You are on page 1of 75

A.

CENTRAL NERVOUS SYSTEM


63 Cancer of the Central Nervous System
Jay F. Dorsey, Ryan D. Salinas, Mai Dang, Michelle Alonso-Basanta,
Kevin D. Judy, Amit Maity, Robert A. Lustig, John Y.K. Lee,
Peter C. Phillips, and Amy A. Pruitt

S UMMARY OF K EY P OI N T S
Incidence • Most brain tumors are supratentorial; • Pineal region tumors (germ cell
• 78,980 new primary brain tumors will notable exceptions include brainstem tumors, pineocytomas,
be diagnosed in the United States in gliomas, cerebellar pilocytic pineoblastomas, and gliomas of this
2018, of which approximately astrocytomas, medulloblastomas, region) may compress the aqueduct
one-third will be malignant. and ependymomas that involve the of Sylvius, causing hydrocephalus.
• 3,560 new childhood (ages 0–19 posterior fossa. Compression of the pretectal area
years) primary benign and malignant • Glioblastoma (World Health produces Parinaud syndrome, with
CNS tumors will be diagnosed in Organization grade IV astrocytoma) paralysis of upgaze, ptosis, and loss
2018. Brain tumors are now the most and brainstem gliomas in children of pupillary light reflexes, along
commonly diagnosed malignancy carry the poorest prognosis. Pilocytic with retraction-convergence
among children at a rate of 5.47 per astrocytomas carry the best nystagmus.
100,000 population, surpassing prognosis. In 2016, the World Health Diagnostic Studies
leukemia (5.0 per 100,000).1 Organization reclassified brain • Magnetic resonance imaging with
• Radiation exposure is the most tumors to include molecular gadolinium contrast is the most
established risk factor for brain diagnostics based on genetic sensitive technique.
tumors. Hereditary syndromes mutations, which can clarify • Computed tomography scanning is
including neurofibromatosis types 1 prognosis and guide good for visualizing intratumoral
and 2, tuberous sclerosis, von treatment. calcifications and bone erosion but
Hippel-Lindau disease, and poor at visualizing the posterior
Li-Fraumeni syndrome are Clinical Manifestations
• General signs and symptoms from fossa.
associated with a higher risk of brain • Positron emission tomography and
tumors. mass effect, increased intracranial
pressure, edema, or shift or advanced magnetic resonance
Pathology and Classification destruction of surrounding brain imaging techniques may help
• Taking all age groups into account, tissue may include changes in discriminate between tumor
histologic types of CNS tumors personality and cognitive function, recurrence and radiation necrosis or
include meningiomas (36.6%), headaches, nausea, vomiting, pseudoprogression.
glioblastomas (14.9%), other seizures, and papilledema. • Magnetic resonance spectroscopy
astrocytomas (5.6%), nerve sheath • Focal signs and symptoms may may help distinguish a high-grade
tumors (8,2%), tumors of the include focal seizures, visual tumor from a low-grade tumor or
pituitary (15.9%), oligodendrogliomas changes, speech abnormalities, gait radiation necrosis.
(1,5%), ependymomas (1.8%), and abnormalities, and cranial nerve Therapy
embryonal tumors including deficits. • For most brain tumors, tissue
medulloblastomas (1%). • Posterior fossa tumors often diagnosis is required (an exception
• Among children aged 14 years or compress the fourth ventricle, may be selected brainstem
younger, histologic tumor types causing hydrocephalus, and gliomas).
include pilocytic astrocytomas frequently manifest with ataxia and • Treatment for brain tumors is highly
(18%), glioblastomas (2.8%), other intractable nausea and vomiting. dependent on histologic type.
astrocytomas (8.5%), ependymal • Brainstem gliomas often manifest For many tumors (e.g., gliomas,
tumors (5.8%), oligodendrogliomas with a combination of cranial nerve meningiomas, primitive
(0.7%), embryonal tumors including palsies and “long tract” signs such neuroectodermal tumors [PNETs],
medulloblastomas (13.8%), as hemianesthesia or hemiparesis and ependymomas), maximal
craniopharyngiomas (4%), and germ coupled with ataxia in cases with surgical resection that is safely
cell tumors (3.8%). cerebellar involvement. feasible is the primary treatment.

906
Cancer of the Central Nervous System • CHAPTER 63 907

• For some tumors (e.g., irradiation or surgery can offer management of many brain
glioblastomas, PNETs, and germ cell successful control; the decision tumors (e.g., glioblastomas,
tumors), radiation therapy is an between the two is based germ cell tumors, anaplastic
essential adjunct treatment after on assessment of adverse oligodendrogliomas, PNETs, and
surgery. effects. CNS lymphomas).
• For some tumors (e.g., acoustic • Chemotherapy is assuming an
neuromas and glomus tumors), either increasingly important role in the

The World Health Organization (WHO) classification of central in Fig. 63.1. The incidence of all brain tumors is highest in the 75- to
nervous system (CNS) tumors is based on tumor histology and cellular 84-year-old age group. The incidence of meningiomas increases with
origin and was established to provide a common classification and increasing age, whereas for gliomas and pituitary adenomas the incidence
grading system of human CNS tumors that is accepted and used increases with age but then declines at the highest age category (see
worldwide. The WHO divides brain tumors into classes: neuroepithelial Fig. 63.1A). Certain histologic types, such as germ cell tumors,
tumors, tumors of the cranial and paraspinal nerves, tumors of the medulloblastomas, and pilocytic astrocytomas, are far more common
meninges, lymphomas and hematopoietic neoplasms, and germ cell in children than in adults (Table 63.2; See Fig. 63.1B).
tumors and tumors of the sellar region.2 The different histologic types For most patients with primary CNS tumors, surgery usually is the
of CNS tumors are shown in Table 63.1. Meningiomas, glioblastomas, initial therapy. Radiation therapy is often an important component
and astrocytomas constitute more than half of all CNS tumors. The of treatment after surgery, but for management of malignant disease,
frequency of different histologic tumor types varies with age, as shown chemotherapy has an expanding role. A great deal of knowledge has

80
All primary tumors
70
Rate per 100,000 person-years

Gliomas
60 Meningioma
Nerve sheath tumors
50 Pituitary adenoma
Lymphoma
40

30

20

10

0
A (0–14) 0–19 20–34 35–44 45-54 55–64 65–74 75–84 85+

6
Rate per 100,000 person-years

0
B 0–4 5–9 10–14 15–19

All primary tumors


Figure 63.1 • Age-specific incidence of primary central nervous Gliomas
system tumors by histologic type. (A) Selected histologic types Embryonal tumors, including medulloblastoma
among all age groups. (B) Selected histologic tumor types in children.
(Data from Central Brain Tumor Registry of the United States. Pilocytic astrocytoma
Primary brain tumors in the United States, Statistical Report, 2012. Ependymoma/
Hinsdale, IL: Central Brain Tumor Registry of the United States; Anaplastic ependymoma
2012. Data were collected between the years 2004 and 2008.)
908 Part III: Specific Malignancies

Table 63.1 Primary Central Nervous System Tumors: Distribution by Histologic Type
FREQUENCY (%) OF PRIMARY CNS TUMORS BY HISTOLOGY
WHO 2016 Classification of Primary CNS Tumors Grade In All Age Groupsa Among Young Adultsb
Tumors of neuroepithelial tissue 29.3 28
Astrocytic tumors
Pilocytic astrocytoma I 1.4 2.8
Pilomyxoid astrocytoma
Subependymal giant cell astrocytoma I
Pleomorphic xanthoastrocytoma II
Anaplastic pleomorphic xanthoastrocytoma III
Diffuse astrocytoma II 2.2
Diffuse astrocytoma, IDH-mutant
Diffuse astrocytoma, IDH–wild type
Diffuse astrocytoma NOS
Anaplastic astrocytoma III 1.7
Anaplastic astrocytoma, IDH-mutant
Anaplastic astrocytoma, IDH–wild type
Anaplastic astrocytoma, NOS
Glioblastoma IV 14.9 4.4
Glioblastoma IDH–wild type
Glioblastoma IDH-mutant
Glioblastoma, NOS
Diffuse midline glioma H3K27M-mutant IV
Oligodendroglial tumors 1.0 3.5
Oligodendroglioma II
IDH-mutant and 1p/19q codeleted
Anaplastic oligodendroglioma III
IDH-mutant and 1p/10q-codeleted
Ependymal tumors
Ependymoma II 1.8 3.5
Ependymoma RELA fusion positive II or III
Anaplastic ependymoma III
Subependymoma I
Myxopapillary ependymoma I
Other gliomas
Angiocentric glioma I
Chordoid glioma of third ventricle II
Neuronal and mixed neuronal-glial tumors 1.2
Dysembryoplastic neuroepithelial tumor
Gangliocytoma I
Ganglioglioma I
Anaplastic ganglioglioma III
Dysplastic gangliocytoma of cerebellum (Lhermitte-Duclos) I
Desmoplastic infantile astrocytoma and ganglioglioma I
Papillary glioneuronal tumor I
Rosette-forming glioneuronal tumor I
Central neurocytoma II
Extraventricular neurocytoma II
Cerebellar liponeurocytoma II
Tumors of the pineal region 0.2
Pineocytoma I
Pineal parenchymal tumor of intermediate differentiation II or III
Pineoblastoma IV
Papillary tumor of the pineal region II or III
Cancer of the Central Nervous System • CHAPTER 63 909

Table 63.1 Primary Central Nervous System Tumors: Distribution by Histologic Type—cont’d
FREQUENCY (%) OF PRIMARY CNS TUMORS BY HISTOLOGY
WHO 2016 Classification of Primary CNS Tumors Grade In All Age Groupsa Among Young Adultsb
Embryonal tumors 1.0 1.6
Medulloblastoma (all subtypes) IV
Embryonal tumor with multilayered rosettes, C19MC-altered IV
Medulloepithelioma IV
CNS embryonal tumor, NOS IV
Atypical teratoid or rhabdoid tumor IV
CNS embryonal tumor with rhabdoid features IV
Tumors of cranial and paraspinal nerves 8.3 8.6
Schwannoma (neurilemmoma, neurinoma) I
Neurofibroma I
Perineuroma I
Malignant peripheral nerve sheath tumor II, III, IV
Tumors of the meninges 37.8
Tumors of meningiothelial cells
Meningioma I 36.6 15.9
Atypical meningioma II
Anaplastic (malignant) meningioma III
Other neoplasms related to the meninges
Solitary fibrous tumor, hemangiopericytoma I, II, or III
Hemangioblastoma, hemangioma I 3.5
Lymphomas and hematopoietic neoplasms 2.0 1.5
Germ cell tumors 0.4 1.3
Tumors of the sellar region 16.7 32.2
Craniopharyngioma I 0.8 1.2
Tumors of the pituitary I 15.9 31
Granular cell tumor I
Pituicytoma I
Spindle cell oncocytoma I

a
Frequency among all patients with primary brain and CNS tumors espectively (N = 368,117). Data from CBTRUS Statistical Report: Primary Brain and Other Central Nervous
System Tumors Diagnosed in the United States in 2010–2014 (Ostrom et al. 2016).
b
Frequency among young adults (15–34 years of age) with primary brain and CNS tumors (N = 54,388, respectively).
CNS, Central nervous system; IDH, isocitrate dehydrogenase; NOS, not otherwise specified; WHO, World Health Organization.
Data from Ostrom, Quinn T., et al. “CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2009–2013.”
Neuro-oncology 18.suppl_5 (2016): v1–v75.

been amassed during the past two decades regarding the biology has been increasing, with survival only modestly improved during the
of brain tumors that should lead to improved treatments in the past decade.4
near future. Ionizing radiation is one of the few factors shown to have a strong
association with the development of brain tumors. Exposure to ionizing
EPIDEMIOLOGY radiation represents the most important exogenous risk factor for
childhood brain tumors. Prenatal diagnostic x-ray exposure increases
An estimated 79,270 new cases of primary CNS tumors were expected the risk of childhood brain tumors,5 and various reports describe the
to be diagnosed in the United States in 2017.1 Approximately 25,850 occurrence of gliomas, meningiomas, and other brain tumors in children
of these tumors were expected to be malignant, representing 1.4% of who received radiation therapy to the head for tinea capitis and for
all primary malignant cancers diagnosed in the United States.3 prior malignancies.6–9 A dose of 1 to 2 Gy of radiation, which was
Malignant CNS tumors were expected to cause approximately 17,000 used to treat tinea capitis in Israeli children, was associated with an
deaths in 2017. increased risk of the development of brain tumors—specifically,
On the basis of data provided by the Surveillance, Epidemiology, meningiomas, gliomas, and nerve sheath tumors.10 A dose-response
and End Results (SEER) Program, Deorah and colleagues3 found correlation in the induction of brain tumors was seen, with a relative
that the incidence of brain cancer increased until 1987, when the risk of 3.0 at a dose of 1 Gy. Tumors developed at least 6 years after
annual percentage of change reversed direction. Elderly persons irradiation, with a mean interval greater than 15 years. Even lower
experienced an increase in brain cancer until 1985, but their rates doses of radiation delivered with radium-226 that were used to treat
were stable thereafter. Overall, however, the incidence of glioblastoma hemangiomas in Swedish infants (with a mean dose to the brain of
910 Part III: Specific Malignancies

brain tumors in humans (reviewed by Berleur and Cordier18). The


Table 63.2 Primary Central Nervous System relationship of human cytomegalovirus (HCMV) and gliomas remains
Tumors of Childhood: Distribution by controversial. The HCMV and gliomas symposium held in April 2011
Histologic Type resulted in consensus that sufficient evidence exists to conclude that
HCMV viral gene expression exists in most, if not all, malignant
FREQUENCY (%)
gliomas and that HCMV could influence the phenotype of malignant
Age 0–14 Yr Age 15–19 Yr gliomas by interacting with signaling pathways.19 Other authors who
Histologic Tumor Type (n = 16,653) (n = 6869) have used the data regarding cytomegalovirus (CMV) seroprevalence
Pilocytic astrocytoma 18 9.4 in the United States from the National Health and Nutrition
Examination Surveys, 1988 to 2004, have concluded that a possible
Glioma malignant NOS 14.3 5.1
CMV-glioblastoma association could not be corroborated with CMV
Embryonal tumors, including 13.8 3.6 seropositivity rates.20
medulloblastoma Also lacking is conclusive evidence that occupational exposure to
All other 10.2 11.5 industrial chemicals leads to the development of brain tumors, although
Other astrocytomas 8.5 7.4 a number of studies have suggested such a link (reviewed by Wrensch
Neuronal and mixed 6.6 7.9 and coworkers).21 Some of the chemicals that can induce brain tumors
neuronal-glial tumors in laboratory animals, such as polycyclic aromatic hydrocarbons, can do
Ependymal tumors 5.8 3.9
so only when administered by direct contact or transplacentally, but not
by inhalation or dermal contact; the latter two modes of exposure are
Nerve sheath tumors 4.9 5.9 more relevant in the occupational setting. Specific chemicals that have
Tumors of the pituitary 4.5 27.8 been examined include cosmetics containing N-nitroso compounds,
Craniopharyngioma 4.0 2,1 organic solvents, chemicals used in the manufacture of synthetic rubber,
Germ cell tumors 3.8 4.1 formaldehyde, phenols, polycyclic aromatic compounds, polyvinyl
Glioblastoma 2.8 3,3
chloride, and pesticides. Although many chemicals can induce brain
tumors in laboratory animals, no definitive associations have been found
Meningioma 1.6 5.0 in humans. For example, N-nitroso compounds, which commonly
Oligodendroglioma 0.7 1.6 are present in foods, are known to be neurocarcinogenic in animals.
Oligoastrocytic tumors 0.4 0.8 Oxidants in the environment can cause DNA damage, so it has been
Lymphoma 0.2 0.4 hypothesized that antioxidants, such as vitamin E, found in certain
foods may protect against the development of cancers. Epidemiologic
NOS, Not otherwise specified. studies, however, have provided mixed support for the idea that the
Data from Ostrom, Quinn T., et al. “CBTRUS statistical report: primary brain and intake of N-nitroso compounds, antioxidants, or specific nutrients in
other central nervous system tumors diagnosed in the United States in 2009–2013.” foods can influence the risk of developing brain tumors.18
Neuro-oncology 18.suppl_5 (2016): v1–v75. A great deal of interest has emerged in a possible association between
the use of cell phones and the risk of brain tumors. In a case-control
study, Inskip and coworkers were unable to show a correlation between
7 cGy) were found to be associated with an excess risk of intracranial the duration of cell phone use and the development of gliomas,
tumors, including pituitary adenomas, gliomas, meningiomas, and meningiomas, and acoustic neuromas.22 Other large case-control studies
nerve sheath tumors.11 also have failed to find any association between cell phone use and
A large amount of data has been accumulated on the incidence of the risk of developing brain tumors.23–25 A meta-analysis of nine
brain tumors in patients who received cranial irradiation for the case-control studies revealed no increased risk in analog or digital
treatment of acute lymphoblastic leukemia (ALL). The estimated cellular phone users.26 Nevertheless, some still claim that there is a
cumulative risk of secondary malignant brain tumors after childhood link between cell phone use and brain tumors.27 In addition, the
ALL therapy is 0.5% at 10 years after completion of therapy.12 In a International Agency for Research on Cancer (IARC), a WHO agency,
study from St. Jude Children’s Research Hospital, the actuarial 20-year has classified radiofrequency electromagnetic fields as “possibly carci-
probability of developing a brain tumor in these patients was 1.4%.13 nogenic to humans.”28
The probability of developing a high-grade glioma was greater in Other factors that have been analyzed for their possible relationship
children younger than 5 years of age at diagnosis than in those 6 years to the development of brain tumors include a history of head trauma
of age or older (1.08% versus 0.45%; P = .045). An apparent dose- and injury, drugs and medications, allergies, seizures, smoking and
response correlation was observed; the 20-year risk of developing a alcohol consumption, and exposure to power-frequency electromagnetic
brain tumor was 3.2% in patients who received more than 30 Gy, fields. None of these factors, however, has been conclusively shown
versus 1.03% in patients who received 21 Gy or less (P = .015). No to be important (reviewed by Wrensch and associates21).
CNS malignancies were seen in patients who did not receive cranial Most brain tumors represent sporadic cases; however, familial
irradiation. The latency period between irradiation and the diagnosis clustering has been noted. It is estimated that hereditary syndromes
of a brain tumor ranged from 5.9 to 29 years (median, 12.6 years) account for 2% of childhood brain tumors, although this may be an
but was longer for meningiomas (median, 19 years) than for high-grade underestimate because hereditary syndromes may be undiagnosed in
gliomas (median, 9.1 years). Very similar results regarding the frequency a number of cases.29 Some hereditary syndromes known to be associated
of brain tumors, latency, and dependence on prior cranial irradiation with brain tumors are listed in Table 63.3 (reviewed by Kimmelman
were seen in studies from the German Berlin-Frankfurt-Munster group14 and Liang30). Some of the associations are extremely strong. Nearly
and the Children’s Cancer Study Group.15 The types of brain tumors 70% of all optic pathway gliomas occur in patients with neuro-
that have been reported in these series include gliomas, meningiomas, fibromatosis type 1 (NF1), and acoustic neuromas commonly occur
and medulloblastomas. Patients who received cranial irradiation for in patients with NF2. In addition, hereditary immunosuppression
ALL also often received intrathecal chemotherapy. It has been suggested disorders such as Wiskott-Aldrich syndrome, treatment-associated
that cranial irradiation and intrathecal chemotherapy may work immunosuppression as in organ-transplant recipients, and exogenous
synergistically to increase the incidence of glial tumors.16,17 immunosuppression as in human immunodeficiency virus (HIV)
Viruses can induce brain tumors in animals in the experimental infection are known to be associated with an increased risk of primary
setting; however, no conclusive data point to viruses as a cause of central nervous system lymphoma (PCNSL).
Cancer of the Central Nervous System • CHAPTER 63 911

