from the American Heart Association/American Stroke
Association Council on Stroke: co-sponsored by the Council
on Cardiovascular Radiology and Intervention: the American
Academy of Neurology affirms the value of this guideline.
Stroke. 2006;37(2):577–617.
7. Lefkovitz NW, Roessmann U, Kori SH. Major cerebral
infarction from tumor embolus. Stroke. 1986; 17(3):
555–557.
8. el-Shami K, Griffiths E, Streiff M. Nonbacterial thrombotic
endocarditis in cancer patients: pathogenesis, diagnosis, and
treatment. Oncologist. 2007;12(5):518–523.

73724_CH08_Printer.indd 176 03/29/09 2:40:00 PM


Cerebrovascular Complications 177

9. Rabinstein AA, Giovanelli C, Romano JG, Koch S, Forteza


AM, Ricci M. Surgical treatment of nonbacterial thrombotic
endocarditis presenting with stroke. J Neurol. 2005;
252(3):352–355.
10. Thaler DE, Saver JL. Cryptogenic stroke and patent fora-
men ovale. Curr Opin Cardiol. 2008;23(6):537–544.
11. Meister SG, Grossman W, Dexter L, Dalen JE. Paradoxical
embolism: diagnosis during life. Am J Med. 1972;53(3):
292–298.
12. Cheng SW, Wu LL, Ting AC, Lau H, Lam LK, Wei WI.
Irradiation-induced extracranial carotid stenosis in patients
with head and neck malignancies. Am J Surg. 1999;
178(4):323–328.
13. Scott AS, Parr LA, Johnstone PA. Risk of cerebrovascular
events after neck and supraclavicular radiotherapy: a sys-
tematic review. Radiother Oncol. 2009;90(2):163–165.
14. Butler MJ, Lane RH, Webster JH. Irradiation injury to large
arteries. Br J Surg. 1980;67(5):341–343.
15. Perry A, Schmidt RE. Cancer therapy-associated CNS neu-
ropathology: an update and review of the literature. Acta
Neuropathol. 2006;111(3):197–212.
16. Kayser-Gatchalian MC, Kayser K. Thrombosis and intracra-
nial tumors. J Neurol. 1975;209(3):217–224.
17. van der Sande JJ, Veltkamp JJ, Bouwhuis-Hoogerwerf ML.
Hemostasis and intracranial surgery. J Neurosurg. 1983;
58(5):693–698.
18. Ulmer S, Braga TA, Barker FG II, Lev MH, Gonzalez RG,
Henson JW. Clinical and radiographic features of peritu-
moral infarction following resection of glioblastoma.
Neurology. 2006;67(9):1668–1670.
19. Ferreri AJ, Campo E, Seymour JF, et al. Intravascular lym-
phoma: clinical presentation, natural history, management
and prognostic factors in a series of 38 cases, with special
emphasis on the “cutaneous variant.” Br J Haematol. 2004;
127(2):173–183.
20. Szots M, Szomor A, Kover F, et al. Intravascular lymphoma-
tosis of the nervous system. J Neurol. 2008; 255(10):
1590–1592.
21. Murase T, Nakamura S, Kawauchi K, et al. An Asian variant
of intravascular large B-cell lymphoma: clinical, pathological
and cytogenetic approaches to diffuse large B-cell lymphoma

73724_CH08_Printer.indd 177 03/29/09 2:40:00 PM


178 Chapter 8

associated with haemophagocytic syndrome. Br J Haematol.


2000;111(3):826–834.
22. Ponzoni M, Ferreri AJ, Campo E, et al. Definition, diagno-
sis, and management of intravascular large B-cell lymphoma:
proposals and perspectives from an international consensus
meeting. J Clin Oncol. 2007;25(21):3168–3173.
23. Iijima M, Fujita A, Uchigata M, Katoo H. Change of brain
MRI findings in a patient with intravascular malignant lym-
phomatosis. Eur J Neurol. 2007;14(5):e4–e5.
24. Baumann TP, Hurwitz N, Karamitopolou-Diamantis E,
Probst A, Herrmann R, Steck AJ. Diagnosis and treatment
of intravascular lymphomatosis. Arch Neurol. 2000;
57(3):374–377.
25. Park MS, Kim BC, Kim IK, et al. Cerebral infarction in IgG
multiple myeloma with hyperviscosity. J Korean Med Sci.
2005;20(4):699–701.
26. Carden CP, Larkin JM, Rosenthal MA. What is the risk of
intracranial bleeding during anti-VEGF therapy? Neuro
Oncol. 2008;10(4):624–630.
27. White JB, Piepgras DG, Scheithauer BW, Parisi JE. Rate of
spontaneous hemorrhage in histologically proven cases of
pilocytic astrocytoma. J Neurosurg. 2008;108(2):223–226.
28. Martinez-Lage JF, Poza M, Martinez M, Esteban JA,
Antunez MC, Sola J. Meningiomas with haemorrhagic
onset. Acta Neurochir (Wien). 1991;110(3–4):129–132.
29. Wakai S, Yamakawa K, Manaka S, Takakura K. Spontaneous
intracranial hemorrhage caused by brain tumor: its inci-
dence and clinical significance. Neurosurgery. 1982;
10(4):437–444.
30. Qureshi AI, Mendelow AD, Hanley DF. Intracerebral haem-
orrhage. Lancet. 2009;373(9675):1632–1644.
31. Rohr UP, Augustus S, Lasserre SF, Compton P, Huang J.
Safety of bevacizumab in patients with metastases to the
central nervous system. J Clin Oncol (Meeting Abstracts).
2009;27(Suppl):abstr 2007.
32. Sathornsumetee S, Vredenburgh JJ, Rich JN, et al. Phase II
study of bevacizumab and erlotinib in patients with recur-
rent glioblastoma multiforme. J Clin Oncol (Meeting
Abstracts). 2008;26(Suppl):abstr 13008.
33. Cloughesy TF, Prados MD, Wen PY, et al. A phase II, ran-
domized, non-comparative clinical trial of the effect of beva-
cizumab (BV) alone or in combination with irinotecan (CPT)

73724_CH08_Printer.indd 178 03/29/09 2:40:00 PM


Cerebrovascular Complications 179

on 6-month progression free survival (PFS6) in recurrent,


treatment-refractory glioblastoma (GBM). J Clin Oncol
(Meeting Abstracts). 2008;26(Suppl):abstr 2010b.
34. Rich JN, Desjardins A, Sathornsumetee S, et al. Phase II
study of bevacizumab and etoposide in patients with
recurrent malignant glioma. J Clin Oncol (Meeting Abstracts).
2008;26(Suppl):abstr 2022.
35. Vredenburgh JJ, Desjardins A, Herndon JE II, et al. Phase II
trial of bevacizumab and irinotecan in recurrent malignant
glioma. Clin Cancer Res. 2007;13(4):1253–1259.
36. Vredenburgh JJ, Desjardins A, Herndon JE, 2nd, et al.
Bevacizumab plus irinotecan in recurrent glioblastoma mul-
tiforme. J Clin Oncol. 2007;25(30):4722–4729.
37. Kreisl TN, Kim L, Moore K, et al. Phase II trial of single-
agent bevacizumab followed by bevacizumab plus irinotecan
at tumor progression in recurrent glioblastoma. J Clin Oncol.
2009;27(5):740–745.
38. Nghiemphu PL, Green RM, Pope WB, Lai A, Cloughesy
TF. Safety of anticoagulation use and bevacizumab in
patients with glioma. Neuro Oncol. 2008;10(3):355–360.
39. Bousser MG, Ferro JM. Cerebral venous thrombosis: an
update. Lancet Neurol. 2007;6(2):162–170.
40. Poon CS, Chang JK, Swarnkar A, Johnson MH, Wasenko J.
Radiologic diagnosis of cerebral venous thrombosis: pictorial
review. AJR Am J Roentgenol. 2007;189(6 Suppl):S64–S75.