Table 63.3 Hereditary Syndromes Associated With Brain Tumors


Chromosomal
Syndrome Associated CNS Tumors Gene Locus Defective Protein and Normal Function
Neurofibromatosis type 1 Optic pathway gliomas, NF1 17q11-12 Neurofibromin; GTPase-activating protein that
meningiomas, neuromas negatively regulates Ras
Neurofibromatosis type 2 Bilateral acoustic neuromas, NF2 22q12 Merlin; related to membrane cytoskeleton linker
meningiomas, gliomas protein 4.1 superfamily
Tuberous sclerosis Cerebral hamartomas TSC1 9q34 Hamartin
Subependymal giant cell TSC2 16p13 Tuberin; associates with hamartin; both are involved in
astrocytoma (SEGA) signaling downstream of Akt
von Hippel-Lindau syndrome Hemangioblastomas VHL 3p25-29 VHL protein; degrades HIF-1α
Li-Fraumeni syndrome Malignant gliomas TP53 17p13 p53; maintains genomic stability
Cowden syndrome Meningiomas PTEN 10q23 PTEN; lipid phosphatase, counters PI3 kinase activation
Gorlin syndrome (nevoid basal Medulloblastomas PTCH 9q22 Cell surface receptor; regulates normal brain
cell carcinoma syndrome) development
Turcot syndrome Medulloblastomas APC 5q21 APC; part of Wnt/β-catenin signaling pathway
Malignant gliomas hMLH1 3p21 Involved in mismatch repair
Malignant gliomas PMS2 7p22 Involved in mismatch repair
Familial retinoblastoma Pineoblastomas RB 13q14 Rb protein; regulates entry into S phase
Ataxia-telangiectasia CNS lymphoma ATM 11q22-23 ATM protein; involved in DNA damage sensing
Multiple endocrine neoplasia Pituitary adenomas MEN1 11q13 Menin
syndrome 1

APC, Adenomatous polyposis coli; ATM, ataxia-telangiectasia mutated; CNS, central nervous system; GTPase, guanosine triphosphatase.

is seen (see Table 63.3). This loss of neurofibromin leads to the increased
TUMOR BIOLOGY Ras activation seen in NF1-associated astrocytomas. Pituitary adenomas
Cell Proliferation often show activation of the alpha subunit of the large heterotrimeric
Gs protein, resulting in mitogenic signaling.
Normal cells rely on growth factors secreted in their local environment The Wnt pathway has been shown to play a role in glioma
to stimulate their growth. However, many CNS tumors have developed tumorigenesis. Investigators have shown that the Wnt-specific secretory
the ability to express their own growth factors along with the respective protein Evi is overexpressed in astrocytomas.40 Furthermore, the same
receptors, resulting in an autocrine loop that allows for self-stimulation.31 researchers showed that the depletion of this protein in glioma cells
Platelet-derived growth factor receptor alpha (PDGFRα), for example, leads to decreased cell proliferation and apoptosis. In addition, silencing
is overexpressed in all grades of astrocytomas, but only higher-grade of Evi in glioma cells leads to reduced cell migration and the ability
tumors overexpress the ligands PDGFA and PDGFB.32 Insulin-like to form tumors in vivo.40 Involvement of Wnt/β-catenin signaling in
growth factors and their receptors both are expressed in brain tumors, gliomas has been reviewed by Zhang and colleagues.41
including gliomas and meningiomas.33 The epidermal growth factor Cell proliferation is intimately tied to cell cycle regulation. Mutations
receptor (EGFR) is amplified or overexpressed in 50% of glioblasto- in the p16/Cdk4 or Cdk6/cyclin D/Rb pathway or the p21/p53/
mas,31 and expression of transforming growth factor–α (TGF-α), a Mdm2/p19ARF pathway are common in many brain tumors, particularly
ligand that binds to this receptor, is increased in many gliomas.34,35 gliomas.
Both scatter factor (also known as hepatocyte growth factor) and its
receptor c-Met are expressed in gliomas, with the highest level of Invasion
expression seen in the most malignant tumors.36
As a result of increasing expression of receptors and ligands, increased Many brain tumors, particularly gliomas, display an invasive phenotype
signaling occurs in many brain tumors, resulting in activation of many with infiltration of tumor cells into surrounding tissues, making a
different pathways that are important in proliferation (reviewed by cure very difficult to achieve. In fact, tumor recurrence was reported
Rao and James34). The best studied of these pathways is the mitogen- in one case even after resection of the entire hemisphere in which a
activated protein kinase (MAPK) pathway, which involves Ras and glioma was located.42 The process of invasion involves several steps:
Raf. Another pathway that has attracted much attention is the detachment of the invading cell from the primary tumor mass, adhesion
phosphatidylinositol 3 (PI3) kinase pathway, which leads to activation to extracellular matrix (ECM), degradation of ECM, and cell motility
of Akt. Mutation of PTEN, which occurs in 30% to 40% of glio- and contractility.43 The details of the mechanisms of glioma invasion
blastomas, also can lead to increased Akt activation.37 are only beginning to be understood. Numerous molecules associated
Ras activation commonly is seen in human astrocytomas and with invasion have been found to be upregulated in gliomas, including
neurofibromas despite the fact that these tumors rarely contain Ras tenascin-C, secreted protein acidic and rich in cysteine (SPARC),
mutations. In astrocytomas, Ras activation probably occurs through various integrins, and matrix metalloproteinases (reviewed by Demuth
activation of growth factor receptors such as EGFR and PDGFR.38 and Berens44).
Other mechanisms of activation of aberrant G proteins have been Some controversy exists regarding deregulated Rho guanosine
identified in other CNS tumors (reviewed by Woods and coworkers39). triphosphatase (GTPase) signaling in brain tumors. For example,
In NF1, loss of expression of neurofibromin, an inactivator of Ras, although RhoA and RhoB are reduced in some astrocytic tumors,
912 Part III: Specific Malignancies

some studies report Rho-dependent lysophosphatidic acid–induced Stem Cells


migration in glioma cells. Expression of another member of the
Rho subfamily, Rac1, has been found to be reduced in astrocytic The tumor stem cell hypothesis may help explain the resistance of
tumors; in addition, this protein has been shown to be overex- glioblastoma to current therapies. The tumor stem cell hypothesis
pressed and to have induced invasion in medulloblastoma tumors. suggests that a subset of tumor cells, called tumor stem cells, stemlike
The role of Rho GTPases has been reviewed by Khalil and cells, or tumor-initiating cells, are endowed with characteristics of normal
El-Sibai.45 stem cells, such that they can self-renew and are able to produce a
variety of cell types. Because the stemlike cells have properties different
Angiogenesis and Hypoxia from the cells constituting most of the tumor, a different approach
to eradicating these stemlike cells may be necessary.
For a tumor to grow beyond a certain size, it must develop a blood Identification of glioblastoma stemlike cells has proved to be
supply. The vasculature of normal brain, which is composed of endo- challenging. Early studies showed differences between CD133+ and
thelial cells, pericytes, and astrocytes, is highly specialized. Together CD133− glioma cells, including differences in tumor-forming capacity
these cells form the blood-brain barrier, which selectively restricts in xenografts.65 However, other studies have shown that CD133− stem-
exchange of molecules between the intracerebral and extracerebral like cells may also be capable of forming tumors, although these two
circulatory systems. As primary brain tumors or brain metastases grow, cell populations exhibit distinct molecular profiles and growth
the integrity of the blood-brain barrier is compromised.46–49 characteristics.66
A number of growth factors are known to be important in angio- Efforts are being made to elucidate transduction pathways that
genesis. The most prominent of these is vascular endothelial growth influence tumorigenicity and are unique to glioma stemlike cells to
factor (VEGF), which is overexpressed in many brain tumors. In one allow treatments to target these pathways. It has been demonstrated
study, increasing VEGF expression correlated with increasing malignant that glioma stemlike cells may be relatively radioresistant as a result
grade in astrocytomas, oligodendrogliomas, and ependymomas.50 In of overexpression of DNA damage repair proteins, including the
this study it also was found that increased expression of the VEGF checkpoint kinases Chk1 and Chk2.67 The cells may be made more
receptors Flt-1 and KDR in tumor vasculature correlated with increasing radiosensitive by using specific inhibitors of Chk1/Chk2. Piccirillo
VEGF expression and malignant grade. Hypoxia and acidosis have and colleagues68 showed that CD133+ glioblastoma stem cells have
been shown to independently regulate VEGF transcription in brain a functional bone morphogenetic protein receptor pathway. By exposing
tumors in vitro and in vivo.51 In addition, both tumor suppressors mice implanted with orthotopic glioblastomas to bone morphogenetic
and oncogenes, hormones, cytokines, and other signaling molecules protein 4, these researchers were able to cause the CD133+ cells to
have been shown to regulate VEGF expression.49,52–55 Growth factors differentiate toward glial cells and reduce tumor growth. These two
other than VEGF that may play a role in angiogenesis in gliomas strategies have the potential to be used to target stem cells in glio-
include members of the TGF-β family, PDGF, placental growth factor, blastomas. Another potential strategy is to target the tumor stroma,
basic fibroblast growth factor, and scatter factor/hepatocyte scatter which provides a specialized niche for tumor cells. Calabrese and
factor.56 Angiogenesis can also be negatively regulated by factors such associates69 showed that stem cells in brain tumors occupied a peri-
as thrombospondins 1 and 2 (TSP1 and TSP2). TSP1 is positively vascular niche and could be targeted by antiangiogenic therapy.
regulated by p53, and thus loss of p53, which commonly occurs Other potential targets include signal transducer and activator of
in gliomas, can lead to decreased TSP1 expression and increased transcription 3 (STAT3), a transcription factor activated by interleukin
angiogenesis. (IL)-6, a molecule that is overexpressed in glioblastoma. STAT3 is
Angiogenesis is particularly prominent within glioblastomas. Among constitutively active in numerous cancers, including glioblastoma.
the characteristic features of these tumors are endothelial proliferation Expression of a dominant negative mutant STAT3 or silencing STAT3
and neovascularization. A variety of growth factors that can increase with a lentivirus have been shown to reduce proliferation of a glio-
angiogenesis are expressed by glioblastomas, with the foremost being blastoma cell line.70 Other groups are investigating nanotechnology
VEGF. VEGF, also known as vascular permeability factor, is a potent to target glioma stemlike cells. Some investigators have used iron
inducer of capillary permeability. The high levels of VEGF expression oxide nanoparticles conjugated to a glioblastoma-specific target of
in glioblastomas may be responsible for the edema associated with EGFRvIII in an animal model,71 whereas others have used curcumin
these tumors. Some evidence indicates that genetic changes common nanoparticles to inhibit glioblastoma and medulloblastoma cell prolifera-
to glioblastomas, such as EGFR activation and PTEN mutation, may tion in culture.72 Glioblastoma stemlike cells have been reviewed in
contribute to high levels of VEGF expression, perhaps through activa- detail by Nduom and colleagues73 and by Tabatabai and Weller.74 The
tion of the PI3 kinase pathway.57–59 stemlike cell hypothesis also applies to other brain tumor histologic
Despite expressing high levels of VEGF, glioblastomas contain types. The roles of medulloblastoma and ependymoma tumor-initiating
significant regions of hypoxia, which may be a cause of treatment cells have been reviewed by Manoranjan and colleagues75 and Poppleton
resistance. Hypoxia has been shown to be present in malignant gliomas and Gilbertson, respectively.76
with both polarographic needle electrode measurement60,61 and binding
of the 2-nitroimidazole EF5.62 Hypoxia, as measured by binding of
EF5, has been shown to correlate with more rapid tumor recurrence.62 CLINICAL PRESENTATION
Evidence indicates that hypoxia is involved in many aspects of tumor Pathophysiology of Signs and Symptoms
growth, including induction of angiogenesis, increased invasion and
metastases, resistance to chemotherapy, resistance to radiotherapy, Parenchymal brain tumors produce clinical signs and symptoms by
increased brain tumor stem cell population, and stem cell genetic three main mechanisms, each of which has important implications
instability.63 for therapy. First, infiltration along nerve fiber tracts is typical of
The histologic features of glioblastoma are consistent with this primary brain tumors such as low-grade astrocytomas and oligoden-
notion of hypoxia, including the presence of pseudopalisading necrosis drogliomas. The first clinical manifestation may be seizures. Normal
and proliferative blood vessels. Pseudopalisades seen within glioblas- brain tissue may be present within areas appearing abnormal on
tomas represent a wave of tumor cells actively migrating away from magnetic resonance imaging (MRI), and functional mapping may be
central hypoxia and are thought to arise as a result of vasoocclusion necessary for safe resection of these slowly growing tumors.77–80 Second,
and intravascular thrombosis.64 High VEGF levels and robust neo- displacement of brain tissue with production of vasogenic edema is
vascularization may be explained by hypoxia-induced VEGF simulation typical of cerebral metastases. Such tumors sometimes can be resected
of new vessels. or irradiated focally with less risk to adjacent normal brain tissue than
Cancer of the Central Nervous System • CHAPTER 63 913

is the case with infiltrating tumors. Third, rapidly growing aggressive palsy. The posterior cerebral artery may be compressed against the
tumors such as high-grade astrocytomas may enlarge as a mass and tentorium, leading to homonymous hemianopia. Brainstem compression
also destroy surrounding neuropil to such an extent that surgical can cause compression of the contralateral cerebral peduncle against
resection, although helpful for the reduction of mass effect and intra- the edge of the tentorium, causing what is known as the Kernohan
cranial pressure (ICP), may not alleviate local symptoms and signs. notch phenomenon.81 Patients with uncal herniation initially may
Intracranial neoplasms usually produce progressive symptoms. be awake, but progression to obtundation, coma, and death may
Location and rate of growth determine both general and specific occur rapidly.
localizing symptoms and signs. Thus a patient with a low-grade glial The central herniation syndrome results from tumors that arise
tumor may have seizures for many years or may exhibit behavioral along the midline axis of the brain, especially those deep in the basal
alteration for many months before focal signs develop. With a more ganglia and thalamus regions. The initial signs and symptoms are due
aggressive glial neoplasm, headache and focal signs may develop over to diencephalic compression. The syndrome often first manifests with
a few weeks. Apoplectic onset is associated with hemorrhage or the an alteration in the level of alertness and behavior. Some patients
development of hydrocephalus. become agitated, whereas others become very drowsy. Hemisensory
Brain tumor symptoms may be general, localizing, or falsely local- or hemiparetic deficits and periodic Cheyne-Stokes respirations may
izing. General symptoms include headache, lethargy, nausea, vomiting, develop in these patients. Initially with diencephalic compression,
and nonspecific cognitive or balance difficulties. These symptoms may pupils are small (1–3 mm), but as the syndrome progresses to compress
be manifestations of increased ICP from a combination of expanding the midbrain and upper pons, the pupils dilate moderately and fix at
tumor volume and the production of associated vasogenic cerebral midposition (3–5 mm). As the syndrome progresses, patients become
edema. Tumors also may cause raised ICP by obstruction of the progressively lethargic and apathetic and Cushing’s signs of hypertension
ventricular system or blockage of the venous sinuses. Abrupt headache and bradycardia may develop as a result of direct compression of the
and exacerbation of neurologic signs may accompany the plateau hemodynamic control nuclei within the brainstem.82,83
waves of sudden increased ICP. Tonsillar herniation may be caused by an expanding mass in the
Sustained ICP in excess of 200 mm H2O causes brain shifts that posterior fossa. Tonsillar herniation results in the cerebellar tonsils
can displace brain tissue through fixed intracranial openings, producing being pushed through the foramen magnum. The syndrome is character-
life-threatening herniation syndromes (Fig. 63.2). The uncal herniation ized by posterior headache, vomiting, stiff neck, and sometimes
syndrome is caused by tumors arising in the lateral aspect of the opisthotonic posturing. Other features may include dysconjugate eye
brain, most commonly the temporal lobe. An early, consistent sign movements and syncope with cough or sudden postural change. As
of compressive uncal herniation is a unilaterally dilated pupil due to a result of direct compression of the medulla and its respiratory center,
compression of the ipsilateral third cranial nerve, which is followed by irregular breathing and acute apnea may develop.
extraocular movement abnormalities consistent with an oculomotor These herniation syndromes can rapidly progress from the onset
of symptoms to death. They can be precipitated by medical procedures.
Lumbar puncture may lead to tonsillar herniation, and ventriculostomy
may result in upward herniation in which the brainstem is forced up
through the tentorial notch. A high index of suspicion is required to
Falx cerebri successfully diagnose and treat this condition with emergency intubation
and administration of appropriate therapy (see the section on treatment
Cingulate of brain tumor symptoms).
gyrus