73724_CH08_Printer.indd 179 03/29/09 2:40:00 PM


73724_CH08_Printer.indd 180 03/29/09 2:40:00 PM
C H A P T E R 9

Medical Complications
of Brain Tumor Patients
Seizures
Epidemiology
■ Up to 60% of patients with brain tumors may present
with seizures or may have a seizure for the first time after
diagnosis of the tumor.1
■ The risk of seizure depends on the type of brain tumor,

grade, location, changes in peritumoral environment, and


genetic factors.1,2
• Slow-growing tumors (i.e., low-grade gliomas) are the
most epileptogenic, although the high frequency of
seizures may be related to longer survival.
• Seizure frequency is based on tumor type: dysem-
bryoplastic neuroepithelial tumors 100%, low-grade
astrocytomas and oligodendrogliomas 60% to 85%,
glioblastomas 30% to 50%, and brain metastases 20%
to 35%.
• Cortical tumors (frontal, temporal, parietal more than
occipital) cause seizures more frequently than infraten-
torial or sellar tumors.
■ In patients with systemic cancer, the incidence of seizures

unrelated to brain metastases is only 5%.3


■ Incidence of seizures in patients with brain metastases is

approximately 25% to 40%.4

Clinical Features
■ May present as simple or complex seizures with or with-

out secondary generalization


■ Causes of seizures in patients with cancer

• Related to parenchymal tumor: epileptogenic activity


likely arises from adjacent cortex since tumors are
usually electrically inert5

181

73724_CH09_Printer.indd 181 04/01/10 12:07:00 AM


182 Chapter 9

• Related to infiltrative lesions such as leptomeningeal


carcinomatosis or intravascular lymphomatosis
• Related to radiation necrosis, including temporal lobe
radionecrosis from head and neck radiation
• Related to central nervous system infection or oppor-
tunistic infection
• Related to epileptogenic medications including some
antidepressants, neuroleptics, antibiotics, and chemo-
therapy (e.g., high-dose busulfan, cisplatin, chloram-
bucil, isophosphamide, paclitaxel, methotrexate)
• Related to paraneoplastic syndromes such as limbic
encephalitis
• Related to metabolic disorders
• Related to cerebrovascular complications such as intrac-
ranial hemorrhages, venous sinus thrombosis, or stroke

Seizure Prophylaxis
Perioperative Period
■ Conflicting data regarding the prophylactic use of anti-

epileptic drugs in patients undergoing surgery for a brain


tumor2

Outside the Perioperative Period


■ American Academy of Neurology Practice Parameters6

• “Because of their lack of efficacy and their potential


side effects, prophylactic anticonvulsants should not
be routinely used in patients with newly diagnosed
brain tumors.”
• “In patients with brain tumors who have not had a
seizure, tapering and discontinuing anticonvulsants
after the first postoperative week is appropriate. . . .”
■ Cochrane Review: “Evidence for seizure prophylaxis

with phenobarbital, phenytoin, and divalproex sodium in


people with brain tumors is inconclusive. . . . Therefore,
there are no data supporting the use of prophylactic
antiepileptics and the risk of adverse events lessens their
overall potential benefit.”1

73724_CH09_Printer.indd 182 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 183

■ Sirven et al. meta-analysis: “In patients with no history of


seizures, evidence does not support the use of seizure
prophylaxis with phenobarbital, phenytoin, or valproic
acid for gliomas, cerebral metastases or meningiomas.”7

Seizure Treatment
■ Limited data are available on the efficacy of specific anti-

epileptic medications in patients with brain tumors.


• Several prospective and retrospective studies showed
that add-on levetiracetam decreased seizure frequency
in 27% to 90% of patients with brain tumors.2
• Small prospective study of add-on gabapentin showed
seizure reduction in all patients with 57% becoming
seizure free.8
■ Some patients may develop medical refractory epilepsy

due to several reasons, including multidrug resistance.


Treatment options include
• Surgical resection of epileptogenic zone: 70% to 90%
of patients become seizure free or enjoy a substantial
reduction in seizure frequency after total resection of
the epileptogenic zone.2
• Radiation: small series demonstrated reductions in
seizure frequency following radiation.9,10
• Chemotherapy: temozolomide reduced seizure fre-
quency in 50–60% of glioma patients.11,12
■ Side effects are more frequent in patients with brain

tumors compared with the overall epilepsy population.2

Antiepileptic Drug–Drug Interactions


■ Enzyme-inducing antiepileptic drugs (EIAED) (Table 9-1)
• Includes phenobarbital, primidone, carbamazepine,
and phenytoin
• Induce cytochrome P450 coenzymes, resulting in
decreased effectiveness of medications metabolized
by the P450 system, including several chemothera-
peutic agents and corticosteroids

73724_CH09_Printer.indd 183 04/01/10 12:07:00 AM


Table 9-1: Common Antiepileptic Medications

Generic Name Trade Name Special Considerations in Brain Tumor Patients

Drugs that cause little or no induction of P450 enzymes

73724_CH09_Printer.indd 184
184 Chapter 9

Clonazepam Klonopin Used as adjuvant therapy; side effects that include sedation

Gabapentin Neurontin Three to four times daily dosing, perceived as less efficacious than other antiepileptics,
although data are conflicting

Lamotrigine Lamictal Slow titration to minimize rash risk and therefore may take several weeks to get to
therapeutic levels

Levetiracetam Keppra Therapeutic starting dose; side effects that include depression

Lorazepam Ativan Used for status epilepticus; side effects that include sedation

Tiagabine Gabitril Side effects that include depression

Topiramate Topamax Weak enzyme inducer; side effects that include cognitive slowing; slow titration needed to
minimize CNS adverse effects.

(Continues)

04/01/10 12:07:00 AM
Table 9-1: Common Antiepileptic Medications (Continued)

Generic Name Trade Name Special Considerations in Brain Tumor Patients

Valproic Acid Depakote, Side effects that include hepatotoxicity, thrombocytopenia and abnormal coagulation at higher

73724_CH09_Printer.indd 185
Depakene doses, enzyme inhibiting effects (which may raise levels of certain chemotherapeutic agents)

Zonisamide Zonegran Side effects that include cognitive impairment; slow titration necessary

Drugs that induce P450 enzymes

Carbamazepine Tegretol Side effects that include hyponatremia, small risk of bone marrow suppression

Oxcarbazepine Trileptal Dose-related effects that are similar to carbamazepine, although oxcarbazepine is a weaker
P450 inducer

Phenobarbital Luminal Behavioral and cognitive side effects

Phenytoin Dilantin Strong P450 inducer

Primidone Mysoline Similar side effects as phenobarbital but possibly more sedating that phenobarbital

Data are from Bromfield EB. Epilepsy. In: Samuels MA, ed. Manual of Neurologic Therapeutics, 7th ed. Philadelphia: Lippincott Williams &
Medical Complications of Brain Tumor Patients 185

Wilkins; 2004:33–64. Porter RJ, Meldrum BS. Antiseizure drugs. In: Katzung BG, Masters SB, Trevor AJ, eds. Basic and Clinical Pharmacology,
11th ed. New York: McGraw-Hill Companies; 2009:399–422.

04/01/10 12:07:00 AM
186 Chapter 9

• Chemotherapeutic agents affected by EIAEDs include


nitrosureas (e.g., temozolomide), paclitaxel, cyclo-
phosphamie, etoposide, topotecan, irinotecan,
thiotepa, doxorubicin, and methotrexate
• Phenytoin and phenobarbital shorten the half-life of
dexamethasone and prednisone13
■ Valproic acid is an enzyme-inhibiting antiepileptic drug,
which may result in raised plasma concentrations of other
agents and possibly increased toxic effects.
■ Many chemotherapeutic agents may affect the plasma
concentrations of antiepileptic medications.2
■ Agents with minimal or no induction of P450 enzymes
are found in Table 9-1.

Practice Considerations
■ New-onset seizures in a patient with known systemic
cancer should raise suspicion for brain metastases.
A brain MRI with contrast is the imaging modality of
choice for brain metastases.
■ Seizure prophylaxis is generally not recommended in

patients with no history of seizures.


■ Antiepileptic treatment is recommended in patients with

brain tumors after the first seizure.


■ The length of treatment after the first and only seizure is

unclear, although patients may require lifetime antiepi-


leptic treatment, especially if the tumor is still present.
■ Each state has specific laws regarding driving after a

seizure. In general, physicians should warn their patients


regarding the risks of driving and other behaviors that
could result in injury or death should a seizure occur
(i.e., swimming).
■ Consider drug–drug interactions and side-effect profiles

(e.g., bone marrow suppression, cognitive impairment)


when choosing an antiepileptic for a patient.

Venous Thromboembolism (VTE)


■ VTE includes deep vein thrombosis (DVT) and pulmonary
embolism (PE).

73724_CH09_Printer.indd 186 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 187

Epidemiology
■ VTE is common in patients with cancer, including
primary brain tumors.
■ The incidence of symptomatic VTE in patients with

high-grade glioma outside the perioperative period is 20%


to 30%.14
■ The risk for VTE in high-grade glioma patients is

higher in the postoperative period and in patients with


hemiplegia.
■ The incidence of symptomatic VTE in primary CNS

lymphoma is 18%.14
■ The incidence of VTE is higher in patients with cancer

than patients without cancer.15


■ It is the second leading cause of death in cancer

patients.16

Risk Factors14
■ Postoperative period

■ Hemiparesis

■ Age ⬎60

■ Large tumor size

■ Chemotherapy

■ Hormonal therapy

■ Steroids and osmotic diuretic agents, which may also

increase the rate of clot formation17

Clinical Features
■ DVT: erythema, warmth, pain, tenderness, and edema in

one leg
■ PE: chest pain, cough, tachypnea, tachycardia, and short-

ness of breath

Diagnosis
■ DVT: duplex ultrasonography

■ PE: chest CT angiogram

Prophylaxis
■ Prophylaxis is recommended in the perioperative setting
and during hospitalization.