General Signs and Symptoms


Headache results from traction on pain-sensitive structures of the
intracranial contents, including the large cerebral vessels, the dura
and meninges, the venous sinuses, cranial nerves V and IX, and the
second and third cervical nerve roots. Headache is the most common
symptom of a brain tumor and occurs in approximately 50% of
patients at some time during the course of the illness. Headache more
frequently accompanies rapidly growing than slowly growing tumors.
Third
ventricle Tumors located in neurologically noneloquent brain areas such as the
nondominant frontal and temporal lobes may manifest with headache
as the sole clinical manifestation. The “classic” brain tumor–associated
headache often is worse in the morning and lateralized to the affected
side of the brain. Brain tumor–associated headache may be worsened
Hippocampal
gyrus by coughing or straining. A majority of patients with brain tumors,
Tentorium however, do not have these classic symptoms but instead have headaches
cerebelli that are deep, aching, and difficult to distinguish from tension-type
headache. Headaches from posterior fossa tumors are localized to the
back of the head or neck, whereas headaches from tumors of the
anterior and middle cranial fossae may be referred to the forehead or
Foramen Cerebellar eye, at times being misconstrued as a sinus headache.
magnum tonsil Brain tumor–associated cognitive changes initially may be subtle
and frequently are misdiagnosed as depression or, in the older patient,
Figure 63.2 • Intracranial herniation syndromes evoked by supratentorial age-associated forgetfulness or early Alzheimer disease. Frontal lobe
masses. The tumor and its edema (arrows) have produced the following (curved
arrows): cingulate gyrus herniation under the falx cerebri; diencephalic herniation tumor location is the most common site for tumors producing these
across the midline compressing the ipsilateral ventricle and producing the symptoms as the initial manifestation of a neoplasm.
hydrocephalus in the contralateral ventricle; hippocampal gyrus herniation Several other symptoms may emerge that roughly parallel cognitive
through the tentorial notch compressing the posterior cerebral artery and decline and are of poor localizing value by themselves. Dysphagia is
brainstem; and herniation of the cerebellar tonsils through the foramen magnum. a common complaint, and caregivers may note that the patient with
914 Part III: Specific Malignancies

cognitive impairment seems to take a long time to chew and swallow. Tumors in the occipital lobes cause a contralateral quadrantic or
Oral candidiasis is a potential complicating issue in patients undergoing hemianopic defect, sometimes with sparing of central macular vision.
long-term corticosteroid therapy, and appropriate treatment should Tumors of the brainstem cause a great many focal deficits early in
be instituted. Similarly, incontinence tends to parallel the degree of their course. Typically, a combination of cranial nerve palsies and long
cognitive impairment. Urinary retention may occur in patients with tract signs such as hemianesthesia or hemiparesis coupled with ataxia
bifrontal brain disease or with spinal cord problems. Opiate medications reflecting cerebellar involvement give a clue to the localization. Patients
may exacerbate the urologic problems. with brainstem tumors experience difficulty with swallowing and speech
Seizures occurring for the first time in adults are more likely to be articulation and are at risk for aspiration.
due to focal brain pathology, particularly neoplasms, than are seizures Cerebellopontine (CP) angle tumors such as acoustic neuromas
occurring in childhood. Intracranial tumors produce generalized impair function of the eighth cranial nerve and produce unilateral
tonic-clonic seizures, secondarily generalized tonic-clonic seizures, and hearing loss, tinnitus, and, later, vertigo. Involvement of adjacent
partial, localization-related seizures. Seizures are more common in cranial nerves VII and V leads to facial palsy and facial anesthesia.
persons with cortical tumors than in persons with infratentorial or Later cerebellar dysfunction reflects tumor growth in this area.
deep thalamic lesions, and they occur more often in low-grade Tumors of the pituitary and suprasellar region produce endocri-
astrocytomas, oligodendrogliomas, or oligoastrocytomas (60%–80%) nologic abnormalities either by hormonal production by secretory
than in high-grade gliomas (20%–40%) or cerebral metastases adenomas or by impingement on hypothalamic-pituitary connections.
(15%–20%)84,85 Seizures occur at some time in 25% to 50% of patients Visual defects reflect chiasmatic involvement. The most common
with brain tumors. Seizure frequency may increase during radiation pituitary region field defect is a bitemporal hemianopia.
treatment and continue at an increased frequency for several months Pineal region tumors (e.g., germ cell tumors, pineocytomas,
thereafter because of localized swelling. pineoblastomas, gliomas of this region) may compress the aqueduct
Papilledema—that is, swelling of the optic nerve head with engorge- of Sylvius, causing hydrocephalus. Compression of the pretectal area
ment of retinal veins—usually indicates raised ICP. Currently, papil- produces a characteristic syndrome (Parinaud syndrome) with paralysis
ledema is seen in fewer than 20% of patients at presentation, down of upgaze, ptosis, and loss of pupillary light reflexes, along with
from 59% in the 1971 report by Huber.86 The development of retraction-convergence nystagmus.
papilledema is dependent on the location of the tumor and the rate Many primary tumors are capable of diffuse infiltration of the
of growth. meninges. Gliomas, lymphomas, and oligodendrogliomas all may
Vomiting with or without nausea may occur as a result of direct invade the subarachnoid space. They produce variable cranial nerve
simulation of emetic centers in the floor of the fourth ventricle. This and spinal root dysfunction and diffuse headache, and sometimes lead
mechanism explains the nausea and vomiting seen with raised ICP, to communicating hydrocephalus. Elevated levels of cerebrospinal
particularly when the rise in pressure has been rapid and is associated fluid (CSF) protein, low levels of CSF glucose, and positive results
with hemorrhage or herniation. Patients with posterior fossa tumors on cytologic studies are the diagnostic hallmarks.
frequently experience nausea and vomiting. More commonly, however, On occasion, the clinician will be faced with symptoms that appear
nausea is a nonlocalizing symptom, and thus the differential diagnosis to give a clue to the patient’s tumor site but in fact are false localizing
must include adverse drug reactions from antiepileptic drugs (AEDs), symptoms. Palsies of the abducens nerve (cranial nerve VI) may reflect
analgesics, or other concurrent medications and gastritis from high-dose brainstem involvement but commonly are a nonlocalizing sign of
corticosteroid treatment. raised (or, much less commonly, low) ICP. Ocular pain is of little
localizing value because it may reflect any of the structures innervated
Localizing Signs of Intracranial Tumors by the first division of the fifth cranial nerve. Thus eye pain may
reflect any process in the anterior or middle cranial fossa. Diplopia
Focal clinical signs of an intracranial tumor reflect the location of the may result from cranial nerve invasion by brainstem or leptomeningeal
mass and its associated vasogenic edema, which often is of much tumor but also can be caused by excess levels of AEDs.
greater volume. This chapter does not provide a detailed discussion Posterior head pain may reflect posterior fossa disease but also may
of every possible localizing sign but rather focuses on the general come from the upper cervical segments, and spinal cord tumor or
principle of neurologic localization and the specific emergent syndromes caudal extension of a primary brainstem tumor should be considered
that should be recognized because of their localizing and management if the patient reports pain in the occiput. Another sign of cervical
importance. cord disease is bilateral upper limb weakness or numbness.
Disorders associated with frontal lobe lesions include early impair- Gait disorders pose another potential hazard for falsely localizing
ment of intellectual function and language function if the dominant signs and symptoms. A frontal lobe gait ataxia may mimic basal ganglia
frontal lobe is involved. Patients with bilateral frontal tumors appear or even cerebellar disease. The affected patient walks slowly, with a
to lack initiative and spontaneity—a state called abulia. Such patients wide-based gait, and seems to have difficulty initiating movements.
may have an impaired gait with difficulty initiating walking. Personality Proximal leg weakness may reflect spinal metastases from intracranial
changes include inattentiveness, apathy, depression, and the less or systemic tumor, but probably the most common cause of proximal
common disinhibition and inappropriate affect leading to socially leg weakness is corticosteroid-induced myopathy.
inappropriate behaviors. As tumors enlarge to involve the motor cortex,
contralateral motor problems, such as hemiparesis or monoparesis, Treatment of Brain Tumor Symptoms
may develop.
Receptive language, auditory discrimination, and memory all are Acute Raised Intracranial Pressure
important functions of the dominant temporal lobe. Patients with The most immediate life-threatening syndromes are the herniation
nondominant temporal lobe tumors may have seizures involving visual, syndromes, but rapid increase in vasogenic edema also mandates
olfactory, or gustatory hallucinations. Deep temporal tumors may aggressive treatment of evolving neurologic signs and symptoms.
cause a contralateral visual field cut (superior quadrantanopia). If the patient’s condition is rapidly deteriorating, intubation and
The parietal lobe is demarcated from the frontal lobe by the central hyperventilation aiming for a pressure of carbon dioxide (Pco2) of
sulcus. Parietal lobe functions include tactile perceptions; integration 25 to 30 mm Hg are required. Mannitol is administered intravenously
of sensory, visual, and auditory information; and visual discrimination at a loading dose of 1 to 2 g/kg, followed by 0.5 to 1 g/kg every 6
in the inferior contralateral quadrant. When tumor involves the hours as needed to control ICP. Mannitol may be transiently effective,
nondominant parietal lobe, inattention to the deficit (anosognosia) but a rebound pressure increase sometimes develops after 24 to 48
may be a prominent feature of the presentation. hours. Intravenous dexamethasone at a loading dose of 10 mg followed
Cancer of the Central Nervous System • CHAPTER 63 915

by 4 to 10 mg every 6 hours is also appropriate initial treatment, In view of all of the potential hazards of AEDs, it is worthwhile
but the full effect of corticosteroid therapy takes 24 to 72 hours. to consider whether their prophylactic use is justified in the brain
Although it is frequently given every 6 hours, dexamethasone has tumor population with several studies showing mixed results regarding
a long half-life and can be given in a twice-daily dosing regimen. efficacy.97–99 For all of these reasons, a practice parameter published
Electrolytes and glucose levels must be monitored, and gastritis pro- by the Quality Standards Subcommittee of the American Academy
phylaxis is required. Acute neurosurgical interventions include ICP of Neurology concluded that no benefit could be established for routine
monitoring devices and ventricular drainage or tumor decompression prophylactic use of AEDs in patients with brain tumors.100 However,
in patients with obstruction. Fluid management requires avoidance of the advent of AEDs that do not induce hepatic enzymes has recently
hyponatremia. caused some neurologists to reconsider prophylactic use of seizure
medicines in high-risk patients, particularly those with hemorrhagic
Chronically Increased Intracranial Pressure metastases, such as patients with melanoma.
Dexamethasone in doses of 8 to 40 mg per day upregulates angiopoietin Of the newer agents, levetiracetam (Keppra) has emerged as a first
1 to stabilize the blood-brain barrier and downregulates VEGF.87–89 line antiepileptic agent in patients with brain tumors. Its therapeutic
Controlling the edema also helps to control headache, nausea, and window is wider than that of phenytoin, as the level is not affected
seizures. Because chronically raised ICP can cause visual loss, careful by chemotherapy drugs, and it does not alter the metabolism of any
ophthalmologic follow-up evaluation with visual field assessment is chemotherapeutic agents. Transitioning from phenytoin to levetiracetam
essential. Acetazolamide therapy for symptomatic plateau waves has in glioma-related seizures did not elicit significant differences in seizure
been useful for some patients with raised ICP from intracerebral or freedom, suggesting safety in switching to levetiracetam.101,102 However,
leptomeningeal tumor.90 the clinician must be familiar with some adverse effects specific to
In the doses required to treat cerebral edema, corticosteroids produce these drugs (summarized in Table 63.4) to be able to diagnose symptoms
many adverse effects that range from the easily managed gastritis accurately, eliminate the offending drug, and avoid unnecessary
symptoms and glucose intolerance to insomnia, steroid psychosis, diagnostic and therapeutic interventions. One of the most common
intractable hiccoughs, and disabling myopathy. The psychosis may adverse side effects of levetiracetam is irritability and aggression.
readily respond to reduction of the steroid dose and the addition of Lacosamide is an alternative first-line103 or add-on therapy104 to
neuroleptic agents. Treatment of the steroid myopathy, however, will levetiracetam with similar efficacy. Use of lacosamide as monotherapy
require weeks of physical therapy with attempts at steroid reduction. or in combination with levetiracetam may be considered in patients
Anecdotal reports suggest that substitution of a nonfluorinated steroid who are experiencing adverse side effects.
such as methylprednisolone for fluorinated steroids such as dexa-
methasone may help minimize the weakness.91 Deep Venous Thrombosis
Many patients with brain tumors are treated with corticosteroids Deep venous thrombosis (DVT) is extremely common in patients
for prolonged periods and are thus susceptible to infection, particularly with brain tumors, with a reported incidence of 28% to 45%.100,105
with Pneumocystis jiroveci, which carries a 50% mortality rate. Among The risk is greatest in the postoperative period and in patients with
solid tumors, primary and metastatic brain tumors have the highest hemiplegia.
rate of Pneumocystis infection, which occurs in 2% of patients and For prophylaxis, low-molecular-weight heparin drugs have a good
proves fatal to 40% of these patients.92 The mean duration of steroid safety profile in the neurosurgical population.106 Prophylaxis with
therapy before infection was 7 months, with a mean dexamethasone enoxaparin, 40 mg, administered subcutaneously daily, plus use of
equivalent dose of 1 to 2 mg per day.93 In view of these statistics, external compression stockings has been found to be superior to the
prophylaxis with one double-strength tablet of trimethoprim- use of compression stockings alone for the prevention of venous
sulfamethoxazole three times a week should be provided for patients thromboembolism after neurosurgery.107
with brain tumors during steroid administration and for 1 month Patients with brain tumors who have diagnosed DVT or pulmonary
afterward. Alternatives for patients who are allergic to trimethoprim- embolism (PE) will often receive inferior vena cava filters to prevent
sulfamethoxazole include aerosolized pentamidine, dapsone, or ato- further thromboembolic complications. However, retrospective studies
vaquone. Withdrawal of corticosteroids after use for more than 2 have demonstrated a low risk of hemorrhage relative to a 60% rate
weeks may result in an adrenal insufficiency state with symptoms of of complications. Complications included PE, filter thrombosis, and
lethargy, hypotension, electrolyte imbalance, arthralgias, and diffuse postphlebitic syndrome.109
weakness. Chronic glucocorticoid supplementation with hydrocortisone, Although no absolute guidelines are available, most neurosurgeons
10 to 20 mg per day, is essential for these patients.94,95 A VEGF would be reluctant to institute full anticoagulation in these patients
inhibitor such as bevacizumab or cediranib may provide an alternative within 2 weeks of surgery. Because most primary and secondary brain
treatment for controlling peritumoral edema.96 tumors pose a continuing risk of thromboembolism, lifelong antico-
agulation is recommended.
Seizures
Neurooncologists often find themselves working closely with epileptolo-
gists as they seek to control seizures with minimal adverse effects.
Because patients with brain tumors frequently require corticosteroids,
analgesics, anxiolytics, and antiemetics, drug-drug interactions make Table 63.4 Adverse Effects of Antiepileptic Drugs:
the management of epilepsy complex. Special Issues in Patients With Brain
The potential interaction of AEDs with other medications required Tumors
for patients who have a brain tumor has led to reconsideration of the
prophylactic use of antiseizure drugs. Cytochrome P450 enzyme– Drug Potential Adverse Effect(s)
inducing drugs such as phenytoin and carbamazepine are now used Oxcarbazepine Hyponatremia (confusion, seizures)
less frequently because of their side effects. Gabapentin Sedation, ataxia, weight gain
A rash develops in approximately 20% of patients with gliomas Depakote Weight gain, platelet dysfunction
who are treated with phenytoin or carbamazepine and cranial irradiation;
Topiramate Memory and word-finding problems, weight loss
the most serious and sometimes life-threatening reaction, Stevens-
Johnson syndrome, occurs in a few patients. Treatment is controversial, Zonisamide Sedation
but often the corticosteroid dose is doubled as AEDs are withdrawn Levetiracetam Psychosis, irritability, lethargy
abruptly.
916 Part III: Specific Malignancies

No clear-cut guidelines exist for the use of direct thrombin inhibitors Multiple sequences may be obtained from the MRI scans, highlight-
in patients with brain tumors, and the lack of agents to reverse ing different properties of both brain and tumor. Of particular interest
anticoagulation is a drawback in this population of patients who may are the T2-weighted and fluid-attenuated inversion recovery (FLAIR)
need urgent surgical intervention. However, the recent development images. These sequences highlight tumor-related vasogenic edema
of new reversal agents may lead to changes in practice patterns, given and nonenhancing portions of the tumor, which appear as areas of
the inherent decreased risk of intracranial hemorrhage with direct increased T2 signal compared with normal brain. Evaluating this signal
thrombin inhibitors. pattern is very important in assessing the degree of mass effect, because
the increased signal may represent the true limits of a glial tumor, as
tumor cells are invariably present in the nonenhancing tissue sur-
DIAGNOSTIC IMAGING rounding the enhancing component.110 This phenomenon provides
Magnetic Resonance Imaging the rationale for extending the irradiated zone in standard external
beam radiation therapy to include the edematous volume of brain
MRI is the study of choice for evaluating brain tumors. The increased plus a margin. The presence of an edema pattern extending into the
tissue contrast resolution provides exquisite anatomic definition, which corpus callosum implies direct tumor invasion into the corpus callosum.
is accentuated after administration of gadolinium contrast material. The extremely compact fiber tracts passing through the corpus callosum
Therefore when possible, MRI sequences should be performed before impede passage of edematous fluid, and thus any increased T2 signal
and after administration of contrast material. Much like with the use that is present is likely created by tumor infiltration. This point is
of iodinated contrast material in computed tomography (CT) imaging, very important to keep in mind in assessing the extent of tumor.
tumor-related disruption of the blood-brain barrier allows interstitial Endothelial proliferation with neovascularity is a hallmark of
leakage of contrast material, and depending on scanning parameters, malignancy in brain neoplasms. Both MRI and CT perfusion imaging
it may appear as increased (spin echo T1) or decreased (gradient echo can be performed in combination with conventional MRI examinations
imaging) signal. Gadolinium enhancement can be crucial in identifying to determine the extent of neovascularity. The correlation between
leptomeningeal disease, which appears as thickened, enhancing areas relative cerebral blood volume (rCBV) and grade of malignant glioma
of the dura and leptomeninges. The absence of bony artifacts common appears to be very good.112 Perfusion imaging with dynamic susceptibil-
with CT has enabled MRI to provide exceptional information regarding ity contrast-enhanced MRI is especially helpful in distinguishing grade
brain tumors, particularly in the posterior fossa. In addition, localization III from grade IV malignant gliomas, but it can also be used to dis-
of the lesion is precise because MRI is usually reconstructed in three tinguish gliomas from other neoplasms such as intracranial metastases
orthogonal planes. (Fig. 63.3).113,114 It has been shown that anaplastic astrocytomas that

A B C

D E F
Figure 63.3 • Differentiation of tumor types with perfusion magnetic resonance imaging (MRI). Axial T1-weighted postgadolinium magnetic resonance
image demonstrating a metastasis from a lung carcinoma in one patient (A) and glioblastoma multiforme in another (D). Respective axial T2-weighted images
demonstrate moderate edema with questionable involvement of the corpus callosum genu by the metastasis (B) and splenium of the corpus callosum by the
glioblastoma (E). Perfusion magnetic resonance image demonstrates hypovascularity in the peritumoral region, with only a minor increase in perfusion at the
rim of enhancing tumor (C, white arrow), consistent with vasogenic edema without infiltrating tumor, compared with (F), which demonstrates increased perfusion
in the peritumoral region, implying hypervascularity consistent with infiltrating high-grade glioma. (From Law M. Perfusion and MRS for brain tumor
diagnosis. In: Edelman RR, Hesselink JR, Zlatkin MB, Crues JV, eds. Clinical Magnetic Resonance Imaging. Vol. 3. 3rd ed. Philadelphia: WB Saunders; 2006.)
Cancer of the Central Nervous System • CHAPTER 63 917

enhance have a higher vascularity index than do nonenhancing metabolites provides the most useful information. N-acetylaspartate
anaplastic astrocytomas. Perfusion imaging may enable monitoring (NAA) is a marker of healthy neurons, whereas increases in choline
of the antiangiogenic effects of treatment in brain tumors. Furthermore, are associated with high cellular turnover. Creatine serves as an
perfusion imaging is also useful in determining vascularity in benign internal marker and is related to the energy state of the tumor. A
tumors such as meningiomas, a phenomenon that may be associated lipid peak indicates the presence of necrosis and suggests higher-grade
with disease progression.115 malignancy, whereas lactate indicates the presence of hypoxia.121,122
Functional MRI techniques have been used to identify areas of Malignant gliomas exhibit a higher choline peak relative to creatine
brain activity in real time, which is quite useful in identifying regions and a lower NAA peak relative to choline, with increasing grade of
of eloquent brain adjacent to tumors.116,117 By identifying language malignancy (Fig. 63.4). Lipid and lactate peaks, which also constitute
and motor areas adjacent to tumors, one can determine the extent of markers of malignancy, can be seen both in primary gliomas and in
surgical resection that can be carried out while attempting to preserve metastatic tumors. MRS can help determine whether an enhancing
functional activity.118 It is now possible to import functional MRI region in a previously treated tumor bed represents active tumor or
images into frameless stereotactic units that are used for intraoperative radiation necrosis. MRS is also used to monitor treatment effects over an
surgical planning.119 Through use of discrete motor, sensory, language, extended period.
or visual paradigms, the functional activity of relevant cortical brain MRI has the capability to specifically identify vascular structures.
can be imaged. One limitation to functional MRI scanning is its As noninvasive techniques, magnetic resonance angiography (MRA)
inability to identify subcortical white matter tracts adjacent to the and magnetic resonance venography can be useful in evaluating major
gray matter activated by the investigated activity. For this reason, vessels at the base of the skull in preparation for surgical treatment
diffusion tensor imaging (DTI) is increasingly being used to map of skull base tumors.
subcortical white matter tracts. It has been shown that combining
DTI with conventional anatomic images of the lesion of interest Computed Tomography
facilitates extended resection of tumor while preserving function.120
A great deal of interest has centered on the capability of clinical MRI A CT scan usually is the first study obtained in a patient with a
scanners to perform magnetic resonance spectroscopy (MRS) to evaluate neurologic complaint. In part because of its extensive availability and
molecular components of a defined voxel within the brain tissue. rapid acquisition times, CT provides a robust screening tool. Hemor-
Today’s smaller voxel sizes allow a much greater degree of selectivity rhagic lesions can be seen as a result of the increased density of
in evaluating components of a tumor. MRS of malignant gliomas extravasated blood products in comparison with surrounding tissues.
has been evaluated extensively, and the presence or absence of five Infarcts manifest acutely as edematous hypodense tissue and can be