73724_CH09_Printer.indd 187 04/01/10 12:07:00 AM


188 Chapter 9

■ An optimal prophylactic regimen has not been established.


■ Options for prophylaxis include the following:
• Compression stockings and sequential pneumatic
compression devices
• Unfractionated heparin
• Low molecular weight heparin (LMWH) (e.g., enox-
aparin, dalteparin, tinzaparin, and nadroparin)
■ In patients with brain tumors, LMWH and unfraction-
ated heparin reduce the risk of VTE from 12.5% to 6.2%
and carry a 2% risk for major bleeding.18
■ The need for prophylaxis beyond the perioperative period
is less clear.
• A randomized phase III clinical trial of malignant
glioma patients that tested the safety and efficacy
of long-term dalteparin for prevention of VTE versus
placebo closed early due to expiration of study med-
ication.19
• In this study, there was a trend in favor of dalteparin
in reducing VTE but the results were not statistically
significant (although the study may have been under-
powered).
• There was also a trend for increased intracranial
hemorrhage in the dalteparin group, although the
difference was not statistically significant.

Treatment
■ The main objective of treatment of a DVT is to prevent

PE, as pulmonary emboli are associated with increased


morbidity and mortality.
■ Consider a noncontrast head CT before initiating therapy

to evaluate for intracerebral hemorrhage.


■ Options for treatment include the following:

• Low molecular weight heparin (LMWH)


• Unfractionated heparin IV or LMWH with transition
to warfarin
• In patients with a contraindication to therapeutic anti-
coagulation (i.e., intracerebral hemorrhage, recent
craniotomy, prolonged thrombocytopenia), an inferior
vena cava (IVC) filter should be placed.

73724_CH09_Printer.indd 188 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 189

■ An IVC filter is not necessary if the patient is able


to receive therapeutic anticoagulation.
■ IVC filters are associated with a higher complica-

tion rate and are less effective in preventing PEs as


compared to anticoagulation.20
■ IVC filter complications include pneumothorax,

infection, bleeding, IVC wall damage, recurrent


VTE, postphlebitic syndrome, and IVC thrombosis.
• In patients with heparin-induced thrombocytopenia,
direct thrombin inhibitors (e.g., lepirudin, argatroban,
bivalirudin) should be used instead.
■ Clinical trials and systematic reviews comparing differ-
ent anticoagulants in cancer patients with VTE suggest
that LMWHs are more effective than unfractionated
heparin and at least as effective or possibly more effec-
tive than warfarin.
• In a multicenter, randomized, open-label trial com-
paring enoxaparin versus warfarin for 3 months, war-
farin was associated with a high bleeding rate,
although the differences were not statistically signifi-
cant.21 When combining recurrent VTE and major
hemorrhage as the primary endpoint, the enoxaparin
group had statistically significant lower rates of this
combined endpoint.
• In a multicenter, randomized, open-label trial compar-
ing dalteparin followed by warfarin versus dalteparin
alone for 6 months, rates of recurrent VTE were lower
in the dalteparin alone group. There was no signifi-
cant difference in the rate of major bleeding or any
bleeding.
• Meta-analyses show that LMWHs are more effective
than unfractionated heparin for the initial treatment
of VTE and are associated with less major
bleeding.22,23
■ Initial treatment with unfractionated heparin (as opposed
to LMWH) should be considered in patients with serious
PEs and in patients at high risk for hemorrhage (as
protamine is able to reverse unfractionated heparin more
completely than LMWH).

73724_CH09_Printer.indd 189 04/01/10 12:07:00 AM


190 Chapter 9

■ Properly monitored anticoagulation can be relatively safe


in patients with primary and metastatic brain tumors.20
• The incidence of cerebral hemorrhage is generally not
significantly elevated in studies.
• Hemorrhagic complications most commonly occur in
the setting of supratherapeutic anticoagulation.
■ The optimal length of treatment after an initial VTE is
unknown.
• No clinical trials have been performed to answer this
question.
• Some advocate using therapeutic anticoagulation
indefinitely in patients with cancer or at least as long
as the cancer is active.24

Cerebral Edema
■ There are two main types of cerebral edema.
• Vasogenic edema is most commonly associated with
brain tumors.
• Cytotoxic edema is due to hypoxia and is often caused
by ischemic strokes or traumatic brain injury.
■ Vasogenic edema results from the extravasation of intra-
vascular fluid and proteins into cerebral parenchyma
extracellular space due to breakdown of tight endothelial
junctions in the blood–brain barrier.

Clinical Features
■ Depend on location of cerebral edema

■ May include focal neurologic deficits, headaches, nau-

sea, vomiting, and lethargy

Diagnosis
■ The diagnosis is based on clinical history, symptoms, and

imaging.
■ MRI is the best imaging modality for assessment of

cerebral edema.
• Cerebral edema is hypointense on T1-weighted images
and hyperintense on T2-weighted images.
• It may be difficult to differentiate cerebral edema
from an infiltrating tumor.

73724_CH09_Printer.indd 190 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 191

Management14
Corticosteroids
■ Indicated for symptomatic peritumoral edema (usually

not necessary in asymptomatic patients with peritumoral


edema on neuroimaging)
■ Usually adequate for managing most peritumoral edema

■ Mechanism of action not well understood

■ Side effects (also discussed later in “Treatment Compli-

cations and Their Management” section)


• The side effects include osteoporosis, compression
fractures, avascular necrosis, steroid myopathy, gastro-
intestinal bleeding, pneumocystis jiroveci pneumoni-
tis (PJP), Cushingnoid features, adrenal insufficiency,
and cataracts.
• The severity of complications corresponds to the dose
and duration of symptoms.
• Most resolve after stopping steroids (except for osteo-
porosis and posterior subcapsular cataracts).
• The severity may be reduced with using the lowest
dose possible.14
■ Dexamethasone

• This is the preferred corticosteroid because of less


mineralocorticoid activity, and possibly lower risk of
infection and cognitive impairment compared with
other corticosteroids.25
• Dosing depends on the severity of symptoms.
■ The typical starting dose is 10 mg.

■ The typical initial maintenance dose is 16 mg per

day (divided into two to four times a day dosing),


but lower doses may be used if less symptomatic.
■ PO and IV dosing are equivalent.

■ In general, use the lowest dose that still provides

symptomatic benefit.
■ If lowering the dose, the dose should be tapered.

The longer the patient has been on steroids, the


slower the taper, although the length of the taper
will vary from patient to patient according to
symptoms.
• This induces improvement of neurologic symptoms
caused by peritumoral edema within 48 hours.

73724_CH09_Printer.indd 191 04/01/10 12:07:00 AM


192 Chapter 9

• Improvement on imaging studies often lags behind


clinical improvement.
• Prophylactic treatment for potential side effects from
dexamethasone should be considered for the following
complications:
■ Gastrointestinal complications: histamine H 2

receptor blocker (e.g., famotidine, ranitidine) or


proton pump inhibitor (e.g., omeprazole, lansopra-
zole, pantoprazole)
■ Pneumocystis jiroveci pneumonitis (PJP)

■ Osteoporosis

Vascular Endothelial Growth Factor (VEGF)


Pathway Inhibitors
• VEGF is a major proangiogenic peptide that is partly
responsible for the loss of integrity of the blood–brain
barrier in brain tumors.26
• Anti-VEGF agents could be used to reduce vasogenic
cerebral edema.
■ In a phase II study of cediranib in patients with

glioblastoma patients, this agent significantly


reduced tumor-associated edema.27
■ For significantly elevated intracranial pressure and mass

effect not responsive to corticosteroids or for impending


herniation, hospitalization is warranted.
• Immediate measures to decrease intracranial pressure
such as elevation of the head of the bed, hyperventila-
tion, and fluid restriction should be started before
providing more definitive therapy.
• Hypertonic fluids
• Mannitol
• Surgical debulking

Pain
Headaches
Causes
■ These are typically caused by increased intracranial pres-

sure from the brain tumor and/or peritumoral edema.

73724_CH09_Printer.indd 192 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 193

■ Other causes should be considered in the correct clinical


context, including intratumoral hemorrhages, meningeal
processes, venous sinus thrombosis, analgesic medica-
tion overuse, depression, and migraines.
Clinical Features
■ Headaches that cause early morning awakening or are
more prominent on waking should raise suspicion for
increased intracranial pressure.
■ Headaches are typically associated with nausea and

vomiting.
■ Headaches are worse with coughing or Valsalva maneuver.

■ Papilledema may be present on funduscopic examination.

Treatment
■ Steroids are an option.

■ All classes of pain medications, including opioids, may be

considered; however, nonsteroidal anti-inflammatory


medications may increase the risk of gastric toxicity from
corticosteroids.

Neuropathic Pain
Potential Causes
■ Leptomeningeal disease

■ Lymphomatous invasion of nerves

■ Skull base involvement

■ Herpes zoster

■ Radiation

■ Chemotherapy (see Chapter 6 for chemotherapy-induced

peripheral neuropathy)
Clinical Features
■ Painful dysesthesias typically localized to specific der-

matomes, nerve root distributions, or peripheral extremi-


ties in a stocking-glove distribution
■ May be spontaneous and/or evoked by a stimulus

Treatment
■ May require a combination of agents
■ Spontaneous resolution

73724_CH09_Printer.indd 193 04/01/10 12:07:00 AM


194 Chapter 9

• Pain associated with acute chemotherapy-induced


neuropathy may resolve after discontinuation of the
culprit chemotherapy.
• Neuropathic symptoms may progress for up to 2
months after stopping oxaliplatin or cisplatin.
• Recovery may be incomplete in some patients, result-
ing in chronic neuropathic pain.
■ Antiepileptic medications for neuropathic pain, includ-
ing gabapentin, carbamazepine, and pregabalin
• High doses of gabapentin may be required for pain
control (maximum up to 3,200 mg per day divided in
TID or QID dosing).
■ Antidepressants for neuropathic pain, including tricy-
clics (desipramine, amitriptyline, nortriptyline) and
serotonin/norepinephrine reuptake inhibitors (venlafax-
ine, duloxetine)
■ Topical or transdermal agents, including lidocaine
patches
■ All classes of pain medications, including opioids; how-
ever, nonsteroidal anti-inflammatory medications may
increase the risk of gastric toxicity.
■ Corticosteroids for patients with neuropathic pain from
leptomeningeal disease

Cognitive Impairment
Potential causes
■ Brain tumor
■ Radiation (see Chapter 5)

■ Chemotherapy (see Chapter 6)

■ Epilepsy and antiepileptic drugs

• In a study of low-grade gliomas, the presence of epi-


lepsy and the use of antiepileptic drugs were indepen-
dently associated with changes in memory, attention,
and communication.28
• The use of antiepileptics is associated with a sixfold
increase in cognitive deficits (such as attention, psy-
chomotor speed, executive function) as compared
with radiation.29
■ Other medical factors and complications contributing to

cognitive and neurobehavioral changes include endocrine

73724_CH09_Printer.indd 194 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 195

dysfunction, metabolic disturbances, infection, pain,


fatigue, anxiety, and depression.

Treatment
■ Only a few treatment trials in patients with brain tumors
have been conducted.
■ Psychostimulants

• Methylphenidate
■ Case study of methylphenidate in three high-grade

glioma patients with cognitive deficits from primary


brain tumors or radiation described improvements
in arousal, attention, initiation speed of tasks, and
mood.30
■ Open-labeled study of methylphenidate in high-

grade glioma patients showed significant improve-


ments in psychomotor speed, memory, visual–motor
function, executive function, motor speed, and dex-
terity, as well as subjective improvements in their
ability to function despite progressive disease and
increasing radiation damage.31
• Modafinil
■ In a pilot study of primary brain tumor patients

with fatigue or cognitive dysfunction, modafinil


improved cognition, mood, and fatigue.32
■ Donepezil

• In a study of brain tumor patients who received radia-


tion, donepezil improved cognitive functioning, includ-
ing memory, mood, and quality of life in patients with
brain tumors who received radiation therapy.33
■ Ventriculoperitoneal shunting

• Could be considered in patients with cognitive dys-


function due to communicating hydrocephalus34
■ Cognitive rehabilitation

Fatigue
■ Negatively affects quality of life

Potential Causes
■ Radiation (fatigue tends to increase with the number of
fractions)

73724_CH09_Printer.indd 195 04/01/10 12:07:00 AM


196 Chapter 9

■ Antiepileptics
■ Chemotherapy
■ Anemia
■ Metabolic disturbances
■ Depression
■ Endocrine dysfunction

Treatment
■ Psychostimulants (e.g., methylphenidate, pemoline, dex-
troamphetamine, modafinil) are generally well tolerated
in brain tumor patients but may lower the seizure thresh-
old (except for modafinil)

Treatment Complications and Their


Management
Musculoskeletal
Osteoporosis and Compression Fractures
■ Chronic corticosteroids (prednisone dose greater than

2.5 to 5 mg/day) increase the risk of osteoporosis, which,


in turn, predisposes a patient to fractures such as verte-
bral compression fractures.35,36
■ Clinical features associated with compression fractures

include back pain, nerve impingement (numbness, tin-


gling, weakness), urinary/stool incontinence, or retention
due to spinal cord compression.
■ Diagnosis

• Osteoporosis is diagnosed with a bone mineral density


(BMD) test.
• Vertebral compression fractures can be seen on X-rays
or spine CT scans, but a spine MRI may be necessary
to evaluate for spinal cord compression (if neurologic
symptoms are present).
■ Prophylactic therapy should be provided to patients on

chronic steroids.
• Calcium supplementation 1,500 mg/day
• Vitamin D 800 IU daily or activated vitamin D such as
calcitriol 0.5 µm/day

73724_CH09_Printer.indd 196 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 197

■ Treatment
• Calcium and vitamin D should be given to all patients
with osteoporosis.
• Bisphosphonates (e.g., etidronate, alendronate,
risedronate, zoledronate) improve BMD and may
reduce the risk of fractures although the data are
not definitive.20
• Compression fractures may require hospitalization if
neurologic symptoms are present or if the pain is too
severe to manage as an outpatient.
• Spine surgeon consultation is warranted if the patient
has spinal cord compression or spine instability.
• Vertebroplasty is often used to relieve pain from com-
pression fractures, but two recent multicenter ran-
domized, placebo-controlled trials demonstrated no
benefit for painful osteoporotic vertebral fractures.
■ In a randomized trial of 131 patients with painful

osteoporotic vertebral compression fractures com-


paring vertebroplasty versus a simulated procedure
without cement, improvements in pain and pain-
related disability at 1 month were similar in both
groups.37
■ In a randomized trial of 78 patients with painful

osteoporotic vertebral fractures comparing verte-


broplasty with a sham procedure, the authors found
no beneficial effect at 1 week or at 1, 3, or 6 months
after treatment.38

Steroid Myopathy
■ Occurs in up to 60% of cancer patients39
■ More common in older persons and after prolonged use

of high-dose corticosteroids
■ Clinical features

• Subacute onset proximal muscle weakness and


wasting (especially in the lower extremities) result-
ing in difficulty climbing stairs or rising from a
seated position
• Normal muscle enzymes

73724_CH09_Printer.indd 197 04/01/10 12:07:00 AM


198 Chapter 9

■ Diagnosis
• Diagnosis is primarily based on clinical history.
• Electromyography is nonspecific and may demonstrate
myopathic changes.
• Muscle biopsy is only necessary if the diagnosis is in
question and may contain type IIb muscle fiber
atrophy.
■ Prophylactic therapy
• Nonfluorinated steroids (e.g., prednisone, hydrocorti-
sone) may carry a lower risk of steroid myopathy than
fluorinated steroids (e.g., dexamethasone).20
• Regular exercise may attenuate symptoms of steroid
myopathy.
■ Treatment
• Stopping steroids or decreasing to the lowest possi-
ble dose
• Physical therapy
■ Prognosis: recovery may take several months

Syndromes Associated With Steroid Withdrawal


Adrenal Insufficiency
■ Caused by impaired adrenal steroid synthesis

■ Seen with mitotane, ketoconazole, aminoglutethimide,

megestrol, prior cranial irradiation for childhood brain


tumors, or rapid reduction in corticosteroid therapy
■ Rarely seen with replacement of both adrenal glands by

metastases
■ Clinical features

• Patients may develop nausea, vomiting, anorexia, and


lethargy.
• Acute adrenal crisis (hypotension, mental status
changes, hypoglycemia) may occur with abrupt ces-
sation.
■ Diagnosis: rapid ACTH stimulation test

■ Treatment

• Mild symptoms can be managed as an outpatient by


increasing the steroid dose and then tapering more
gradually or using alternate day administration.
• Acute adrenal insufficiency may require hospitaliza-
tion for intravenous hydration, glucose, and steroids.

73724_CH09_Printer.indd 198 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 199

Steroid-Withdrawal Syndrome
■ Clinical features include nausea, headache, myalgias,
malaise, and weight loss.40
■ Treatment includes slower steroid taper or alternate day

administration.