Cho After resection


NAA
Cho Cr

Normal-appearing spectrum Histologically proved Presumed necrosis


tumor

Post contrast Choline NAA NAA + Cho

Figure 63.4 • Magnetic resonance imaging/magnetic resonance spectroscopy (MRI/MRS) studies of World Health Organization grade II oligoastrocytoma.
Axial T1-weighted postgadolinium MRI performed the day before surgery (top left panel) revealed an area of hypodensity that did not enhance (bottom left
panel). MRS image (middle panel) shows an area of high choline (Cho; 1.8 times greater than adjacent brain) and low N-acetylaspartate (NAA) levels, in
addition to a smaller region of no significant metabolite levels, which is presumed to be necrosis. The resection was limited to a region of elevated choline
with no significant Cr, Creatine. (From Vigneron D, Bollen A, McDermott M, et al. Three-dimensional magnetic resonance spectroscopic imaging of histologically
confirmed brain tumors. Magn Res Imaging. 2001;19:93.)
918 Part III: Specific Malignancies

confused with tumors, especially low-grade gliomas, which may not claustrophobia, implanted pacemakers, defibrillators, or other metal
enhance with contrast agents. However, cytotoxic edema associated devices that preclude imaging with MRI.
with infarcts often involves both gray and white matter, whereas
vasogenic edema as a result of the presence of tumor predominately Imaging of Supratentorial Gliomas
involves the white matter. Iodinated contrast agents may be useful to
differentiate tumor from the surrounding edematous brain tissue WHO grade I (pilocytic) astrocytomas show enhancement on CT
because disruption of the blood-brain barrier permits leakage of the and MRI scans. By contrast, WHO grade II astrocytomas typically
contrast agent into the interstitial space. The pattern of ring enhance- are poorly defined, hypodense, or isodense lesions on CT scans and
ment of tumors may be difficult to distinguish from the ring enhance- do not enhance. Either localized or homogeneous enhancement,
ment of a cerebral abscess, although brain tumors are far more common however, can be seen in up to 30% to 40% of cases, with calcification
than brain abscesses in the United States. Infarcted brain tissue may in 5% to 10% of cases.124,125 MRI shows low signal intensity on
also enhance after administration of contrast material. This finding T1-weighted images and high signal intensity on T2-weighted images
is most common 2 to 3 weeks after a stroke, and thus clinical history (Fig. 63.5A–B). Grade III anaplastic astrocytomas and grade IV
should complement image interpretation.123 glioblastomas typically enhance with contrast, although in one study,
CT has limitations in evaluating structures near the irregular bony enhancement was not seen in 40% of the former and in 4% of the
surfaces of the skull base because of volume averaging and streak latter (see Fig. 63.5C–D)126 The region of enhancement typically has
artifact. This limitation makes interpretation in and around the skull a ringlike appearance surrounding an area of necrosis. The perimeter
base and in the posterior fossa (the region between the tentorium and of the enhancing region does not define the border of the tumor, and
the foramen magnum) difficult. Nevertheless, CT is the study of malignant cells may be present beyond this perimeter. T2-weighted
choice in patients who cannot undergo MRI, such as patients with MRI images show abnormalities that are more extensive than those

A B

C D
Figure 63.5 • Magnetic resonance imaging: low-grade astrocytoma versus glioblastoma. (A) World Health Organization (WHO) grade II astrocytoma.
T1-weighted postgadolinium axial image shows no enhancing mass; however, a region of hypodensity is seen. (B) A corresponding fluid-attenuated inversion
recovery (FLAIR) image of the tumor in (A) shows abnormality consistent with edema. (C) WHO grade IV glioblastoma. A T1-weighted postgadolinium
axial image shows the presence of a large mass compressing the right lateral ventricle. A rim of enhancement with central necrosis is typical of these tumors.
(D) A corresponding FLAIR image of the tumor in (C) shows extensive peritumoral edema.
Cancer of the Central Nervous System • CHAPTER 63 919

seen on a contrast-enhanced CT scan or a T1-weighted MRI scan. in Neuro-Oncology, or RANO). First, pseudoprogression is defined as
In one study in which stereotactic biopsy findings were correlated an increase in contrast enhancement or edema on MRI without true
with radiologic findings in patients with gliomas, isolated tumor cells tumor progression, a phenomenon made more common by the addition
often were found as far away as the region showing increased signal of temozolomide to radiotherapy.130,131 The first postradiation MRI
intensity on T2-weighted images.110 Perfusion MRI can measure rCBV image shows increased contrast enhancement in up to 50% of patients
and cerebral blood flow, which can be used to determine tumor grade (Fig. 63.7), in half of whom the enhancement eventually subsides.
and to assess progression after treatment.127 The phenomenon may be asymptomatic but can also cause neurologic
At least 50% of oligodendrogliomas show calcifications, which can decline. Failure to recognize pseudoprogression may cause the physician
be appreciated on plain films of the skull and on CT scans.128 Enhance- to prematurely terminate effective therapy.132,133 Second, pseudoregression
ment of oligodendrogliomas can be seen on CT and MRI scans but refers to a decrease in contrast enhancement on MRI without true
is often mild and poorly defined. response, a phenomenon increasingly seen in patients treated with
VEGF pathway antiangiogenic agents.134 Infiltrative tumor may be
Positron Emission Tomography distinguished from radiation-related FLAIR and T2 signal MRI changes
by diffusion-weighted MRI sequences that show diffusion restriction
Positron emission tomography (PET) imaging uses a radioactive in areas infiltrated by neoplastic cells.135
isotope analogue of glucose, fluorine-18 fluorodeoxyglucose (FDG), Some of the important features of the updated RANO criteria
to image glucose metabolism. Because glucose is the sole fuel for brain that are just now being implemented in clinical trials include precise
tissue, this metabolic imaging allows visualization of brain physiology. definitions of measurable disease, exclusion of patients within the first
Because glioblastomas, untreated PCNSL, oligodendrogliomas, and 3 months after radiotherapy from clinical trials to avoid confounding
malignant meningiomas have increased glucose metabolism compared chemotherapy failure with pseudoprogression, and inclusion of stable
with normal brain tissue, these tumors have greater FDG avidity and or decreased nonenhancing disease as a criterion for tumor response
will be PET positive. Anaplastic astrocytomas, oligodendrogliomas, in patients who have received antiangiogenic therapy.
meningiomas, and metastatic tumors show variable FDG uptake (Fig. Similarly, confusion may arise when neuroimaging documents
63.6), whereas low-grade gliomas and radiation necrosis show little enlargement of a mass after stereotactic radiosurgery (SRS) that may
to no FDG uptake. PET imaging using targeted tracers, such as the progress over many months. Because PET scanning may not reliably
somatostatin-receptor ligand gallium-68 DOTATOC in meningiomas, distinguish radiation necrosis from tumor progression, the results of
might be more useful for tumors in which FDG is too variable for concurrent chemotherapy may be ambiguous. Multicentric recurrence
accurate delineation.129 outside original radiation fields and after temozolomide appears to
be more common when patients have positive O6-methylguanine
Challenges to Imaging Modalities DNA-methyltransferase (MGMT) promoter methylation status.136,137

Complicating assessment of chemotherapy in patients with a brain Lumbar Puncture


tumor is the observation that corticosteroids also often reduce MRI
contrast enhancement and relieve symptoms, making it difficult to Lumbar punctures are not commonly used for the diagnosis of brain
distinguish chemotherapy effects from corticosteroid effects. For almost tumors owing to the low yield of the procedure in primary gliomas.
20 years, standard response criteria have been the Macdonald criteria, Leptomeningeal seeding from supratentorial malignant gliomas is
which use two-dimensional measurements of enhancing tumor and usually an end-stage finding. Once life-threatening conditions such
account for corticosteroid use and clinical status. However, it has been as mass effect have been excluded, lumbar puncture may be performed
recognized that reliance on contrast enhancement is a limitation in for confirmation of leptomeningeal disease. Lumbar puncture remains
many anaplastic gliomas. Two situations in particular have led to the part of the staging workup for some primary brain tumors, such as
development of a new set of response criteria (Response Assessment medulloblastoma and CNS lymphoma.

Figure 63.6 • Positron emission tomography (PET) scan of recurrent glioma. The patient had undergone surgical resection of an anaplastic oligodendroglioma
15 months earlier. Areas of contrast enhancement on T1-weighted magnetic resonance imaging (MRI) scan (left) have high fluorodeoxyglucose (FDG) accumula-
tion, consistent with recurrent high-grade tumor on the coregistered FDG-PET image (right). (From Hagge RJ, Wong TZ, Coleman RE. Positron emission
tomography. Brain tumors and lung cancer. Radiol Clin North Am. 2001;39:874.)
920 Part III: Specific Malignancies

Figure 63.7 • Pseudoprogression after chemoradiation. Gadolinium-enhanced T1 magnetic resonance image before chemoradiation in a patient with
anaplastic astrocytoma (left). First posttreatment scan (right) 8 weeks after completion of radiation and temozolomide therapy shows enlargement of a ring-
enhancing mass that correlated with clinical deterioration. Symptoms and magnetic resonance imaging findings had regressed on the next scan 6 weeks later
(not shown).

SURGERY: GENERAL CONSIDERATIONS including three-dimensional conformal therapy, stereotactic radio-


therapy, and intensity-modulated radiation therapy are being used in
The successful resection of a brain tumor requires removing the tumor a majority of patients with brain tumors. Proton therapy, which is
while minimizing injury to the surrounding normal brain. The approach increasingly available in the United States, may be used for specific
to removal of brain tumors follows the mantra of real estate agents: cases, especially for pediatric brain tumors and for tumors at the base
“location, location, location.” Deep tumors that are in noneloquent of the skull. Heavy ion therapy, such as with carbon ions, is used in
regions of brain can be easily accessed and removed, whereas superficial a few centers worldwide.
tumors may not be easily resectable because of their location within
extremely eloquent brain tissue. Radiation Therapy: Technical Details
The goals of surgery are to (1) establish the histology of the lesion,
which is frequently better achieved via surgical resection over stereotactic Treatment planning starts with a review of the MRI scan to identify
biopsy because greater tissue sampling reduces the risk of sampling the target volumes. The appropriate volume to be irradiated varies
error; (2) debulk the mass effect of the tumor to correct a neurologic according to the tumor type. Most brain tumors including gliomas
deficit and to prevent imminent death in patients with large tumors and meningiomas are treated with focal radiation to the lesion plus
and early herniation syndromes; and (3) achieve surgical cure when a margin. The actual abnormality seen on imaging studies is termed
possible (as in WHO I gliomas) or, when the lesion is infiltrative and the gross tumor volume (GTV). Additional tissue surrounding the
surgical cure is impossible, to debulk the tumor to increase the efficacy GTV that is thought to potentially contain tumor cells is included
of radiation therapy and chemotherapy, which produce the best response in the clinical target volume (CTV). For some tumors such as glio-
rate when they are used with minimal tumor burden. blastoma, which can be highly infiltrative, the CTV includes the area
The most common presenting manifestations in patients with of edema as demonstrated on the FLAIR or T2 sequences plus up to
glioma are headache and seizures.135 Debulking large tumors will reduce a 2.5-cm margin. For other tumors that do not infiltrate, such as
dural stretch, thus decreasing headaches. The incidence of seizures in meningiomas, pituitary adenomas, acoustic neuromas, and cranio-
patients with malignant gliomas can be decreased by at least 75% pharyngiomas, the CTV typically would be much tighter than that
with attempts at gross total resection.136 Recovery from other neurologic used for glioblastomas. An additional volume is included around the
deficits such as hemiparesis, visual field loss, or aphasia will depend CTV to take into account day-to-day setup error and to allow for
on whether the deficit is due to mass effect or due to infiltration and buildup of dose, resulting in the planning target volume (PTV).
damage to neural tracts. Treatment planning begins with CT simulation of the patient with
Another rationale for maximal debulking is to reduce the cellular appropriate immobilization devices. Fig. 63.8 shows thermoplastic
burden in order to increase efficacy of both radiation therapy and mask and bite-block immobilization devices. A previously obtained
chemotherapy following resection. Extent of resection in primary glial MRI image or an MRI image obtained at the time of simulation may
tumors has been found to correlate with survival.138–140 be fused with the CT images in the treatment planning system to
facilitate better target volume delineation. Computerized treatment
RADIATION THERAPY: GENERAL planning is then used to generate radiation beam angles, blocks, and
CONSIDERATIONS dose distributions.
Most brain tumors are treated with focal radiation therapy, and
Radiation is commonly used to treat many different types of brain often brain metastases and some leukemias and lymphomas require
tumors. The radiation oncologist must decide on many factors in the whole-brain radiotherapy; however, for some tumors, such as primitive
treatment plan for an individual patient, including treatment volume, neuroectodermal tumors (PNETs) and metastatic germ cell tumors
dose, fractionation, and normal tissue constraints. Treatment techniques of the CNS, it may be necessary to include the entire craniospinal
Cancer of the Central Nervous System • CHAPTER 63 921

A B
Figure 63.8 • Patient immobilization devices for radiation therapy delivery. (A) Thermoplastic mask head-immobilization device. (B) Bite-block head
immobilization device. (From Gunderson LL, Tepper JE. Clinical Radiation Oncology. 2nd ed. Philadelphia: WB Saunders; 2006.)

axis in the irradiated zone. Proton therapy for craniospinal irradiation limits to a portion of the structure, and the maximum point dose
has the advantage of reducing dose to the vertebral bodies and essentially within the structure. The most common mechanism of delivering this
eliminating the exit dose through the thorax, abdomen, and pelvis.141 dose is use of multiple fields, typically five to nine, with use of a special-
E-Fig. 63.1 shows sample treatment plans for craniospinal irradiation ized blocking apparatus called a multileaf collimator to divide each
using three-dimensional conformal radiotherapy, tomotherapy, and field into multiple beamlets. From 50 to more than 100 beamlets
proton beam techniques.142 may be used.

Stereotactic Radiotherapy Heavy Charged Particle Radiation Therapy


Stereotactic radiotherapy is a technique for delivering high-dose The delivery of charged particles, primarily protons, has been used
radiation to a target volume with very tight margins, thereby sparing to treat brain tumors. The potential benefit of protons is the result
surrounding normal tissues. This technique can be implemented of their physical properties, which are different from those of photons.
with various devices, including (1) a Gamma Knife machine, The advantage of proton radiation therapy compared with photon
which, depending on the model, typically contains 192 or 201 fixed radiation therapy is the reduction of the total dose to the patient as
cobalt-60 sources that converge to a single point; (2) a conventional a result of the absence of an exit dose. This attribute results in a
linear accelerator (“linac”) outfitted with additional hardware so it decreased dose to the normal part of the brain, which may decrease
can deliver focused radiation, generally with three to five arcs; (3) acute and long-term toxicities, which is particularly important in
CyberKnife, which is a radiosurgery system composed of a linac pediatric patients.
mounted on a robot and directed under image guidance; and (4)
delivery of charged particles such as protons, which, because of their Adverse Effects After Irradiation of the Brain
physical properties, deposit energy in a narrower region than is possible or Spine
with x-rays.
Stereotactic radiation can be delivered in a single large fraction; Acute and Early Delayed Effects After Cranial Irradiation
this technique is termed stereotactic radiosurgery, although no surgery A variety of adverse effects may occur after irradiation of the CNS
is performed. A stereotactic head frame often is screwed to the skull. (reviewed by Schultheiss and associates143). These effects can be divided
The head frame permits three-dimensional localization and immobiliza- into acute, which occur during the treatment; delayed early, which
tion of the head during treatment, thereby allowing delivery of radiation occur within a few months of radiation; and late, which occur months
with precision to a very tightly defined volume defined with MRI to years later.
imaging. The usual dose of single-fraction radiation used in SRS Acute effects of cranial irradiation include fatigue, nausea, headaches,
ranges from 12 to 24 Gy and is based on the histologic tumor type anorexia, and alopecia. Patients may experience nausea within hours
and the volume of tissue irradiated. The doses used are based on the after the administration of the radiation and headaches during the
likelihood of tumor control and the risk of development of radiation course of treatment. Nausea and headaches are thought to be caused by
necrosis. radiation-induced edema and can be ameliorated with corticosteroids.
Stereotactic radiotherapy also can be delivered in a fractionated Nausea usually is well controlled with antiemetics such as ondansetron,
regimen over several days or weeks. CyberKnife does not require the granisetron, or prochlorperazine. In patients receiving craniospinal
use of a head frame but instead uses real-time image tracking while radiation, the nausea and anorexia may be compounded by radiation
patients are immobilized with a face mask. that the upper gastrointestinal tract receives as an exit dose from the
spinal field. Hair loss generally starts after the scalp has received 20
Intensity-Modulated Radiation Therapy to 30 Gy. It usually is not permanent, but regrowth of hair may
take months, and the new hair often is thinner than the original
Intensity-modulated radiation therapy uses multiple segmented fields hair or even of a different color. In areas that receive a high dose
with inverse treatment planning algorithms. In addition to defining of radiation, especially with tangentially directed fields, alopecia
the doses to be delivered to the GTV and CTV, dose constraints are may be permanent. Other acute adverse effects of cranial irradiation
placed on normal structures to limit their radiation exposure. Con- may include accumulation of cerumen in the ear canals and serous
straints include the maximum or mean dose to the entire structure, otitis media.
Cancer of the Central Nervous System • CHAPTER 63 921.e1
921.e1

100%

A B 30%

100%

C D 30%

100%

E F 30%

E-Figure 63.1 • Sample treatment plan for craniospinal irradiation showing dose distributions in sagittal and axial views for (A–B) three-dimensional
conformal radiotherapy, (C–D) tomotherapy, and (E–F) proton beam therapy. (From Yoon M, Shin DH, Kim J, et al. Craniospinal irradiation techniques:
a dosimetric comparison of proton beams with standard and advanced photon radiotherapy. Int J Radiat Oncol Biol Phys. 2011;81:3.)
922 Part III: Specific Malignancies

The most common delayed early effect from radiation is the Management of radiation necrosis may be either medical or surgical.
somnolence syndrome, which is characterized by excessive drowsiness, Corticosteroids may counteract the vascular endothelial damage and
nausea, and irritability.144 If this syndrome occurs, it generally does also decrease the inflammatory response.156 Other therapies, such as
so 1 to 3 months after radiation has been completed. This syndrome hyperbaric oxygen,157 anticoagulants,158 and vitamins159 have met with
is thought to be due to transient, diffuse demyelination. It usually is varying degrees of success, and no large trials have been conducted
seen after whole-brain irradiation but also can develop after limited-field to support their use.160 A promising therapy for brain necrosis is the
irradiation. The syndrome resolves spontaneously, but steroids can anti-VEGF agent bevacizumab, which may result in decreased vessel
shorten its duration. Delayed early effects occurring after cranial permeability. A double-blind placebo-controlled trial of 14 patients
irradiation can also take the form of focal neurologic signs due to with CNS radiation necrosis showed that patients receiving the antibody
intralesional reactions related to tumor response or perilesional reactions demonstrated a radiographic response, whereas those treated with
related to edema or demyelination. placebo did not demonstrate a response. Important to note, all patients
treated with bevacizumab showed clinical improvement.161 Surgical
Late Effects exploration often is necessary not only to establish a diagnosis but
The pathophysiology of late effects from CNS irradiation is poorly also as a therapeutic intervention to reduce mass effect and edema.
understood. Some of the effects may be caused by degenerative changes This results in clinical improvement in the majority of patients.147
in the supporting glial cells, whereas others may be caused by vascular
changes due to endothelial cell loss and capillary occlusion. The clinical Neurocognitive Deficits After Cranial Irradiation
and imaging manifestations of these changes may include cognitive Neurocognitive deficits often are observed after cranial irradiation,
deficits and neurobehavioral changes, pituitary-hypothalamic dysfunc- especially in young children. Many of the sequelae appear several
tion, and brain necrosis. years after treatment of children with brain tumors, which mandates
long-term follow-up. Sequential assessments of neurocognitive
Radiation Necrosis of the Brain function demonstrated progressive deterioration during 6 years after
One of the most serious late effects of cranial irradiation is brain radiotherapy in children with ALL who were treated with 18 Gy
necrosis, which may cause significant and persistent neurologic of whole-brain irradiation.162 Numerous studies of neurocognitive
injury.145,146 Histopathologic changes seen in radiation necrosis include function in children after whole-brain irradiation for ALL have been
coagulative and liquefactive necrosis, mostly in white matter, and performed.163–167 In summary, these findings show that whole-brain
associated endothelial thickening, vascular hyalinization, fibrinoid irradiation can lead to decline in neurocognitive function, an effect
deposition, and calcification.147 It may produce progressive cerebral that appears to be greater in younger children and with higher doses of
edema and mass effect requiring prolonged corticosteroid use or surgery. radiation (e.g., 24 Gy) but can be seen after treatment with 18 Gy.162
Radiation necrosis may be confused with tumor growth, resulting in It also is possible that an interaction between methotrexate (intrathecal
the inappropriate use of antitumor therapies. The onset of radiation or high-dose systemic) and whole-brain irradiation causes these late
necrosis includes behavioral changes, such as lethargy and dementia, effects.167 In one study, serial intelligence quotient (IQ) examinations
headache and papilledema, and seizure. Clinical signs and symptoms were used to follow 44 pediatric patients with medulloblastoma who
are usually identified 2 to 3 years after irradiation, although confirmed were treated with postoperative radiation therapy with or without
cases have been detected as early as 9 months and as late as 16 years chemotherapy. At a mean of 5.2 years from completion of radiotherapy,
after completion of radiation therapy. The signs and symptoms are the mean estimated full-scale IQ was greater than one standard
strongly related to the site of radiation necrosis; however, the most deviation below the expected population norm, and a mean loss of
common clinical signs are focal motor deficits. 2.55 estimated full-scale IQ points occurred per year. This study
Accurate data regarding the incidence of brain necrosis based suggests that the decline in IQ was not because of a loss of previously
on CT and MRI findings come from a randomized trial of irra- known skills and information, but rather an inability to acquire new
diation for low-grade gliomas.148 In this study, the 2-year actuarial skills and information at a rate comparable with healthy control
incidence of brain necrosis was 2.5% for patients receiving 50.4 Gy subjects.168
and 5% for those receiving 64.8 Gy. A retrospective review of 426 In a North Central Cancer Treatment Group (NCCTG) randomized
patients treated for glioma with radiation found that radionecrosis study of a dose of 64.8 versus 50.4 Gy for treatment of low-grade
developed in 4.9% of patients.149 The risk of necrosis increased gliomas,148 data regarding cognitive performance were collected
with longer survival and with increasing dose of radiation. The prospectively. With a median follow-up time of 7.4 years in survivors,
use of adjuvant chemotherapy was associated with increased risk of the vast majority of patients with normal baseline findings on the
radiation necrosis. Mini Mental State Examination (MMSE) maintained normal findings
Often radiation necrosis is difficult to distinguish from recurrent after undergoing radiotherapy.169 Patients with abnormal MMSE results
tumor with CT or MRI, which may show increased signal intensity before radiotherapy were more likely to have an improvement in
on T2-weighted images and contrast enhancement on T1-weighted cognitive abilities than deterioration after being treated with radio-
images.150 As discussed earlier in the section on diagnostic imaging, PET therapy. Armstrong and coworkers170 conducted prospective, compre-
scanning with FDG may help distinguish viable tumor from necrotic hensive, longitudinal neuropsychologic testing on 26 adult patients
tissue.151–153 Advanced MRI techniques may also help differentiate tumor with low-grade supratentorial brain tumors, mostly gliomas, who had
from necrosis. On MRS, NAA is significantly reduced in patients with been treated with radiotherapy. Nine patients underwent testing 6
radiation necrosis, whereas changes in choline and creatine are more years after being treated with radiotherapy. No declines were noted
variable. A high choline level is more consistent with disease progression, on most neurocognitive tests. Results in 7 of the 37 neuropsychologic
whereas a decreased creatine level is generally associated with radiation tests showed improvement over 6 years. However, declines emerged
injury.147 Ratios of metabolites are considered, and pooling ratios of only after 5 years on selected tests of cognitive function such as visual
different metabolites may be helpful in differentiating tumor from memory.
necrosis.155 Perfusion imaging techniques that measure rCBV may also Some evidence indicates that adults treated with brain radiotherapy
help differentiate recurrence from necrosis by allowing for indirect may experience long-term cognitive sequelae. One study aimed to
assessment of tumor angiogenesis and vascularity. Hyperperfusion is seen differentiate causes of cognitive disability—either the tumor itself or
with tumor recurrence, and necrosis is associated with ischemia-related various treatments, including radiotherapy, surgery, or medication—in
change.147 Still, rCBV may be increased in areas of radiation-induced a population of patients with a low-grade glioma at a mean of 6 years
inflammation. Advanced MRI techniques of late radiation-induced from diagnosis. Their findings suggest that the tumor had the greatest
injury are reviewed by Pružincová and colleagues.155 deleterious effect on cognitive functioning and that radiotherapy
Cancer of the Central Nervous System • CHAPTER 63 923

resulted in long-term cognitive disability mostly when high doses per impaired by spinal irradiation, which directly affects the vertebral body
fraction (>2 Gy) were used.171 An update of this study, which assessed growth center.
cognitive abnormalities at a mean of 12 years after diagnosis, showed Precocious puberty may occur after relatively low doses, on the
that patients who were treated with radiation therapy had greater order of 18 Gy.185 Deficiencies in other hormones, such as gonado-
deficits than did control subjects who did not undergo radiotherapy. tropins, thyroid-stimulating hormone (TSH), and adrenocorticotropic
The patients who underwent radiation therapy had more deficits in hormone (ACTH), are rare after doses lower than 40 Gy.181 Thyroid
attention functioning at follow-up, regardless of fraction dose, and dysfunction is common in patients with brain tumors who are treated
they also performed worse in tests of executive functioning and with high-dose radiotherapy.186 Constine and associates studied
information processing speed.172 endocrine function in 32 patients with brain tumors not involving
Data regarding treatment of these cognitive deficits in brain tumor the hypothalamic-pituitary region who received 39.6 to 70.2 Gy to
survivors are limited and conflicting. In a randomized trial of 83 this region.187 Hypothalamic or pituitary hypothyroidism developed
childhood survivors of ALL or malignant brain tumors who had in 65% of the patients. Fourteen of 23 postpubertal patients (61%)
problems with academic achievement, the use of methylphenidate had evidence of hypogonadism manifesting with oligomenorrhea, low
was investigated, and it was revealed that the drug at least temporarily estradiol levels, or low testosterone levels. Half of the patients had
reduced some attentional and social deficits.173 However, a phase III mild hyperprolactinemia. Subtle abnormalities in adrenal function
placebo-controlled trial in patients with a brain tumor failed to show were seen in 35% of patients.
that prophylactic use of methylphenidate improved quality of life or In patients receiving craniospinal radiation, hypothyroidism also
cognitive function.174 A phase II trial of the acetylcholinesterase inhibitor may occur as a result of the exit dose to the thyroid gland. In one
donepezil in patients who received radiation for brain tumors showed study, the incidence of hypothyroidism was 15% or 33% (P = .013),
improved cognitive functioning, mood, and health-related quality of respectively, for patients receiving cranial or craniospinal irradiation.188
life after a 24-week course of the drug.175 Cranial transplantation of The mean spinal dose was 29 Gy, and the exit dose to the thyroid
human stem cells is a treatment currently under investigation in the gland ranged from 10 to 15 Gy.
laboratory. Radiation-induced depletion of stem cells in the brain,
especially in the area of the hippocampus, may be partially responsible Optic Neuropathy After Cranial Irradiation
for radiation-induced cognitive deficits. Using rats subjected to cranial Irradiation of tumors that are close to the optic nerves or chiasm may
irradiation, researchers have transplanted human embryonic stem result in a sufficient dose to these structures to cause concern about
cells176 or human neural stem cells177 into the hippocampal formations. the development of optic neuropathy. Two major classes of optic
Animals receiving stem cells showed superior performance on neuropathy are recognized: anterior optic neuropathy and retrobulbar
hippocampal-dependent cognitive tasks. optic neuropathy.189 The former is thought to be due to vascular injury
Young age also is an important risk factor for leukoencephalopathy, affecting the nerve head inside the globe anterior or adjacent to the
which can affect all age groups. Histologically, multifocal white matter lamina cribrosa. This vascular injury is associated with swelling of the
destruction with loss of myelin can be seen, especially in the periven- optic head, in contrast with retrobulbar optic neuropathy, which is
tricular regions; MRI scans show these periventricular abnormalities. due to more proximal injury to the optic nerve. Diagnostic criteria
CT scans also may reveal the presence of intracerebral microcalcifications for retrobulbar optic neuropathy include (1) visual loss (monocular
due to mineralizing microangiopathy. The clinical expression of or binocular) accompanied by corresponding visual field defects, (2)
leukoencephalopathy ranges from mild evidence of white matter injury funduscopic examination, which often shows a pale optic disc but
on neuroimaging studies to severe necrotizing leukoencephalopathy without edema, (3) onset 6 months to several years after radiation
with profound neurologic impairment and, in some cases, death. Mild therapy that delivered a significant dose to the optic nerve and chiasm,
or subclinical cases are more common than severe necrotizing leuko- and (4) no radiologic evidence of visual pathway compression.190 MRI
encephalopathy. The bulk of the experience comes from children who scans may show pathologic contrast enhancement of the region of
received 24 Gy of whole-brain radiation along with high doses of the optic nerve and chiasm that received a high dose of radiation.191
intravenous and intrathecal methotrexate. The frequency of leukoen- Parsons and colleagues189 examined radiation-induced optic neu-
cephalopathy is low in patients who receive cranial radiotherapy and ropathy in patients receiving radiation treatment for primary extracranial
intrathecal methotrexate or cranial irradiation and intravenous head and neck tumors. Of 215 optic nerves at risk, these investigators
methotrexate but may be as high as 45% in patients who receive all found anterior optic neuropathy in 5 nerves and retrobulbar optic
three treatments.178 In general, methotrexate is most toxic when given neuropathy in 12 nerves. No injuries were observed in 106 optic
during or after radiation. nerves that underwent irradiation with less than 59 Gy. The 15-year
Cranial irradiation can result in arterial vascular problems such as actuarial risk of optic nerve neuropathy after administration of 60 Gy
vessel obliteration or narrowing, resulting in a strokelike syndrome.179 or more was 11% when daily fractions less than 1.9 Gy were used,
These complications are rare, but when they occur they are more versus 47% when 1.9 Gy or more was used.
likely to happen after irradiation of the parasellar region. Although these data suggest that the optic nerve–chiasm tolerance
is at least 59 Gy, their population did not include patients with
Endocrine Deficits After Cranial or Spinal Irradiation intracranial tumors compressing the optic nerve and chiasm. It is
Endocrine problems are common after cranial irradiation, particularly possible in the latter situation that the tolerance of the optic nerve
in children.180–182 Such problems include growth hormone (GH) and chiasm is lower as a result of ischemic injury. In some patients
deficiency and thyroid and gonadal dysfunction. with pituitary adenomas or craniopharyngiomas treated with radiation,
The hypothalamus is more radiosensitive than the pituitary optic neuropathy developed after doses as low as 45 to 50 Gy, although
gland and is responsible for endocrine dysfunction at lower doses. in many of these patients the daily fraction size was greater than
At higher doses (>40 Gy), however, both the anterior pituitary gland 2 Gy.190,192,193 On the basis of these results, most investigators currently
and the hypothalamus contribute to endocrine dysfunction. Of all the recommend limiting the dose to the optic chiasm and nerve to 50 Gy
hormones, GH is the most likely to show deficiency after irradiation in 1.8- to 2-Gy fractions in the treatment of pituitary adenomas or
and is seen in a majority of children who have received whole-brain craniopharyngiomas. For other brain tumors requiring higher doses,
irradiation. One study found that in children with ALL who received most radiation oncologists would restrict the dose to the optic nerve
a dose of 24 Gy to the whole brain, 56% experienced GH deficiency, and chiasm to 54 Gy or lower. In one series in which the risk of optic
whereas no such problems were seen in children who received a neuropathy after SRS was examined, a risk of 1.1% for patients receiving
dose of 18 Gy, at least 4 years later.183 The latency of onset is dose 12 Gy or less was found, and the risk was increased in patients who
dependent and is shorter with higher doses.184 Growth may be further received previous or concurrent external beam radiation therapy.194 A
924 Part III: Specific Malignancies

conservative estimate for single-dose tolerance of the optic nerve and of chronic myelopathy was extremely low. Even if the spinal cord dose
chiasm in SRS is 8 Gy.195 Adherence to these guidelines should keep is limited to 45 Gy, myelitis may still develop. This report cited
the risk of radiation-induced optic neuropathy extremely low (1% or published cases in which myelitis developed with doses less than 45 Gy
less) but unfortunately will not completely eliminate it. Possible or even less than 40 Gy, given in 1.8- to 2-Gy daily fractions. Such
treatments include use of hyperbaric oxygen, corticosteroids, or cases, however, are extremely rare, with an estimated risk of 0.2% or
anticoagulation, but none is particularly effective.196 less for the development of chronic myelopathy at this dose.143 No
effective treatment exists for this condition, and only case reports are
Second Malignant Neoplasms Developing After found in the literature suggesting possible instances of benefit with
growth factors, anticoagulation, and bevacizumab.
Cranial Irradiation
Second malignant neoplasms including malignant gliomas and GENERAL PRINCIPLES OF CHEMOTHERAPY
meningiomas remain relatively uncommon consequences of radiation
therapy for brain tumors.197 Concern exists that the addition of adjuvant Chemotherapy for brain tumors involves many of the same problems
chemotherapy may increase the risk of second malignancies in long-term as chemotherapy for systemic cancer, including lack of specificity,
survivors of childhood brain tumors.16,17 This outcome may be especially intrinsic or developing cellular resistance, intolerance of normal tissue
true after prolonged use of alkylating agents and etoposide with or to drug toxicity, synergistic toxicity between chemotherapy and radiation
without irradiation. therapy, and systemic toxicity. Brain tumor therapy also is associated
with specific problems of drug delivery across the blood-brain or
Myelopathy After Spinal Irradiation blood-tumor barrier. Cerebral edema may impede drug delivery, and
A delayed early effect that can be seen after irradiation of the cervical corticosteroids that effectively treat the edema may “repair” tumor
spine is Lhermitte syndrome, which is characterized by an electric vessels and impede the delivery of chemotherapy to the tumor. Radiation
shock–like sensation precipitated by forward neck flexion.198 The therapy also may result in endovascular changes, and the sequencing
symptoms typically start weeks to a few months after completion of of chemotherapy administration with irradiation remains an area of
radiation therapy. They are maximal at first but abate with time, active investigation. Thus a recurring disappointment in clinical
without the development of any objective signs. The paresthesias most chemotherapy trials has been the failure to translate promising preclini-
commonly occur in the lumbosacral region but also can involve the cal chemotherapy findings into meaningful improvement in patient
upper and lower extremities and the upper back. This transient form survival.
of Lhermitte syndrome can occur at radiation doses within accepted Clinical criteria for chemotherapeutic success can be confusing.
spinal cord tolerance and is not associated with any permanent late Performance status, measured in many clinical trials with the Karnofsky
sequelae. The pathogenesis is thought to involve an inhibitory effect Performance Scale (KPS), is affected by tumor site, presence of seizures,
on oligodendrocytes that results in transient reversible demyelination. and adverse effects of AEDs and steroids. Therefore an appropriate
A more ominous form of Lhermitte syndrome can manifest after a measure of chemotherapeutic efficacy should not be median survival
longer latency period after completion of radiation therapy (at least alone. Median progression-free survival (PFS) or 6-month PFS and
a year), which then progresses to chronic radiation myelopathy. reduction in seizure frequency, corticosteroid requirement, and focal
Chronic myelopathy is the most catastrophic late effect that can deficits all are parameters that must be measured. For long-term
occur after spinal cord irradiation. Nearly half of the affected patients survivors, cognitive impairment and other serious acute and chronic
will die from complications.199 A biphasic distribution in the latency neurologic toxicities of cytotoxic chemotherapy and newer molecular
period from irradiation to the onset of myelopathy has been observed. strategies factor into the assessment of chemotherapeutic program
The early peak is from 12 to 14 months, and the second peak is from efficacy.202
24 to 28 months. The pathologic insults that lead to myelopathy Finally, chemotherapy of primary brain tumors for approximately
include damage to oligodendroglial cells, causing demyelination and the past 20 years has been designed on the assumption that the problem
white matter necrosis, and death of endothelial cells, resulting in is one of local control, with few recurrences outside the original tumor
vascular injury. In some patients, partial neurologic dysfunction site.203 With longer survival periods and better local tumor control,
develops; others progress to complete paraplegia or quadriplegia. No however, a higher rate of multicentric cerebral and even leptomeningeal
clinical or radiologic findings are pathognomonic for radiation-induced disseminated disease has been noted among patients with high-grade
myelopathy. Therefore the diagnosis usually is made by a combination glial tumors, and oncologists may need to design future systemic
of (1) neurologic abnormalities corresponding to a level just below regimens with these considerations in mind. Recent studies with an
the irradiated region, (2) a history of spinal cord irradiation with a antibody specific to a mutation of the isocitrate dehydrogenase (IDH1)
high total dose (greater than 45 Gy) at least 6 months before the biomarker identified single tumor cells remote from the presenting
onset of signs and symptoms, (3) MRI findings of increased signal mass and lend credence to the notion that what may appear to be
intensity on T2-weighted images in the irradiated region,200 and (4) focal glioma may represent systemic brain disease.204,205
exclusion of other etiologic factors. The MRI findings in spinal cord An important consideration in the chemotherapy of brain tumors
myelopathy can mimic tumor recurrence with gadolinium enhancement is the ability of the drugs to cross the blood-brain barrier. This physi-
on MRI. ologic barrier defines the restricted transport between blood and the
The probability of development of chronic radiation myelopathy CNS of water-soluble, ionized molecules larger than about 200 daltons.
is dependent not only on the total dose delivered to the spinal cord The blood-brain barrier is formed by the endothelial cells of brain
but also on the fraction size. Larger fraction sizes are associated with capillaries, with some contribution from astrocytes. Brain capillaries
more severe later effects (Schultheiss and associates143). Using standard differ from capillaries elsewhere in the body by the presence of tight
fraction sizes (1.8–2 Gy per day), a commonly observed limit for dose intercellular junctions. Tight junctions consist of a protein complex
to the spinal cord is 45 Gy. In a study of the incidence of myelitis of two groups, the transmembrane proteins occludin and claudin,
after irradiation of the cervical cord, Marcus and Million201 found and junction-associated molecules and cytoplasmic scaffolding and
that of 1112 patients, chronic radiation myelopathy developed in regulatory proteins. The brain’s extracellular or interstitial fluid is an
only 2 (0.18%). This complication developed in 2 out of 471 patients ultrafiltrate essentially identical to CSF. Capillaries of the choroid
(0.43%) receiving between 45 and 50 Gy. No patients who received plexuses are fenestrated, allowing access of large protein molecules
between 40 and 45 Gy or who received more than 50 Gy developed into the plexus stroma. The epithelial cells separating stroma from
myelopathy. The investigators’ conclusion was that even with doses CSF, however, have apical tight junctions. The brain lacks a lymphatic
of up to 50 to 55 Gy to the cervical cord, the likelihood of development system. Brain tumors and their associated blood vessels have altered
Cancer of the Central Nervous System • CHAPTER 63 925