Steroid Pseudorheumatism41
■ Clinical features include diffuse arthralgias.

■ Treatment includes slower steroid taper and NSAIDs.

Gastrointestinal Complications
Peptic Ulceration
■ Peptic ulceration may theoretically occur with continued

use of corticosteroids.
■ From studies of patients on steroids, the overall inci-

dence of peptic ulceration is low, but the incidence is


increased in patients on both steroids and nonsteroidal
anti-inflammatory medications.14
■ Clinical features include nonspecific epigastric pain with

variable relationship to meals, nausea, and anorexia.


■ Diagnosis

• Upper endoscopy
• Abdominal CT only when complications such as per-
foration, penetration, or obstruction suspected
■ Prophylaxis includes histamine H 2 receptor blocker

(e.g., famotidine, ranitidine) or a proton pump inhibitor


(e.g., omeprazole, lansoprazole, pantoprazole)
■ Treatment regimens differ based on presence or absence

of H. pylori infection

Upper Gastrointestinal Hemorrhage


■ This may occur as a complication from peptic ulcer

disease.
■ Clinical features include melena, hematemesis, melenem-

esis, and hemodynamic instability.


■ Prophylaxis includes histamine H receptor blocker
2
(e.g., famotidine, ranitidine) or a proton pump inhibitor
(e.g., omeprazole, lansoprazole, pantoprazole).
■ Hospitalization is indicated, as this could be a life-

threatening emergency.

73724_CH09_Printer.indd 199 04/01/10 12:07:00 AM


200 Chapter 9

Bowel Perforation
■ This is a rare complication of VEGF inhibitors such as
bevacizumab or peptic ulcer disease.
■ Clinical features include typically sudden-onset severe

abdominal pain. Shock, fever, nausea, and vomiting may


be present, although patients may be asymptomatic as
steroids can mask symptoms.
■ Hospitalization is indicated, as this could be a life-

threatening emergency.

Opportunistic Infections
Oropharyngeal Candidiasis (Thrush)
■ Most common opportunistic infection from steroid

immunosuppression
■ Caused by the fungus Candida albicans

■ Clinical features

• Oral pain, dysphagia, odynophagia, dysgeusia, and


aversion to food
• Thrush: characterized by white patches on the sur-
face of the buccal and labial mucosa, tongue, and soft
palate
■ Treatment

• Nystatin oral suspension 100,000 units four times a


day (swish and swallow) should be continued for sev-
eral days after clinical healing.
• If thrush does not improve with nystatin oral suspen-
sion, an azole antifungal agent should be provided.
■ Fluconazole 100 mg daily for 1 to 2 weeks

■ Clotrimazole troche 5 mL three to four times per

day for a 2-week minimum


■ Itraconazole capsule or oral solution 100 mg daily

for 2 weeks
• “Miracle mouthwash” (containing diphenhydramine,
lidocaine, and a Maalox-type aluminum/magnesium
antacid) may help with symptomatic relief of pain but
does not treat thrush.

Pneumocystis Jiroveci Pneumonitis (PJP)


■ PJP was previously known as pneumocystis carinii pneu-

monitis (PCP).

73724_CH09_Printer.indd 200 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 201

■ It is caused by the fungus Pneumocystis Jiroveci.


■ The frequency of PJP is low in cancer (187 cases per
100,000 patients with hematological cancer, 16 cases per
100,000 patients with solid tumors)42 but can be life
threatening.
■ Predisposing factors include corticosteroids, intensity
of chemotherapy treatment (e.g., daily temozolomide
treatment in glioma patients), and low CD4 cell count
(⬍200 cells/mm3).43
■ Clinical features are similar to acute pneumonia.
■ Diagnosis is as follows:
• Radiological imaging demonstrates bilateral interstitial
infiltrates.
• Definitive diagnosis requires cytologic examination of
bronchoalveolar lavage.
■ There are no published guidelines for prophylaxis in
patients without HIV, but the following regimens are
commonly used:
• Trimethoprim-sulfamethoxazole (TMP-SMZ) double
strength three times a week or single strength daily
• Aerosolized pentamidine 300 mg monthly
• Dapsone 50 mg twice per day or 100 mg daily
• Atovaquone 1,500 mg daily
■ For treatment, consider consultation with an infectious
disease specialist.

Behavioral and Psychiatric Changes


■ Clinical features

• Steroids may induce behavioral or psychiatric changes,


including anxiety, insomnia, emotional lability, eupho-
ria, and psychosis.44
• Steroids may also induce cognitive deficits such as
distractibility and memory impairment.45
■ Treatment

• Stopping or tapering steroids to the lowest possible


dose
• Consider neuroleptics (e.g., haldol, olanzapine, ris-
peridone, quetiapine) for more severe behavioral
changes such as psychosis, aggression, or agitation
• Consider antidepressants for depression

73724_CH09_Printer.indd 201 04/01/10 12:07:00 AM


202 Chapter 9

• Consider mood stabilizers for mania


• Consider consultation with a psychiatrist
Endocrine Abnormalities
■ Hyperglycemia should be monitored in patients on ste-

roids because diabetes predisposes patients to infections.


■ Hyperglycemia is associated with shorter survival in

patients with glioblastoma, even after controlling for


mean daily dexamethasone dose.46

References
1. Tremont-Lukats IW, Ratilal BO, Armstrong T, Gilbert MR.
Antiepileptic drugs for preventing seizures in people with
brain tumors. Cochrane Database Syst Rev. 2008 (2):
CD004424.
2. van Breemen MS, Wilms EB, Vecht CJ. Epilepsy in patients
with brain tumours: epidemiology, mechanisms, and man-
agement. Lancet Neurol. 2007;6(5):421–430.
3. Villemure JG, de Tribolet N. Epilepsy in patients with cen-
tral nervous system tumors. Curr Opin Neurol. 1996;9(6):
424–428.
4. Villanueva V, Codina M, Elices E. Management of epilepsy
in oncological patients. Neurologist. 2008;14(6 Suppl 1):
S44–S54.
5. Wolf HK, Roos D, Blumcke I, Pietsch T, Wiestler OD.
Perilesional neurochemical changes in focal epilepsies. Acta
Neuropathol. 1996;91(4):376–384.
6. Glantz MJ, Cole BF, Forsyth PA, et al. Practice parameter:
anticonvulsant prophylaxis in patients with newly diagnosed
brain tumors. Report of the Quality Standards Subcommittee
of the American Academy of Neurology. Neurology.
2000;54(10):1886–1893.
7. Sirven JI, Wingerchuk DM, Drazkowski JF, Lyons MK,
Zimmerman RS. Seizure prophylaxis in patients with brain
tumors: a meta-analysis. Mayo Clin Proc. 2004;79(12):
1489–1494.
8. Perry JR, Sawka C. Add-on gabapentin for refractory sei-
zures in patients with brain tumours. Can J Neurol Sci.
1996;23(2):128–131.
9. Chalifoux R, Elisevich K. Effect of ionizing radiation on par-
tial seizures attributable to malignant cerebral tumors.
Stereotact Funct Neurosurg. 1996;67(3–4):169–182.

73724_CH09_Printer.indd 202 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 203

10. Rogers LR, Morris HH, Lupica K. Effect of cranial irradia-


tion on seizure frequency in adults with low-grade astrocy-
toma and medically intractable epilepsy. Neurology. 1993;
43(8):1599–1601.
11. Brada M, Viviers L, Abson C, et al. Phase II study of primary
temozolomide chemotherapy in patients with WHO grade II
gliomas. Ann Oncol. 2003;14(12):1715–1721.
12. Pace A, Vidiri A, Galie E, et al. Temozolomide chemotherapy
for progressive low-grade glioma: clinical benefits and radio-
logical response. Ann Oncol. 2003;14(12):1722–1726.
13. Ruegg S. Dexamethasone/phenytoin interactions: neuroon-
cological concerns. Swiss Med Wkly. 2002;132(29–30):
425–426.
14. Wen PY, Schiff D, Kesari S, Drappatz J, Gigas DC, Doherty L.
Medical management of patients with brain tumors.
J Neurooncol. 2006;80(3):313–332.
15. Zacharski LR, Prandoni P, Monreal M. Warfarin versus
low-molecular-weight heparin therapy in cancer patients.
Oncologist. 2005;10(1):72–79.
16. Pruemer J. Prevalence, causes, and impact of cancer-
associated thrombosis. Am J Health Syst Pharm. 2005;62(22
Suppl 5):S4–S6.
17. Hamilton MG, Hull RD, Pineo GF. Venous thromboembo-
lism in neurosurgery and neurology patients: a review.
Neurosurgery. 1994;34(2):280–296; discussion 296.
18. Iorio A, Agnelli G. Low-molecular-weight and unfraction-
ated heparin for prevention of venous thromboembolism in
neurosurgery: a meta-analysis. Arch Intern Med. 2000;
160(15):2327–2332.
19. Perry JR, Rogers L, Laperriere N, et al. PRODIGE: A phase
III randomized placebo-controlled trial of thromboprophy-
laxis using dalteparin low molecular weight heparin
(LMWH) in patients with newly diagnosed malignant
glioma. J Clin Oncol (Meeting Abstracts). 2007;25(Suppl):
abstr 2011.
20. Drappatz J, Schiff D, Kesari S, Norden AD, Wen PY. Medical
management of brain tumor patients. Neurol Clin.
2007;25(4):1035–1071, ix.
21. Meyer G, Marjanovic Z, Valcke J, et al. Comparison of low-
molecular-weight heparin and warfarin for the secondary
prevention of venous thromboembolism in patients with
cancer: a randomized controlled study. Arch Intern Med.
2002;162(15):1729–1735.