expression and function of membrane transporters and enzymes leading that extend from the astrocytes, and thus antibodies against this protein
to vasogenic edema and inflammation.206,207 can be used in immunohistochemical studies.
These features of the blood-brain barrier and the blood-CSF barrier Over the years, many different histopathologic staging systems
exclude entry of large molecules such as proteins and limit entry of have been used.213–215 Historically systems classify tumors in up to
smaller molecules to their ability to cross the lipid bilayer of cells. four categories with increasing malignancy. The most widely used
Lipid-soluble molecules such as nicotine, ethanol, heroin, and the system today for classification of astrocytomas is part of the WHO
alkylating agents bis-chloroethylnitrosourea (BCNU; carmustine) and classification of tumors of the CNS, which was last updated in 2016.216
temozolomide (Temodar) pass readily across the blood-brain barrier. Compared with the 2007 classification, which relied solely on histologic
Another function of the blood-brain barrier may be that of pumping characteristics, the 2016 WHO classification of tumors of the CNS
out chemicals potentially harmful to the brain. P-glycoprotein, a now also uses molecular data to define CNS tumors. Notable changes
blood-brain barrier protein, is present in the membrane of some brain include the removal of oligoastrocytoma as a distinct entity and the
tumor cells and serves to reduce the intracellular concentration of subclassification of tumors based on IDH1 mutation status, which
several chemotherapeutic agents. confers a better prognosis in both high-grade and low-grade astrocy-
Quantitative examination of the blood-brain barrier and the brain- tomas. The 2016 CNS WHO classification uses the classical histologic
tumor barrier has resulted in new avenues of investigation for brain features to make an initial diagnosis, which is then refined on the
tumor chemotherapy. Methotrexate entry into brain tumors can be basis of molecular testing (Fig. 63.9).
enhanced by intraarterial delivery, but the use of hyperosmolar mannitol
for this purpose results in a far greater increase of drug entry into Histologic Classification of Supratentorial Gliomas
normal brain tissue than into tumor. Human and rat studies of BCNU,
cisplatin, and other agents after blood-brain barrier disruption in both The category of grade I astrocytic tumors is reserved for subependymal
primary glial tumors and PCNSL have demonstrated significant giant cell astrocytoma and pilocytic astrocytoma, which are the most
toxicity.208,209 Intraarterial delivery of such drugs has been largely benign in terms of histologic characteristics; they are generally curable
abandoned. with surgery alone. Because grade I astrocytomas are more common
In brain tumors, the degree to which the tight endothelial cell in children than in adults, they are discussed in greater detail later in
junctions of the blood-brain barrier are disrupted and vascular perme- the section on childhood brain tumors. The remaining categories are
ability is thereby increased varies even within individual tumors. The grade II (diffuse astrocytomas), grade III (anaplastic astrocytomas),
persistence of a relatively intact barrier impedes entry of some water- and grade IV (glioblastomas).
soluble chemotherapeutic agents into areas of tumor, leading investiga- Diffuse astrocytomas (grade II) are poorly defined gray tumors
tors to look for methods of opening the blood-brain barrier that are that expand the parenchyma and obliterate normal gray matter–white
less toxic than intraarterial mannitol, but efforts have not been suc- matter boundaries217 (Fig. 63.10). Microscopically, increased numbers
cessful.210 Radiation-induced blood-brain barrier disruption may offer of irregularly distributed astrocytes with mildly atypical nuclei are
another avenue to deliver chemotherapy to brain tumors. For example, noted (Fig. 63.11A). Tumor cells can be seen infiltrating into normal
evidence suggests that radiation therapy may offer a unique window brain tissue. Diffuse astrocytomas often are classified into one of three
extending from 1 week after the initiation of radiotherapy to 1 month subtypes: fibrillary (the most common subtype), protoplasmic, or
after the completion of treatment during which a pharmaceutical gemistocytic.218 Fibrillary astrocytomas are composed of tightly
agent has maximum access to high-grade gliomas.211 interlacing bundles of small, spindle-shaped cells amid a predominantly
fibrillar matrix. Gemistocytic astrocytomas contain plump cells with
distinct, round pink cytoplasm arranged on a more delicately interlacing
SUPRATENTORIAL GLIOMAS fibrillar matrix. Protoplasmic astrocytomas are composed of small,
Clinical Considerations round, regular cells with indistinct cytoplasmic boundaries arranged
on a loosely fibrillar stroma.
Patients with supratentorial gliomas may have general signs and Microscopically, anaplastic astrocytomas (WHO grade III)
symptoms such as changes in mental status, seizures, headaches, are distinguished from grade II tumors by their greater cellular-
papilledema, and nausea and vomiting, as discussed earlier. They also ity, increased nuclear pleomorphism, and, of greatest importance,
may have focal signs and symptoms, dependent on tumor location, mitotic activity (see Fig. 63.11B). In both these and grade IV
as discussed previously. glioblastomas, tumor cells can be seen infiltrating into surrounding
The incidence of low-grade astrocytomas is highest between the normal brain tissue, often at a great distance from the primary tumor
ages of 20 and 40 years and decreases in patients older than 50 years. mass. Histologically, glioblastomas are distinguished from anaplastic
Oligodendrogliomas display a similar age-related frequency. Conversely, astrocytomas by the presence of endothelial proliferation or necrosis
high-grade astrocytomas, including glioblastomas, increase in frequency (Fig. 63.12). The necrosis can form a serpentine pattern referred to
with increasing age (see Fig. 63.1). as pseudopalisading, in which tumor cells crowd around the edges
A history of seizures also is extremely common in patients with of the necrotic region. The presence of necrosis is of particular
oligodendrogliomas, occurring in 70% to 90%. The duration of importance in grading gliomas and has been associated with shorter
symptoms can be prolonged. In a study by Ludwig and colleagues,212 survival.219
55% of patients had symptoms for longer than 1 year before diagnosis, Grade IV astrocytomas (glioblastomas) show areas of hemorrhage,
and 37% had symptoms for longer than 3 years. A few patients had necrosis, and cystic change on gross examination (Fig. 63.13). Glio-
symptoms for 10 to 15 years before diagnosis. blastoma can manifest with multicentric disease, but this occurs in
Most glioblastomas have a very short symptomatic history at less than 5% of cases as determined at the autopsy of patients with
presentation, on the order of days or weeks. Tumors that seem to untreated tumors.203
progress more slowly with a longer antecedent history of seizures or In addition to pathologic grading by morphology, immunohisto-
cognitive decline may have progressed from a lower-grade glial neoplasm chemical techniques have been developed to measure cell kinetics
and thus are considered secondary glioblastomas. based on the idea that faster-growing tumors are more malignant.
Ki-67 (MIB-1) or proliferating cell nuclear antigen is a measure of
Pathologic Classification of Supratentorial Gliomas cellular proliferation. Bromodeoxyuridine (BUdR) or iododeoxyuridine
incorporation into tumor cells can help measure the proportion of
Astrocytomas arise from astrocytes, the supporting cells of the CNS. cells in the S phase of mitosis. The predictive power of all three of
Glial fibrillary acidic protein is expressed in the cytoplasmic processes these techniques has been compared with that for clinical parameters.220
926 Part III: Specific Malignancies

Histology Astrocytoma Oligoastrocytoma Oligodendroglioma Glioblastoma

IDH status
IDH mutant IDH wild-type IDH mutant IDH wild-type

1p/19q and Glioblastoma, IDH mutant


other genetic ATRX loss*
1p/19q codeletion
parameters TP53 mutation*
Glioblastoma, IDH wild-type

Diffuse astrocytoma, IDH mutant

Oligodendroglioma, IDH mutant and 1p/19q codeleted Genetic testing not done
or inconclusive

After exclusion of other entities:


Diffuse astrocytoma, IDH wild-type
Oligodendroglioma, NOS

Diffuse astrocytoma, NOS


Oligodendroglioma, NOS
Oligoastrocytoma, NOS
Glioblastoma, NOS

Figure 63.9 • Molecular classification based on 2016 WHO classification. Both IDH1 mutation status and 1p19q status have become defining feature
in the molecular WHO classification of gliomas. *, Characteristic but not required for diagnosis; IDH, isocitrate dehydrogenase; NOS, not otherwise specified.
(From Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary.
Acta Neuropathol. 2016;131[6]:803–820.)

Immunohistochemical stains for genetic changes found in glial tumors


are increasingly used to assess prognosis.
The different grades of astrocytoma carry very different prognoses.
With WHO grade I (pilocytic) astrocytomas, a cure rate greater than
90% after surgical resection alone can be expected. By contrast, with
WHO grade IV tumors, the median survival period is 1 to 2 years,
even after aggressive combined-modality therapy. With WHO grade
III tumors, long-term survival rates on the order of 20% are usual,
but the median survival period is approximately 2 years. WHO grade
II tumors carry a better prognosis than grade III tumors; nevertheless,
recurrence with a higher-grade tumor is common with grade II tumors,
although recurrence may take 6 to 8 years to occur.
Most oligodendrogliomas occur in the cerebral hemispheres (80%),
but they can rarely occur in the lateral and third ventricles. Oligo-
dendrogliomas arise from oligodendrocytes, which produce myelin.
They often involve the subcortical white matter, with extension into
the cerebral cortex. On gross examination, oligodendrogliomas are
Figure 63.10 • Diffuse low-grade astrocytoma, gross specimen. A World often soft and gelatinous and better circumscribed than astrocytomas.217
Health Organization grade II astrocytoma arises diffusely and infiltrates the They frequently contain calcifications. Despite their gross appearance
right frontal lobe. Gross determination of the tumor’s boundaries is almost suggesting that they are contained, they can infiltrate surrounding
impossible, but the tumor is evident as an ill-defined area of enlargement, tissues, including the subarachnoid space and leptomeninges.
with loss of distinction between the gray and white matter. (From Maher EA, Because of fixation artifact, oligodendrogliomas appear micro-
McKee AC. Neoplasms of the central nervous system. In: Skarin AT, ed. scopically as sheets of cells with nuclei surrounded by a clear halo of
Dana-Farber Cancer Institute Atlas of Diagnostic Oncology. 3rd ed. St Louis: cytoplasm, giving the cells the appearance of a “fried egg” (Fig. 63.14A).
Mosby; 2003.)
Unlike astrocytomas, oligodendrogliomas lack fibrillary cytoplasmic
processes. Calcifications are present in 90% of these tumors. A common
finding is a network of capillaries, lending a “chicken wire” pattern.
The WHO classification for oligodendrogliomas is a two-tiered
system of grade II (oligodendroglioma) and grade III (anaplastic
Cancer of the Central Nervous System • CHAPTER 63 927

A B
Figure 63.11 • Histologic appearance of a low-grade astrocytoma versus an anaplastic astrocytoma. (A) A low-grade astrocytoma (World Health Organization
[WHO] grade II) shows low cellularity, slight nuclear pleomorphism, no endothelial proliferation, and no necrosis. (B) An anaplastic astrocytoma (WHO grade
III) shows increased cellularity, pleomorphism, and mitotic activity compared with the grade II tumor in (A). The anaplastic astrocytoma also lacks endothelial
proliferation and necrosis. (Courtesy Dr. Daniel Skovronsky, Department of Pathology, University of Pennsylvania School of Medicine, Philadelphia, PA.)

A B
Figure 63.12 • Glioblastoma (grade IV astrocytoma): histologic appearance. (A) Pseudopalisading cells surrounding regions of necrosis. (B) Endothelial
proliferation, which in this case has reached dramatic proportions, with the formation of tangled clusters of neovascular channels, often referred to as glomeruloid
blood vessels because of their resemblance to renal glomeruli. (From Maher EA, McKee AC. Neoplasms of the central nervous system. In: Skarin AT, ed.
Dana-Farber Cancer Institute Atlas of Diagnostic Oncology. 3rd ed. St Louis: Mosby; 2003.)

oligodendroglioma).216 Features suggestive of anaplasia include either IDH-mutant or IDH-wildtype. For instance, diffuse astrocytoma
cytologic atypia and increased mitotic activity (see Fig. 63.14B). In can be IDH-mutant or IDH-wildtype; similarly, glioblastoma can
the Mayo Clinic series, the 5- and 10-year survival rates and median also be considered IDH-mutant or IDH-wildtype. IDH-mutant status
survival period for low-grade oligodendrogliomas were 75%, 46%, confers a survival advantage when compared with IDH wild-type
and 9.8 years, respectively, versus 41%, 20%, and 3.9 years for high- tumors of the same grade (Fig. 63.15).
grade tumors.221 Approximately 80% of all glioblastomas develop without any prior
Overall, patients with oligodendrogliomas have a better prognosis history of a glioma and are termed primary glioblastomas. In 20%
than patients with astrocytomas.148 For example, in the Mayo Clinic of the cases, the patient has an antecedent history of a lower-grade
experience, the 5- and 10-year survival rates and the median survival glioma (grade II or III), generally 5 to 10 years previously. These
period were 46%, 17%, and 4.7 years, respectively, for low-grade secondary glioblastomas tend to arise in younger patients, with a
diffuse astrocytomas, compared with 73%, 49%, and 9.8 years for median age of 40 years. Furthermore, IDH mutation is seen more
low-grade oligodendrogliomas.222 commonly in secondary glioblastoma (73%) compared with primary
glioblastoma (4%).223
Molecular Classification of Supratentorial Gliomas A genome-wide mutational analysis of glioblastomas identified
somatic mutations in the isocitrate dehydrogenase 1 gene (IDH1) in
Isocitrate Dehydrogenase Mutations 12% of these tumors. These mutations occurred in a high percentage
The 2016 WHO classification of CNS tumors incorporates IDH of young patients and were seen in most secondary glioblastomas.224
mutation status.216 Each grade of astrocytoma can be classified as In this analysis, IDH1 mutations were associated with longer
928 Part III: Specific Malignancies

overall survival (OS) with a median survival of 3.8 years in patients had both IDH1 mutation and 1p/19q loss. IDH1 mutations occurred
with the mutated gene versus 1.1 years for patients with the wild- in 10% of pilocytic astrocytomas, 5% of primary glioblastomas, and
type gene. none of the ependymomas examined. These findings are important
Both IDH1 and IDH2 mutation status were explored in a variety because they provide evidence that different histologic types of low-grade
of CNS and non-CNS tumors.225 These investigators identified muta- gliomas share a frequent genetic alteration.
tions in amino acid 132 of IDH1 in more than 70% of WHO grade IDH1 and IDH2 are nicotinamide adenine dinucleotide phosphate
II and III astrocytomas and oligodendrogliomas, and in glioblastomas (NADP+)–dependent enzymes that catalyze the oxidative decarbox-
that developed from these lower-grade gliomas. Often tumors without ylation of isocitrate into α-ketoglutarate226 (see Fig. 63.15). The mutated
mutated IDH1 had mutations in the analogous amino acid of the forms of IDH1 and IDH2 have been shown to result in the induction
IDH2 gene. Other investigators have found that IDH1 mutations of hypoxia-inducible factor-1α (HIF-1α), whereas levels of
occurred in 88% of low-grade diffuse astrocytomas and in 82% of α-ketoglutarate decreased (loss of function) and the conversion of
secondary glioblastomas. In addition, no cases of IDH1 mutation α-ketoglutarate to 2-hydroxyglutarate occurred (gain of function).227
occurred after either acquisition of TP53 mutations or 1p/19q loss, The decreased enzymatic activity suggests that IDH1 and IDH2 are
suggesting that IDH1 mutation is a very early event in gliomagenesis. tumor suppressor genes.205 However, the altered substrate specificity
The majority of grade II astrocytomas (63%) contained both IDH1 seen in mutant IDH1 is more compatible with oncogenic function.205,228
and TP53 mutations, whereas 64% of grade II oligodendrogliomas Mutation in either a tumor suppressor or an oncogene would be
expected to be associated with poorer prognosis, but the opposite is
in fact seen in patients with this mutant gene.227
The reason that IDH mutations result in a better prognosis is
unknown. Some investigators have hypothesized that mutated IDH
decreases the reduced form of nicotinamide adenine dinucleotide
phosphate (NADPH) and α-ketoglutarate. Several pathways are affected
by changes in NADPH and α-ketoglutarate concentrations. A decrease
in these molecules, which normally rescue cells from oxidative stress,
results in increased radiosensitivity.229 IDH mutations might also be
important for cellular migration and the cellular hypoxic response.
Increased lactate production results from inhibition of cytosolic IDH1,
which results in increased glioma cell migration.230 In vitro and in
vivo studies have demonstrated that the IDH1 mutation reduces the
aggressiveness of an established glioma cell line.231
The discovery of the role of IDH1/IDH2 in gliomas has important
implications. First, this enzyme previously had no known role in
tumor formation, and it has prompted a resurgence of interest in the
field of altered metabolism in tumorigenesis. Second, it is now used
to more accurately classify patients because it is associated with
prolonged survival. IDH1 mutation is also associated with differentia-
tion between primary and secondary glioblastoma, with a sensitivity
Figure 63.13 • Glioblastoma: gross specimen. The tumor appears as a of 70% to 73% and specificity of 96%.232 In addition, this mutation
necrotic, hemorrhagic infiltrating mass. (From De Girolami U, Anthony DC, can be used to differentiate between diffuse and pilocytic astrocytomas
Frosch MP. The central nervous system. In: Cotran RS, Kumar V, Collins T, with 73% to 83% sensitivity and 100% specificity. Finally, the pathways
eds. Robbins’ Pathologic Basis of Disease. 6th ed. Philadelphia: WB Saunders; affected as a consequence of this mutation may represent novel thera-
2003.) peutic targets (see Fig. 63.15).

A B
Figure 63.14 • Histologic appearance of oligodendroglioma versus anaplastic oligodendroglioma. (A) In World Health Organization (WHO) grade II
oligodendroglioma, oligodendrocytes are uniform cells with small, round nuclei and a characteristic perinuclear halo (“fried egg” cells). (B) A grade III oligo-
dendroglioma shows cytologic atypia and increased mitotic activity compared with the tumor in (A). (Courtesy Dr. Daniel Skovronsky, Department of
Pathology, University of Pennsylvania School of Medicine, Philadelphia, PA.)
Cancer of the Central Nervous System • CHAPTER 63 929

Glucose

Acetyl CoA

OAA Cit Cit

Mal Isocitrate Isocitrate


NAD+ NADP– NADP–
IDH3 IDH2 IDH1
Fum
CO2 + NADPH NADPH + CO2 NADPH + CO2
αKG aKG
Suc

Glutamine

Glutamine
A

Glucose

Acetyl CoA

OAA Cit Cit

Mal Isocitrate Isocitrate


NAD+ NADP+ NADP–
IDH3 IDH2 IDH1
Fum CO2 + NADH NADPH + CO2 NADPH + CO2
αKG αKG
Suc NADPH αKG-dependent enzymes
NADPH and processes
IDH2*
D2

Glutamine IDH1*
HG

NADP+
NADP+
DH

2HG
Glutamine
Transcriptional regulation
2HG Proliferation
Angiogenesis
Apoptosis resistance
Migration and invasion

• Mitochondrial activity
• Ca2+ homeostasis
• 5-ALA and HO-Lys metabolism
B
Figure 63.15 • Metabolic reactions catalyzed by the isocitrate dehydrogenase (IDH) family. (A) IDH1 catalyzes the NADP+-dependent decarboxylation
of isocitrate to α-ketoglutarate (αKG) in the peroxisome and cytoplasm, whereas IDH2 mediates the same reaction in the mitochondria. Also, within the
mitochondrial compartment, heterotetrameric IDH3 catalyzes the conversion of isocitrate to αKG in an NADP+-dependent manner. (B) Mutations in IDH1
and IDH2 impart to these enzymes the ability to catalyze the reduction of αKG to 2-hydroxyglutarate (2HG). On the basis of the chemical similarity between
these two metabolites, it is possible that the activity of αKG-dependent enzymes is affected by the elevated level of 2HG in tumors harboring IDH1 and
IDH2 mutations. In this model, these events may trigger alterations in angiogenesis, epigenetic state, and extracellular matrix dynamics, which in turn may
affect proliferation, survival, and invasive properties of these tumors. Cit, Citrate; Fum, fumarate; Mal, malate; NAD, nicotinamide adenine dinucleotide;
NADPH, reduced form of nicotinamide adenine dinucleotide phosphate; OAA, oxaloacetate; Suc, succinate. (Modified from Yen KE, Bittinger MA, Su SM,
et al. Cancer-associated IDH mutations: biomarker and therapeutic opportunities. Oncogene. 2010;29:6409–6417.)

LOH of 1p and 19q is common in both grade II and grade III


1p19q Codeletion in Oligodendroglioma oligodendrogliomas, and thus these changes probably occur early during
Two chromosomal abnormalities have now become a defining tumorigenesis.234,235 The 2007 WHO CNS classification included
feature of oligodendrogliomas: deletion of 1p and deletion of 19q. oligoastrocytoma, which shared both an astrocytic and oligodendroglial
Oligodendrogliomas exhibit loss of heterozygosity (LOH) at 19q in component; this is discouraged in the 2016 WHO classification. The
up to 88% of cases and LOH at 1p in up to 100% of cases.233 two entities can now be molecularly distinguished through the use
930 Part III: Specific Malignancies

of 1p/19q codeletion, which is seen in oligodendroglioma but not in highly expressed, possibly as a result of higher levels of necrosis and
astrocytoma.216 inflammatory infiltrates seen in this class.239
The proneural subtype samples were associated with alterations
Genome Level Classification of Glioblastoma in PDGFRA and IDH1. Samples in this group also contained most
of the TP53 mutations and TP53 LOH. Less common in proneural
Profiling and sequencing results are now being used to divide samples were chromosome 7 amplification and chromosome 10 loss.
glioblastomas into molecular subgroups. Phillips and colleagues236 The proneural group showed high expression of oligodendrocytic
used gene expression profiling to classify glioblastoma into three genes PDGFRA, NKX2-2, and OLIG2. Other proneural develop-
groups: proneural, mesenchymal, and proliferative. More recent ment genes associated with this group include DCX, DLL3, ASCL1,
work from The Cancer Genome Atlas has provided more detailed and TCF4.239
information about the molecular genetic alteration of glioblastoma, The neural subtype was associated with expression of the neuron
resulting in four distinct subtypes that resemble stages of neurogenesis: markers NEFL, GABRA1, SYT1, and SLC12A5.239
classical, mesenchymal, neural, and proneural (Fig. 63.16).237 The These four glioblastoma subtypes also have clinical implications.
sequential genetic changes in the pathogenesis of glioma according Younger patients were overrepresented in the proneural subtype.
to subgroup have been well reviewed by Van Meir and colleagues238 Furthermore, patients with the proneural subtype showed a trend
(Fig. 63.17). toward longer survival.239 Most secondary glioblastomas were classified
The classical subtype is characterized by chromosome 7 amplification as proneural.239 Interesting to note, aggressive treatment reduced
and chromosome 10 loss, which occur in 100% of classical subtypes. mortality in patients with classical and mesenchymal subtype tumors,
This subtype is associated with EGFR amplification in about 97% and efficacy was suggested in the neural group, but it did not alter
of the samples tested. TP53 mutations are also lacking in this subtype. survival in patients classified as having the proneural subtype.239
Other alterations include deletions of CDKN2A and high expression
of the neural precursor and stem cell marker NES, and signaling Other Genetic Changes in Glioblastomas
pathways Notch and Sonic Hedgehog.239 The Rb pathway, which regulates the G1/S transition, commonly is
The mesenchymal subtype is associated with deletions in regions disrupted in high-grade astrocytomas. This disruption can occur through
of the NF1 gene. These samples also expressed the mesenchymal deletion or mutation of RB itself; deletion or mutation of CDKN2A,
markers CHI3L1 and MET. In addition, genes in the tumor necrosis which encodes the cyclin-dependent kinase inhibitor p16; deletion
factor superfamily and in the nuclear factor (NF)- κB pathway are or mutation of CDKN2B, which encodes the cyclin-dependent kinase

Figure 63.16 • Integrated view of gene expression and genomic alterations across four glioblastoma subtypes: (1) proneural, (2) neural, (3) classical, and
(4) mesenchymal. Key genetic for each subtype mutations are highlighted in the right column. (From Brennan C, et al. The somatic genomic landscape of
glioblastoma. Cell. 2013;155[2]:462–477.)
Cancer of the Central Nervous System • CHAPTER 63 931

Putative GBM
cells of origin: Primary GBM subtypes:

EGFR mutation/amplification/overexpression
Neural stem Oligodendroglioma Classical PTEN loss/mutation
cell IDH1/2 mutations CDKN2A loss
NES overexpression
Notch & Shh pathways activation

Transit NF1 loss/mutation


amplifying cell PT53 loss/mutation
Mesenchymal
PTEN loss/mutation
MET, CHI3L1, CD44, MERTK overexpression
TIC BCPC TNF family & NFκB pathways activation
Neural/glial
progenitor
Sequential genetic EGFR mutation/amplification/overexpression
alterations and clonal Neural Gene signature of normal brain
evolution Neuron marker expression
(NEFL, GABRA1, SYT1, SLC12A5)
Remains to be better defined
Astrocyte
PDGFRA amplification
Juvenile pilocytic Proneural IDH1 mutation
Astrocytoma PIK3A/PIK3R1 mutation
Wild type IDH1/2 TP53, CDKN2A & PTEN loss/mutation
BRAF mutation Proneural marker expression
(SOX, DCX, DLL3, ASCL1, TCF-4)
Oligodendrocytic marker expression
BCPC
Grade II/III Secondary GBM (PDGFRA, OLIG2, TCF3 and NKX2-2)
Astrocytoma HIF, PI3 kinase & PDGFRA pathways
Oligodendrocyte IDH1/2 mutations activation

Figure 63.17 • Sequential genetic changes observed in the pathogenesis of different subtypes of glioblastoma. Some cells in the normal brain undergo
genetic alterations, which leads to a population of tumor-initiating cells (TICs), which can then further accumulate genetic and epigenetic changes and become
brain cancer–propagating cells (BCPC). The latter cells are responsible for the formation of glioblastoma. EGFR, Epidermal growth factor receptor; GBM,
glioblastoma multiforme; HIF, hypoxia-inducible factor; IDH, isocitrate dehydrogenase; PDGFRA, platelet-derived growth factor receptor–A; PI3 kinase,
phosphatidylinositol-3 kinase; PTEN, phosphatase and tensin homolog; TNF, tumor necrosis factor. (Modified from Van Meir EG, Hadjipanavis CG, Norden
AD et al. Exciting new advances in neuro-oncology: the avenue to a cure for malignant glioma. CA Cancer J Clin. 2010;60[3]:166–193.)

inhibitor p15; or amplification of the CDK4 gene. Ichimura and The p53 pathway frequently is disrupted in all grades of astrocy-
coworkers240 noted that one of these changes had occurred in 67% tomas. This disruption can occur by mutation of p53 itself, by
of glioblastomas and 21% of anaplastic astrocytomas but in none of overexpression of MDM2, which degrades p53, or by mutation or
15 low-grade astrocytomas. These findings suggest that progression deletion of p14ARF, which positively regulates p53 by inhibiting MDM2
to a higher-grade glioma is facilitated by deregulation of the G1/S expression. In one study, the p53 pathway was disrupted by one of
transition. these mechanisms in 67% of diffuse astrocytomas, 72% of anaplastic
LOH of 13q and 9p has been identified in high-grade astrocytomas, astrocytomas, and 76% of glioblastomas.242 Therefore disruption of
and these deletions may lead to alterations in the Rb pathway. LOH the p53 pathway appears to be an early event in the genesis of
of chromosome 13 occurs in 30% to 40% of higher-grade astrocy- astrocytomas. It is possible that mutation of p53 in low-grade astro-
tomas.241 The Rb gene (RB), which maps to 13q14, is the likely target cytomas leads to genomic instability that sets the stage for additional
of this deletion. CDKN2A is located on 9p21, and this chromosomal mutations that lead to the formation of higher-grade tumors. The
region is homozygously deleted in at least 30% to 40% of glioblastomas p53 gene (TP53) is located on the long arm of chromosome 17 (17p),
and in approximately 10% of anaplastic astrocytomas, but never in which frequently is lost in both low-grade and high-grade astrocytomas,
diffuse astrocytomas.241 Because two different gene products, CdkN2A suggesting that p53 is the target for this genetic change (reviewed by
and p14ARF, are both transcribed from a single locus on 9p21, deletion Ichimura and coworkers241).
of this region leads to simultaneous disruption of both the Rb and EGFR amplification leading to overexpression is seen in
p53 pathways. approximately 35% of primary glioblastomas, rarely in anaplastic
Astrocytic tumors also frequently display high-level expression of astrocytomas, and never in diffuse astrocytomas.31 Another 15%
PDGF ligands and receptors.32 Most studies have found that the of primary glioblastomas overexpress EGFR without amplification.
PDFGα receptor is overexpressed in 60% to 90% of low- and high- Approximately half of glioblastomas with EGFR amplification express
grade astrocytomas. By contrast, overexpression of the ligand PDGFA a mutant form of the receptor.31 The most common mutant is known
or PDGFB is not very common in low-grade gliomas but is seen in as ΔEGFR or EGFRvIII, which is missing exons 2 to 7, resulting in
higher-grade tumors, suggesting that this growth factor participates an in-frame deletion of 801 base pairs of the coding sequence of the
in an autocrine loop in these tumors.32 extracellular domain.243,244 This particular mutant form of EGFR is
932 Part III: Specific Malignancies

constitutively activated and cannot be downregulated. Its expression craniotomy for resection of the malignant glioma located in eloquent
in glioblastoma cells has been associated with increased proliferation, brain areas.
decreased apoptosis, and increased tumorigenicity and invasion in To specifically address the role of aggressive resection of gliomas
vivo.244 Even in glioblastomas that overexpress wild-type EGFR, which in prolonging survival, Lacroix and coworkers138 performed a retrospec-
is not constitutively active, the ligands EGF and TGF-α often are tive multivariate analysis of outcomes in 416 patients through use of
coexpressed, which may activate an autocrine loop that allows for prospectively collected data. Postoperative volumetric MRI scans were
self-stimulation.31 obtained for all patients so that a blinded neuroradiologist was able
Up to 90% of primary glioblastomas exhibit deletion of 10q, on to determine the extent of resection given as a percentage of the
which is located the tumor suppressor gene PTEN.245 The retained original tumor. Patients with a resection of 98% or more of the tumor
PTEN allele is mutated in approximately 50% of tumors with 10q volume had a median survival period of 13 months, versus 8.8 months
loss, leading to complete loss of functional PTEN protein. Therefore for patients with a less than 98% resection. Also found to be significant
the incidence of PTEN mutation in glioblastoma is at least 45%.241 were age, KPS score, and extent of tumor necrosis. However, when
One study suggested a potential association between lower PTEN statistical analysis controlled for these other three factors, extent of
levels and shorter survival in patients with glioblastomas, although resection remained a significant variable. More recently, Sanai and
this association did not reach statistical significance.246 PTEN mutations coworkers140 analyzed the effect of extent of resection in 500 consecu-
have been found in anaplastic astrocytomas, although at a much lower tively treated patients with newly diagnosed glioblastoma multiforme
frequency than in glioblastomas.247 This study found the presence of (GBM). The authors found that extent of resection equal to or greater
PTEN mutations in anaplastic astrocytoma to be a powerful prognostic than 78% conferred a survival benefit, and reasoned that 78% should
factor portending a poorer outcome. be the new threshold minimum. Using recursive partition analysis,
In contrast to primary tumors, secondary glioblastomas rarely the group further demonstrated that a 95% extent of resection is the
display EGFR amplification.248–250 Secondary glioblastomas have loss single best predictor of survival in patients with glioblastoma.
of 10q, but they do not have mutations of the retained PTEN gene.249
Both the p53 and Rb pathways generally are disrupted in secondary Navigation During Surgery
glioblastomas, as is the case with primary glioblastomas; however, the Intraoperative MRI scanners have allowed for real-time evaluation of
mechanisms tend to be different. Both wild-type p53 alleles are lost the extent of tumor resection. Imaging of relevant anatomy can be
in more than half of all secondary glioblastomas, generally one allele performed during surgery with MRI to provide real-time feedback
by deletion and the second by mutation. By contrast, p53 mutations regarding the surgical resection. Schulder and colleagues257 reviewed
are rarely seen in primary glioblastomas (occurring in fewer than 10% the cases of 93 patients who underwent neurosurgery aided by a
of cases).248,250 The Rb pathway is disrupted in secondary glioblastomas, low-field intraoperative MRI scanner. These investigators found that
often through loss of both wild-type alleles.241 Secondary glioblastomas surgery was directly affected by imaging in 51% of the operations.
may also show disruption of the p53 and Rb pathways through promoter Second lesions not otherwise evident on the initial preoperative scan
methylation of CdkN2A and p14ARF. were seen in 21 patients, and in another 14 patients, unnecessary
dissection was avoided as a result of real-time imaging. Intraoperative
Surgery for Supratentorial Gliomas: Extent of MRI scanners are being used not only for anatomic imaging but also
Surgical Resection for perfusion and MRA imaging. Frameless stereotactic units have
been available for many years, allowing preoperative MRI scans to be
The role of extent of surgical resection in the treatment of gliomas downloaded to an intraoperative computer workstation to create a
has been controversial for many years.138,252,253 Virtually all of the three-dimensional reconstruction of the images. The patient’s head
reviews in this subject area have been retrospective, and thus they are can be directly referenced to the images for precise intraoperative
subject to significant selection bias. It is likely that patients with the localization to less than 2 mm. Investigators have been able to import
most favorable, easily resectable lesions within noneloquent brain PET, functional MRI, and DTI data into the frameless stereotactic
regions have tended to undergo aggressive resections, whereas those units to map out functional brain areas during surgery.119
with deeper lesions in more eloquent brain regions have been selected Intraoperative cortical and subcortical mapping has been used to
for stereotactic biopsy to minimize surgical morbidity. Some experts define eloquent brain areas during resection of gliomas.258,259 Intra-
have advocated maximal resection for gliomas, reasoning that this operative cortical mapping has enabled neurosurgeons to become more
approach is associated with improved survival in children, adults, and aggressive with tumors located within eloquent brain regions. This
even elderly persons.139,140,254 Others have advocated for supratotal application is especially useful with low-grade gliomas that occur
resection in low-grade glioma, with retrospective data demonstrating immediately adjacent to motor and speech areas. Cortical mapping
prolonged survival.255 A possible explanation for this outcome may may be performed with the patient lightly anesthetized but awake
be that fewer cells are left behind that need to be eradicated with enough to respond to questions so that speech areas can be mapped.
irradiation and chemotherapy. This technique has been shown to facilitate the extent of resection,
Other investigators have advocated stereotactic biopsy alone as the thereby improving long-term survival in patients with low-grade
preferred mode of histologic diagnosis in the treatment of patients gliomas.260
with gliomas.253,256 Stereotactic biopsy affords a histologic diagnosis
with a low complication rate (2%–5%). However, stereotactic biopsy Complications of Surgery
may lead to an inaccurate pathologic diagnosis. In a review of data Analysis by the Glioma Outcome Project, a prospectively compiled
for 81 consecutive patients initially diagnosed with stereotactic biopsy database capturing information on 788 patients, reviewed perioperative
who subsequently underwent a craniotomy, Jackson and colleagues252 complications and neurologic outcomes for patients who underwent
showed that the histopathologic classification based on stereotactic craniotomy for the diagnosis and treatment of gliomas.261 Of these
biopsy findings was incorrect in 38% of the patients. This finding patients, 499 underwent either a first or second craniotomy for treat-
represented sampling error in these patients, 96% of whom underwent ment of their malignant glioma, and the remaining 289 patients
biopsy at outside institutions. Despite the fact that these tumors underwent a stereotactic biopsy only. No difference was found in the
were located in eloquent brain areas and were previously treated by characteristics of the patients who underwent a first or second crani-
neurosurgeons, gross total resection could be achieved in 57% of otomy. Analysis of the perioperative symptoms, however, revealed that
these patients once they reached a major tertiary care hospital (The patients undergoing a second craniotomy had a higher incidence of
MD Anderson Cancer Center, Houston, TX). Major complications altered level of consciousness and papilledema, whereas those undergoing
were seen in only 12.3% of the patients undergoing aggressive a first craniotomy had a higher incidence of headache. The incidence
Cancer of the Central Nervous System • CHAPTER 63 933