73724_CH09_Printer.indd 203 04/01/10 12:07:00 AM


204 Chapter 9

22. Segal JB, Streiff MB, Hofmann LV, Thornton K, Bass EB.
Management of venous thromboembolism: a systematic
review for a practice guideline. Ann Intern Med. 2007;
146(3):211–222.
23. van Dongen CJ, van den Belt AG, Prins MH, Lensing AW.
Fixed dose subcutaneous low molecular weight heparins
versus adjusted dose unfractionated heparin for venous
thromboembolism. Cochrane Database Syst Rev. 2004(4):
CD001100.
24. Prevention and treatment of venous thromboembolism.
International Consensus Statement (guidelines according to
scientific evidence). Int Angiol. 2006;25(2):101–161.
25. Batchelor T, DeAngelis LM. Medical management of cere-
bral metastases. Neurosurg Clin N Am. 1996;7(3):435–446.
26. Gerstner ER, Duda DG, di Tomaso E, et al. VEGF inhibi-
tors in the treatment of cerebral edema in patients with
brain cancer. Nat Rev Clin Oncol. 2009;6(4):229–236.
27. Batchelor TT, Sorensen AG, di Tomaso E, et al. AZD2171,
a pan-VEGF receptor tyrosine kinase inhibitor, normalizes
tumor vasculature and alleviates edema in glioblastoma
patients. Cancer Cell. 2007;11(1):83–95.
28. Taphoorn MJ. Neurocognitive sequelae in the treatment of
low-grade gliomas. Semin Oncol. 2003;30(6 Suppl 19):
45–48.
29. Klein M, Heimans JJ, Aaronson NK, et al. Effect of radio-
therapy and other treatment-related factors on mid-term to
long-term cognitive sequelae in low-grade gliomas: a com-
parative study. Lancet. 2002;360(9343):1361–1368.
30. Weitzner MA, Meyers CA, Valentine AD. Methylphenidate
in the treatment of neurobehavioral slowing associated with
cancer and cancer treatment. J Neuropsychiatry Clin
Neurosci. 1995;7(3):347–350.
31. Meyers CA, Weitzner MA, Valentine AD, Levin VA.
Methylphenidate therapy improves cognition, mood, and
function of brain tumor patients. J Clin Oncol. 1998;
16(7):2522–2527.
32. Kaleita TA, Wellisch DK, Graham CA, et al. Pilot study of
modafinil for treatment of neurobehavioral dysfunction and
fatigue in adult patients with brain tumors. J Clin Oncol
(Meeting Abstracts). 2006;24(Suppl):abstr 1503.
33. Shaw EG, Rosdhal R, D’Agostino RB, Jr., et al. Phase II
study of donepezil in irradiated brain tumor patients: effect
on cognitive function, mood, and quality of life. J Clin
Oncol. 2006;24(9):1415–1420.

73724_CH09_Printer.indd 204 04/01/10 12:07:00 AM


Medical Complications of Brain Tumor Patients 205

34. Thiessen B, DeAngelis LM. Hydrocephalus in radiation


leukoencephalopathy: results of ventriculoperitoneal shunt-
ing. Arch Neurol. 1998;55(5):705–710.
35. Van Staa TP, Leufkens HG, Abenhaim L, Zhang B, Cooper C.
Use of oral corticosteroids and risk of fractures. J Bone Miner
Res. 2000;15(6):993–1000.
36. van Staa TP, Leufkens HG, Cooper C. The epidemiology of
corticosteroid-induced osteoporosis: a meta-analysis.
Osteoporos Int. 2002;13(10):777–787.
37. Kallmes DF, Comstock BA, Heagerty PJ, et al. A random-
ized trial of vertebroplasty for osteoporotic spinal fractures.
N Engl J Med. 2009;361(6):569–579.
38. Buchbinder R, Osborne RH, Ebeling PR, et al. A random-
ized trial of vertebroplasty for painful osteoporotic vertebral
fractures. N Engl J Med. 2009;361(6):557–568.
39. Batchelor TT, Taylor LP, Thaler HT, Posner JB, DeAngelis
LM. Steroid myopathy in cancer patients. Neurology.
1997;48(5):1234–1238.
40. Lester RS, Knowles SR, Shear NH. The risks of systemic
corticosteroid use. Dermatol Clin. 1998;16(2):277–288.
41. Rotstein J, Good RA. Steroid pseudorheumatism. AMA Arch
Intern Med. 1957;99(4):545–555.
42. Torres HA, Chemaly RF, Storey R, et al. Influence of type of
cancer and hematopoietic stem cell transplantation on clini-
cal presentation of Pneumocystis jiroveci pneumonia in
cancer patients. Eur J Clin Microbiol Infect Dis. 2006;
25(6):382–388.
43. Mansharamani NG, Balachandran D, Vernovsky I, Garland R,
Koziel H. Peripheral blood CD4 ⫹ T-lymphocyte counts
during Pneumocystis carinii pneumonia in immunocompro-
mised patients without HIV infection. Chest. 2000;118(3):
712–720.
44. Truhan AP, Ahmed AR. Corticosteroids: a review with
emphasis on complications of prolonged systemic therapy.
Ann Allergy. 1989;62(5):375–391.
45. Hall RC, Popkin MK, Stickney SK, Gardner ER. Presentation
of the steroid psychoses. J Nerv Ment Dis. 1979;167(4):
229–236.
46. Derr RL, Ye X, Islas MU, Desideri S, Saudek CD, Grossman
SA. Association between hyperglycemia and survival in
patients with newly diagnosed glioblastoma. J Clin Oncol.
2009;27(7):1082–1086.

73724_CH09_Printer.indd 205 04/01/10 12:07:00 AM


73724_CH09_Printer.indd 206 04/01/10 12:07:00 AM
Index

A Anti-Ma2 (Ta), 140–141


Anti-NMDA receptors, 141
Acute cerebellar toxicity, 113 Anti-voltage-gated potassium
Acute encephalopathy, 113 channels, 141
American Academy of Neurology Astrocytoma
Practice Parameters on seizure anaplastic astrocytoma, 11–15
prophylaxis, 182 diagnosis, 78–79
Amifostine, 122 diffuse astrocytoma, 8–11
Anaplastic astrocytoma (WHO glioblastoma, 15–18
Grade III/IV) pathology of, 78
biology, 11 pilocytic astrocytoma, 8
clinical features, 12 treatment, 79
MRI appearance, 12 Atypical meningiomas, 23
prognosis, 14–15 Autonomic neuropathy, 147
treatment Autosomal dominant
chemotherapy, 12 disorders, 4–5
radiation therapy, 12
recurrent disease, 14
surgery, 12
B
Anaplastic/malignant
meningiomas, 23 Benign meningiomas, 23
Anaplastic oligodendroglioma Bevacizumab, 100
biology, 19 Bowel perforation, 200
clinical features, 19 Brachytherapy, 93
MRI appearance, 19 Brain biopsy, 44
prognosis, 21–22 Brain metastases
treatment breast cancer, 53–55
chemotherapy, 20–21 diagnosis, 44
radiation therapy, 20 epidemiology, 43
recurrent, 21 melanoma, 55–57
surgery, 19–20 non-small cell lung cancer,
Angiotropic large-cell lymphoma. 57–58
See Intravascular lymphoma parenchymal metastases, 43
Antibody-mediated PND, 128–129 prognostic factors
Anticoagulation, 100 RPA classes, 45–46
Anti-CRMP5 (anti-CV2), 140 renal cell carcinoma, 58
Antiepileptic drug-drug surgery approach
interactions, 183–186 for multi brain metastasis, 52
Anti-Hu, 140 for single brain metastasis, 51

207

73724_IDXx_Printer.indd 207 04/02/10 09:50:00 PM


208 Index

Brain metastases (continued) management


vs. SRS, 52 corticosteroids, 191
with/without WBRT, 52 dexamethasone, 191–192
treatment approach VEGF pathway inhibitors, 192
chemotherapy, 52–53 types of, 190
SRS, 48–50 Cerebral venous thrombosis (CVT)
whole-brain radiation therapy, clinical features, 174
46–48 diagnosis, 174–175
Breast cancer, brain metastases prognosis, 175–176
epidemiology, 53–54 risk factors, 174
risk factors, 54 treatment, 175
treatment recommendations Chemical aseptic meningitis, 89
stereotactic radiosurgery, 54–55 Chemotherapy
surgery, 54 agents, 55
systemic treatments, 55 anaplastic astrocytoma, 12
whole-brain radiation therapy, 55 anaplastic oligodendroglioma,
20–21
astrocytomas, 79
C
brain metastases treatment, 52–53
Cancer-associated retinopathy common chemotherapy agents,
(CAR), 143–144 107–109
Capecitabine, 55 diffuse astrocytoma, 10–11
Carcinomatous meningitis, 83 leptomeningeal metastases
Central nervous system (CNS) intrathecal chemotherapy, 88–89
chemotherapy-related cognitive systemic chemotherapy, 89
impairment (Chemobrain), in melanoma, 58
110–111 metastatic epidural spinal cord
focal radiation necrosis, 99–101 compression, 72
imaging abnormalities following nerve sheath tumors, 75
radiation therapy, 96 seizure treatment, 183
metastases treatment schemes, 109
in breast cancer, 54 and WBRT, 48
in melanoma, 57 Chemotherapy-induced peripheral
in NSCLC, 56 neuropathy (CIPN)
neurocognitive dysfunction clinical signs and symptoms,
clinical features, 97 115, 120
clinical studies, 97–98 common chemotherapeutic
pathophysiology, 97 agents, 116–119
radiation, 96–97 prevention, 120–122
treatment, 98 risk factors, 115
neurotoxicity of specific agents, treatments and prognosis, 122
112–115 Chemotherapy-related cognitive
radiation-induced brain impairment (Chemobrain),
tumors, 101 110–111
radiation toxicity, 94–96 CIPN. See Chemotherapy-induced
RPLE, 111–112 peripheral neuropathy
Cerebral edema CNS. See Central nervous system
clinical features, 190 Coagulation disorders, 171
diagnosis, 190 Corticosteroids, 70–71, 191

73724_IDXx_Printer.indd 208 04/02/10 09:50:00 PM


Index 209

Cranial irradiation, 5, 94 5-Fluorouracil (5-FU), 112–113


CVT. See Cerebral venous Focal external beam radiation
thrombosis therapy, 27
Cytarabine, 112 Focal radiation necrosis, 98–101
Fractionation, 93
Frontal skull base meningiomas, 23
D
Deep vein thrombosis (DVT). See
Venous thromboembolism
G
Dermatomyositis, 151 Gastrointestinal complications
Dexamethasone, 191–192 bowel perforation, 200
Diffuse astrocytoma (WHO peptic ulceration, 199
Grade II/IV) upper gastrointestinal
biology of, 8 hemorrhage, 199
clinical features, 8 Glioblastoma (WHO Grade IV/IV)
MRI appearance, 8 biology, 15
prognosis, 11 clinical features, 15
treatment MRI appearance, 15
chemotherapy, 10–11 prognosis, 17–18
radiation therapy, 9–10 treatment
resection benefits, 9 newly diagnosed glioblastoma, 17
surgery, 8–9 recurrent glioblastoma, 17
Diffuse large B-cell lymphoma surgery, 16
(DLBCL), 29. See also Primary Gliomas
CNS lymphoma (PCNSL) astrocytic vs. oligodendroglial
Donepezil, 195 tumors, 6–7
astrocytoma
anaplastic, 11–15
E diffuse, 8–11
Embolism glioblastoma, 15–18
NBTE pilocytic, 8
characteristics of vegetations, 164 oligodendroglioma
clinical features, 164 anaplastic, 19–22
diagnosis and treatment, 165 oligodendroglioma, 18–19
paradoxical, 165–166 Glutamate, 121
septic, 165 Glutathione, 121
tumor, 163
Endocrine abnormalities, 202
Enzyme-inducing antiepileptic H
drugs (EIAED), 183–186 Headaches, 192–193
Epidural hemorrhage, 170 High-dose chemotherapy
Epidural spinal cord space, 68 with autologous stem-cell
External beam therapy (EBRT), 93 transplantation, 32
High-dose cytarabine, 112
F High-dose methotrexate
monotherapy, 31
Fatigue, 195–196 High-grade astrocytoma tumors, 79
Fludarabine, 113 High-grade gliomas, 173

73724_IDXx_Printer.indd 209 04/02/10 09:50:00 PM


210 Index

HIV-related PCNSL, 34–35 Intratumoral hemorrhage


Hodgkin lymphoma, 137 CNS metastases, 169–170
Hyperbaric oxygen therapy, 100–101 imaging studies, 169
Hyperglycemia, 202 primary brain tumors, 170
Hyperviscous obstruction, 168–169 Intravascular lymphoma
clinical features, 167
diagnosis, 167–168
I prognosis and treatment, 168
Intraventricular hemorrhage, 170
Ifosfamide, 113–114
Ionizing radiation exposure, 5
Inclusion body myositis, 151–152
Ischemic strokes, cancer patients
Inflammatory myopathies
management guidelines, 161–162
dermatomyositis, 151
risk factors, 161
inclusion body myositis, 151–152
treatment considerations, 163
polymyositis, 150–151
Intensity-modulated radiation
therapy (IMRT), 93 K
Interferon-␣, 114
Internal carotid artery (ICA) Karnofsky performance status
stenosis, 166 (KPS) scale, 44–45
Intracranial hemorrhages
cancer-related causes L
coagulation disorders, 171
neoplastic aneurysms, Lambert-Eaton myasthenic
171–172 syndrome (LEMS), 128, 148–149
pituitary tumor apoplexy, 172 Lapatinib, 55
VEGF pathway inhibitors, Leptomeningeal carcinomatosis, 83
172–173 Leptomeningeal metastases
clinical features, 171 clinical features, 84
diagnosis, 171 diagnosis, 85–86
management principles, 171 epidemiology, 83–84
types of, 169–170 pathogenesis, 84
Intramedullary astrocytoma. See prognosis, 86–87
Intramedullary spinal cord tumors treatment
Intramedullary metastasis, 79 chemotherapy, 88–89
Intramedullary spinal cord tumors radiation therapy, 88
astrocytoma supportive care, 89–90
diagnosis, 78–79 surgery, 87–88
pathology of, 78 Leukemic meningitis, 83
treatment, 79 Li-Faumeni (TP53), 4
clinical features, 77 Low-grade astrocytoma tumors, 79
ependymoma Low molecular weight heparin
diagnosis, 77 (LMWH), 188–189
treatment, 78 Lymphomatous meningitis, 83
intramedullary metastasis, 79 Lymphoplasmacytic lymphoma, 146
Intraparenchymal hemorrhage, 169
Intrathecal (IT) chemotherapy, M
31–32, 88–89
Intrathecal liposomal Malignant angioendotheliomatosis.
cytarabine, 112 See Intravascular lymphoma

73724_IDXx_Printer.indd 210 04/02/10 09:50:00 PM


Index 211

Malignant peripheral nerve-sheath histology


tumors (MPNSTs), 74 MPNSTs, 74
Melanoma-associated retinopathy neurofibromas, 74
(MAR), 144 schwannomas, 73–74
Melanoma, brain metastases treatment, 74–75
epidemiology, 55 Neurocognitive dysfunction
imaging, 56 clinical features, 97
treatment recommendations, clinical studies, 97–98
56–57 pathophysiology, 97
Meningeal layers, 68 radiation, 96–97
Meningioma, 76 treatment, 98
clinical features, 22–23 Neurofibromas, 74
imaging characteristics, 26 Neurofibromatosis, 4
pathology, 23, 26 Neuromyotonia, 147–148
prognosis, 28 Neuropathic pain, 193–194
risk factors, 22 Newly diagnosed glioblastoma, 17
treatment, 26–27 Nimodipine, 122
Metastatic epidural spinal cord Nonbacterial thrombotic
compression (MESCC) endocarditis (NBTE)
clinical features, 69–70 characteristics of vegetations, 164
diagnosis, 70 clinical features, 164
epidemiology, 68–69 diagnosis and treatment, 165
pathophysiology, 69 Nonionizing radiation exposure, 5
prognosis, 73 Non-small cell lung cancer
treatment (NSCLC), brain metastases
chemotherapy, 72 epidemiology, 55
corticosteroids, 70–71 risk factors, 56
radiation therapy, 71 treatment recommendations, 56–57
recurrence, 72
surgery, 71–72
Methotrexate, 31, 114–115 O
Methylphenidate, 195 Ocular lymphoma, 33
Modafinil, 195 Oligodendroglial tumors, 18–22
Monoclonal IgM antibodies, 146 Oligodendroglioma (WHO Grade
Musculoskeletal complications, II/IV), 18–19
196–198 Ommaya reservoir, 87–88
Myasthenia gravis (MG), 128, Opportunistic infections, 200–201
149–150 Org 2766, 122
Myxopapillary ependymoma, Oropharyngeal candidiasis
76–77 (Thrush), 200
Osteoporosis and compression
fractures, 196–197
N Oxcarbazepine, 122
NBTE. See Nonbacterial
thrombotic endocarditis P
Neoplastic aneurysms, 171–172
Neoplastic meningitis, 83 Pain
Nerve sheath tumors headaches, 192–193
diagnosis, 74 neuropathic, 193–194

73724_IDXx_Printer.indd 211 04/02/10 09:50:00 PM


212 Index

Palliative chemotherapy, 109 PCNSL. See Primary CNS


Paradoxical embolism, 165–166 lymphoma
Paraneoplastic cerebellar Peptic ulceration, 199
degeneration Peripheral nervous system
clinical features, 131, 136 CIPN
differential diagnosis, 136–137 clinical signs and symptoms,
imaging stages, 136 115, 120
prognosis, 137–138 common chemotherapeutic
serologic testing, 137 agents, 116–119
treatment, 137 prevention, 120–122
Paraneoplastic disorders (PND) risk factors, 115
antibodies, paraneoplastic treatments and prognosis, 122
syndromes, and associated disorders, 128
tumors, 132–135 postradiation optic neuropathy,
clinical features, 129 101–102
diagnosis, 127 radiation-induced peripheral
criteria, 130 nerve tumors, 102–103
workup, 131 radiation plexopathy, 102
epidemiology, 127–128 Peripheral neuropathy
pathogenesis autonomic, 147
antibody-mediated PND, causes in cancer patients, 144–145
128–129 chemotherapy-induced
T-cell mediated PND, 129 neuropathy, 115–122
Paraneoplastic limbic encephalitis paraneoplastic sensory, 145–146
clinical features, 139 paraproteinemic, 146
differential diagnosis, 140 sensorimotor neuropathy,
imaging studies, 139–140 145–146
laboratory abnormalities, 140–141 subacute sensory, 145
prognosis, 142 Pilocytic astrocytoma (WHO
treatment, 141–142 Grade I/IV), 8
Paraneoplastic motor neuron Pituitary tumor apoplexy, 172
disease, 144 PND. See Paraneoplastic disorders
Paraneoplastic opsoclonus- Pneumocystis jiroveci pneumonitis
myoclonus, 138–139 (PJP), 200–201
Paraneoplastic optic neuropathy, 144 Polymyositis, 150–151
Paraneoplastic sensory neuropathy, Posterior reversible encephalopathy
145–146 syndrome (PRES), 111–112
Paraneoplastic visual syndromes Postradiation optic neuropathy,
cancer-associated retinopathy, 101–102
143–144 Postradiation vasculopathy, 166
melanoma-associated Primary brain tumors
retinopathy, 144 astrocytic tumors
paraneoplastic optic anaplastic astrocytoma, 11–15
neuropathy, 144 diffuse astrocytoma, 8–11
Paraproteinemic neuropathy, 146 glioblastoma, 15–18
Parasagittal meningiomas, 23 pilocytic astrocytoma, 8
Parenchymal metastases, 43 diagnosis, 5
PCI. See Prophylactic cranial epidemiology, 1
irradiation histological classification of, 1–4

73724_IDXx_Printer.indd 212 04/02/10 09:50:00 PM


Index 213

Primary brain tumors (continued) radiation-induced brain


oligodendroglial tumors tumors, 101
anaplastic oligodendroglioma, radiation toxicity, 94–96
19–22 peripheral nervous system
oligodendroglioma, 18–19 postradiation optic neuropathy,
risk factors, 4–5 101–102
Primary CNS lymphoma (PCNSL) radiation-induced peripheral
clinical features, 29 nerve tumors, 102–103
diagnosis, 29–30 radiation plexopathy, 102
HIV-related PCNSL, 34–35 Radiation-induced brain tumors, 101
pathology, 29 Radiation-induced peripheral nerve
prognosis, 33–34 tumors, 102–103
risk factors, 28 Radiation necrosis, 100
treatment Radiation plexopathy, 102
adjuvant corticosteroids, 32 Radiation sensitizers, 47–48
adjuvant rituximab, 32–33 Radiation therapy
high-dose chemotherapy anaplastic astrocytoma, 12
with autologous stem-cell anaplastic oligodendroglioma, 20
transplantation, 32 Radiation therapy
high-dose methotrexate diffuse astrocytoma, 9–10
monotherapy, 31 leptomeningeal metastases, 88
intrathecal chemotherapy, meningioma, 27
31–32 MESCC, 71
methotrexate-based multidrug Radiation toxicity
regimens, 31 acute effects, 94
ocular lymphoma, 33 early-delayed effects, 94–95
recurrent/refractory PCNSL, 33 late-delayed effects, 95
surgery, 30 risk factors, 95–96
WBRT, 32 Recombinant human leukemia
Prophylactic cranial irradiation inhibitory factor (rhuLIF), 122
(PCI), 94, 98 Recurrent anaplastic
Prophylactic therapy, 196, 198 oligodendroglioma, 23
Psammomatous meningioma, 76 Recurrent glioblastoma, 17
Psychiatric and behavioral Recurrent MESCC, 72
complications, 201–202 Recurrent/refractory PCNSL, 33
Psychostimulants, 195 Recursive partitioning analysis
Pulmonary embolism (PE). See (RPA) classes, 45–46, 55
Venous thromboembolism Renal cell carcinoma (RCC), brain
metastases, 58
Reversible posterior
R
leukoencephalopathy syndrome
Radiation effects (RPLE), 111–112
central nervous system
focal radiation necrosis, 99–101 S
imaging abnormalities following
radiation therapy, 96 Schwannomas, 73–74
neurocognitive dysfunction, Seizures
96–98 antiepileptic drug-drug
pathology, 96 interactions, 183–186

73724_IDXx_Printer.indd 213 04/02/10 09:50:00 PM


214 Index

Seizures (continued) postoperative complications, 166


clinical features, 181–182 postradiation vasculopathy, 166
epidemiology, 181 Trastuzumab, 53–54
practice considerations, 186 Tuberous sclerosis, 4
prophylaxis, 182–183 Tumor embolism, 163
treatment, 183 Turcot syndrome, 5
Sensorimotor neuropathy, 145–146
Septic embolism, 165
Spinal cord tumors V
anatomy of, 67–68
extradural tumors Valproic acid, 186
extradural space, 68 Vascular endothelial growth factor
MESCC, 68–72 (VEGF) pathway inhibitors,
intradural tumors 172–173, 192
extramedullary tumors, 73–76 Vasogenic edema, 190
intramedullary tumors, 77–79 Venous thromboembolism (VTE)
Spinal ependymoma, 77–78 clinical features, 187
Spinal meningioma, 75–76 diagnosis, 187
Stereotactic radiosurgery (SRS), 27 epidemiology, 187
in breast cancer, 54–55 prophylaxis, 187–188
in brain metastases, 48–49 risk factors, 187
description of, 93 treatment, 188–190
with SRS/without WBRT, 50 Vertebroplasty, 197
surgery approach, 52 VTE. See Venous thromboembolism
with WBRT/without SRS, 49
Stereotactic radiotherapy (SRT), 93
Steroid myopathy, 197–198 W
Steroid pseudorheumatism, 199
Whole-brain radiation therapy
Steroid-withdrawal syndrome, 199
(WBRT), 32, 55
Stiff-person syndrome (SPS), 128,
and chemotherapy, in brain
142–143
metastases, 48
Subacute multifocal
description of, 94
leukoencephalopathy, 113
indications in brain
Subacute sensory neuronopathy, 145
metastases, 46
Subarachnoid hemorrhage, 170
melanoma in brain
Subarachnoid spinal cord space, 68
metastases, 57
Subdural hemorrhage, 170
and radiation sensitizers, 47–48
Subdural spinal cord space, 68
and surgery in brain
Systemic chemotherapy, 89
metastases, 52
treatment regimens, 47

T
T-cell mediated PND, 129 X
Thrombotic disease
Xaliproden, 121
hyperviscous obstruction,
168–169
intravascular lymphoma, 167–168

73724_IDXx_Printer.indd 214 04/02/10 09:50:00 PM

Potrebbero piacerti anche