of depression was higher in patients undergoing a second craniotomy been superseded by the results of three randomized trials, two from
(11% in the group undergoing the first craniotomy versus 20% in the European Organisation for Research and Treatment of Cancer
the patients undergoing a second craniotomy). This difference may (EORTC) and one from the NCCTG (see Table 63.4). The EORTC
reflect the presence of a chronic disease state. The rate of systemic 22844 trial randomly assigned adults with supratentorial low-grade
infections also was higher in patients undergoing a second craniotomy astrocytomas, oligodendrogliomas, or mixed oligoastrocytomas to receive
at 4.4%, versus 0% in patients undergoing a first craniotomy. This either 45 Gy or 59.4 Gy of radiation after surgery.263 The NCCTG
finding is not unexpected, because these patients frequently have been study was similar except that patients were randomly assigned to
heavily pretreated with radiation therapy and chemotherapy, in addition receive either 50.4 or 68.4 Gy after surgery.148 Patients underwent a
to long-term steroid use, causing significant immunosuppression. range of surgical procedures, including biopsy and subtotal or gross
Postoperative neurologic status was the same or better in 92% of the total resection. Neither study showed any benefit to the higher dose
patients undergoing their first craniotomy, whereas this rate dropped in terms of OS or PFS. If anything, the NCCTG study showed a
to 82% in patients undergoing their second craniotomy. Nevertheless, worse survival rate in patients receiving the higher dose of radiation
the overwhelming majority of patients benefited from a debulking (64.8 Gy). The 5-year survival rate in both studies ranged from
procedure to reduce neurologic deficits. 58% to 72%.
The most important preoperative factor associated with good The EORTC also performed study 22845, in which patients who
neurologic outcome has been the KPS score. Patients with higher underwent surgical treatment for astrocytomas, oligodendrogliomas,
KPS scores fared better with surgery than did patients with lower or mixed oligoastrocytomas were randomized to receive either adjuvant
scores. This finding is a reflection of degree of neurologic injury. The irradiation at a dose of 54 Gy or no up-front radiation therapy.264
Glioma Outcome Project revealed that the incidence of further Patients in the latter group had the option of receiving radiation if
neurologic deficit with craniotomy is only 8% in patients undergoing progression of disease was observed. No difference was seen in OS
their first operation versus 18% in patients undergoing a second between the early radiotherapy and observation groups (Table 63.5);
craniotomy for resection of a malignant glioma. In view of the severity the 5-year survival rate was 68% versus 66%, and the median survival
of the disease, this risk is certainly acceptable. Of greater importance, period was 7.4 versus 7.2 years. However, a statistically significant
prospectively obtained data show improvement in neurologic outcome advantage was observed in the patients who received radiation in
with aggressive debulking surgery. An important point is that these terms of PFS (55% versus 35% at 5 years; median time to progression,
data were compiled from participating institutions, and the decision 5.3 versus 3.4 years; P < .0001). The absence of any difference in
about whether to perform surgery was left to the discretion of the survival has supported the position of experts who advocate delaying
surgeon; therefore selection bias certainly played a role in terms of radiation. Conversely, persons favoring up-front radiation in the
who underwent radical surgery versus stereotactic biopsy. The investiga- treatment of low-grade gliomas have cited the improved PFS in the
tors do not discuss the frequency of tumors in eloquent versus irradiated group and the fact that at 1 year, seizures were better
noneloquent brain regions. Box 63.1 summarizes the recommended controlled in patients in the radiation treatment arm.
approach to management of supratentorial gliomas. These randomized trials confirmed the importance of certain
prognostic factors in low-grade gliomas. In the NCCTG trial, three
Radiation Therapy for Supratentorial Gliomas prognostic factors—histologic subtype, patient age, and tumor
size—were consistently associated with OS in multivariate analysis.148
Radiation Therapy for Low-Grade Gliomas The 5-year survival rates for patients with oligodendroglioma-
Numerous retrospective reports have been published regarding the use predominant tumors and those with astrocytomas were 74% and
of radiation therapy for low-grade gliomas (summarized by Leighton 56%, respectively (P = .0001); for patients younger than 40 years and
and associates262). The results of these retrospective studies have largely those aged 40 years or older, 5-year survival rates were 77% and 60%,
respectively (P = .025); and for preoperative tumor size less than 5 cm
and 5 cm or greater, 5-year survival rates were 81% and 61%,
respectively (P = .008). In the EORTC 22844 study, the extent of
Box 63.1. MANAGEMENT OF SUPRATENTORIAL resection had a great impact on OS; patients who had more than
ASTROCYTOMAS 90% of their tumor removed did much better than did those who
underwent a biopsy. Multivariate analysis of data from the two EORTC
• Grade I (pilocytic) astrocytomas: Surgery is curative. If residual trials confirmed that astrocytoma histologic characteristics, age older
tumor is seen on postoperative images, the patient should undergo a than 40 years, and preoperative size greater than 6 cm all were unfavor-
second craniotomy to resect the entire tumor. Radiation therapy and able prognostic factors but also uncovered a few other factors, such
chemotherapy have limited usefulness for the treatment of these as tumor crossing the midline and the presence of neurologic deficits
tumors. Radiation can be considered for recurrent or unresectable before surgery.265
tumors. On the basis of these randomized trials, a reasonable management
• Grade II (low-grade) astrocytoma: Surgery is the mainstay of strategy would be observation in a patient with a completely resected
therapy for tumors in noneloquent regions of brain. In patients low-grade glioma in patients younger than 40 years. However, many
younger than 40 years who undergo gross total resection, no investigators still advocate radiation after complete resection in patients
additional therapy is given. Incomplete resection is typically treated older than 40 years or after incomplete resection. The standard dose
with radiation alone; however, increasing evidence suggests that is 54 Gy given in 1.8-Gy daily fractions over 6 weeks. It is common
chemotherapy improves survival. to treat the MRI-defined GTV with a 1-cm margin. Additional
• Grade III astrocytoma (anaplastic astrocytoma) and grade IV prognostic factors include IDH mutation status and 1p/19q deletions,
gliomas (glioblastoma): Surgery is required to establish tissue which have been reviewed previously.
diagnosis, preferably also with debulking. Chemotherapy is begun
with temozolomide during radiation therapy and continued for Radiation Therapy for High-Grade Gliomas
six cycles after completion of the radiation regimen. The radiation The earliest randomized trial to demonstrate that radiation therapy
dose generally is 60 Gy. Tumor tissue is sent for analysis of O6- was beneficial in the treatment of high-grade gliomas was conducted
methylguanine DNA-methyltransferase (MGMT) promoter activity. by the Brain Tumor Study Group (BTSG 69-01)266 (Table 63.6).
Temozolomide is offered to all patients, however, regardless of Patients were randomly assigned to one of four groups: supportive
promoter methylation status. care, whole-brain radiation therapy (WBRT), BCNU, or WBRT plus
BCNU. This trial clearly showed a benefit for radiation alone versus
934 Part III: Specific Malignancies

Table 63.5 Results of Treatment for Low-Grade Gliomas: Select Randomized Trials
Treatment No. of
Study Years Histologic Type Protocol Patients 5-Yr Survival (%) 5-Yr PFS (%)
Karim et al 263
1985–1991 9% A, grade 1 45 Gy 171 58, P = .94 47, P = .73
EORTC 22844 60% A, grade 2 59.4 Gy 172 59 50
22% O
9% mixed
van den Bent et al264 1986–1997 2% A, grade 1 Observation 157 66, P = .87 35, P < .0001
EORTC 22845 60% A, grade 2 54 Gy 154 68 55
25% O
10% mixed
Shaw et al148 1986–1994 32% A or mixed (A > O) 50.4 Gy 101 72, P = .48 55, P = .65
NCCTG 68% O or mixed (O > A) 64.8 Gy 102 65 52
Shaw et al719 1998–2002 See footnotea 54 Gy 126 63, P = .33 46, P = .06
RTOG 98-02 (randomized arms) 54 Gy + PCV 126 72 63

a
Results of this study are currently available only in abstract form. Histologic type was not available in the abstract.
A, Astrocytoma; EORTC, European Organisation for Research and Treatment of Cancer; NCCTG, North Central Cancer Treatment Group; O, oligodendroglioma; PCV,
procarbazine, cisplatin, vincristine; PFS, progression-free survival; RTOG, Radiation Therapy Oncology Group.

supportive care, with a median survival of 9 months versus 3.5 months Table 63.6).272 Hyperfractionation also has been used to try to increase
and a 1-year survival rate of 24% versus 3% (P = .001). On the basis the total dose. In the Brain Tumor Cooperative Group (BTCG) study
of this trial, radiation therapy has remained an essential component 77-02, patients who were randomly assigned to receive hyperfractionated
in the treatment of high-grade gliomas. A second randomized trial WBRT with BCNU showed no difference in survival compared with
from the Scandinavian Glioblastoma Study Group confirmed the those who received conventional WBRT (60 Gy).273 In RTOG trial
efficacy of radiation with a lower dose of WBRT (45 Gy) compared 90-06, patients were randomized to receive either 60 Gy in conventional
with supportive care267 (see Table 63.6). fractionation or 72 Gy in a hyperfractionated regimen (1.2 Gy twice
The results from BTSG 69-01 and two successive BTSG studies daily). No differences in survival were found between the two groups.274
were pooled to examine dose response.268 The original studies were Radioactive implants also have been used to increase dose locally
all randomized, but patients were not randomly assigned to different to the tumor bed. Single-institution data suggested improved outcome
radiation doses, and thus the analysis of dose was retrospective. With with this approach compared with conventional radiotherapy.275 In
this caveat, the study showed a benefit to use of 60 Gy versus 50 Gy the BTCG 87-01 trial, patients were randomized to receive either an
(median survival, 10.5 versus 7 months; P = .004). iodine-125 (125I) seed implant (60 Gy) or no implant at surgery (see
The current standard of care in Radiation Therapy Oncology Group Table 63.6). Thereafter, patients received 60 Gy via external beam
(RTOG) studies is to define the initial volume as the preoperative radiation. Remarkably, no survival benefit was found for use of 120 Gy
lesion with edema as seen on T2-weighted MRI images with a 2-cm delivered with brachytherapy and external beam irradiation.276 Similarly,
margin. This volume is treated with 46 Gy, followed by a boost to a trial conducted at the Princess Margaret Hospital that combined
125
60 Gy to the gadolinium-enhancing lesion seen on T1-weighted images I implants with external beam radiation showed no improvement
with a 2.5-cm margin. with the implant.
By pooling the results of three RTOG studies and using a non- An implantable balloon has been developed for the delivery of
parametric recursive partitioning analysis (RPA), Curran and associates269 brachytherapy (GliaSite). This device has traditionally used a liquid
identified several significant prognostic factors for survival in patients form of 125I (Iotrex) as the source for brachytherapy. This device
with high-grade gliomas: histologic type, KPS score, age, neurologic provides brachytherapy directly to the resection cavity, with minimal
function, and duration of symptoms. Patients could be placed into exposure of unaffected brain to the radiation source, and is used for
groups I through VI with different outcomes on the basis of these treatment of primary gliomas and metastatic tumors.
factors. Classes I and II, which are only found in the RPA for anaplastic Another means of increasing dose to the tumor region after
astrocytomas and not in the RPA classification for glioblastoma, have conventional radiation therapy has been stereotactic radiotherapy. This
the best outcomes, whereas classes V and VI have the worst outcomes. approach was tested in RTOG study 93-05, in which patients in the
The prognostic significance of the RPA classes was confirmed even control group received standard fractionated radiotherapy (60 Gy in
in the era of temozolomide treatment for glioblastoma (discussed in 30 fractions) with BCNU chemotherapy, whereas patients in the
the next section).270 These RPA classes have been updated to reflect experimental group received a radiosurgery boost (16 to 24 Gy based
the more than 16,000 patients in the RTOG glioma database.271 The on size) before external beam radiation therapy. Results showed no
new RPA classification simplified the original model by combining significant difference in 2- and 3-year survival rates or in patterns of
classes V and VI for patients with glioblastoma, resulting in three failure between the two arms of the study.277
distinct prognostic groups for glioblastoma defined by age, performance The one agent that has clearly been shown to be of benefit when
status, extent of resection, and neurologic function. combined with radiation is the alkylating agent temozolomide. The
Because of the poor survival of patients with high-grade astrocy- results with this drug for treatment of glioblastomas are discussed in
tomas, numerous strategies have been tried to improve the results depth in the next section.
with irradiation. These efforts fall into two main classes: first, increasing
radiation dose, and second, modulating the radiation response. Doses Tumor-Treating Fields for Glioblastoma
higher than 60 Gy have been used, but to no avail. The RTOG
74-01–ECOG 1374 trial randomized patients between receiving 60-Gy It has been demonstrated that the use of tumor-treating fields, with
WBRT and a total dose of 70 Gy (60-Gy WBRT followed by a 10-Gy alternating electric fields applied to the scalp, confers a survival advantage
boost) but showed no improved survival with the additional dose (see when performed in addition to standard maintenance temozolomide
Cancer of the Central Nervous System • CHAPTER 63 935

Table 63.6 Results of Treatment for High-Grade Astrocytomas: Select Randomized Trials Focusing on
Radiation and Radiation Modifiers
Median 18-Mo
Gliomas Treatment No. of Survival Survival
Study Years (%) Protocol Patients (mo) (%) Comment(s)
Walker et al282 1969–1972 90 Supportive care 31 3.5 0 RT superior to supportive
BTSG 69-01 BCNU 51 4.6 4 care (P = .001)
RT (60 Gy WBRT) 68 9 4
RT (60 Gy WBRT) + 72 8.6 18 RT + BCNU superior to
BCNU supportive care (P = .001)
Kristiansen et al267 1974–1978 — Supportive care 38 5.2 0 RT superior to supportive
care
SGSG RT (45 Gy WBRT) 35 10.8 13
RT (45 Gy WBRT) + 45 10.8 13
Bleo
Bleehen and 1983–1988 — RT (45 Gy) 144 — 11 60 Gy superior to 45 Gy
Stenning720 RT (60 Gy) 299 — 18 (P = .04)
Chang et al272 1974–1979 80 RT (60 Gy WBRT) 148 9.9 19
RTOG 74-01 RT (60 Gy WBRT) + 105 8.4 22 No difference between any
10 Gy boost groups
RT (60 Gy WBRT) + 165 10.0 29 No improvement with 70 Gy
BCNU
RT (60 Gy WBRT) + 136 9.8 26
DTIC + MeCCNU
Curran et al269 RT (60 Gy) + BCNU 13.2 No improvement with HFX
RTOG 90–06 RT (72 Gy HFX) + 11.2 For patients <50 yr, median
BCNU survival better with standard
fractionation
Selker et al276 1987–1994 85 125
I implant (60 Gy) 137 17 56 No improvement with
(10% AA) + BCNU + RTa implant
(60.2 Gy)
BTCG 87–01 RTa (60.2 Gy) + 133 14.8 44
BCNU
Laperriere et al721 1986–1996 92 RT (50 Gy) + BCNU 69 13.2
PMH RT (50 Gy) + BCNU 71 13.8 No improvement with
+ 125I implant implant
(60 Gy)
Nelson et al722 1979–1983 83 RT + BCNU 146 12.4 34
RTOG 79–18 RT + Miso + BCNU 147 10.7 27 No improvement with
misonidazole
Prados et al723 1994–1997 0b RT (60 Gy) + PCV 134 — 74
RTOG 94–04 RT (60 Gy) + BUdR 134 — 62 No improvement with BUdR;
+ PCV closed early at interim
analysis
Souhami et al277 1994–2000 100 RT (60 Gy) + BCNU 186 (total in 14.1 22 (2 yr)
both arms)
RTOG 93–05 Radiosurgery 13.7 18 (2 yr) No improvement with
boost + RT (60 Gy) radiosurgery boost
+ BCNU
Stupp et al286 2000–2002 100 RT (60 Gy) 286 12.1 10.9 (2 yr)
EORTC NCIC RT (60 Gy) + 287 14.6 27.2 (2 yr) Combined therapy was
26981-22981/CE.3 concurrent and beneficial in all prognostic
adjuvant TMZ groups; methylation of
MGMT promoter was
greatest predictor for
benefit from TMZ

a
During the early part of the study, external beam radiation was given to whole brain to a dose of 43 Gy, followed by a boost to the tumor volume for an additional 17.2 Gy.
From May 1989, WBRT was dropped, and the entire dose was restricted to tumor volume.
b
All patients in this trial had AAs.
AA, Astrocytoma; BCNU, carmustine; Bleo, bleomycin; BTCG, Brain Tumor Cooperative Group; BUdR, bromodeoxyuridine; DTIC, dacarbazine; EORTC, European Organisation for
Research and Treatment of Cancer; HFX, hyperfractionation; 125I, iodine 125; MeCCNU, semustine; MGMT, O6-methylguanine-DNA methyltransferase; Miso, misonidazole; NCIC,
National Cancer Institute of Canada; PCV, procarbazine, cisplatin, vincristine; RT, radiation therapy; RTOG, Radiation Therapy Oncology Group; SGSG, Scandinavian Glioblastoma
Study Group; TMZ, temozolomide; WBRT, whole-brain radiation therapy.
936 Part III: Specific Malignancies

therapy compared with maintenance temozolomide therapy alone.278 In in newly diagnosed glioblastomas.285 This study randomized 573
this study, all patients had completed standard chemoradiation therapy. patients to receive 60 Gy of standard fractionated radiation or the
PFS was 7.1 months in the tumor-treating fields plus temozolomide same radiation regimen with daily temozolomide at 75 mg/m2 followed
group compared with 4.0 months in the temozolomide alone group. by six cycles of adjuvant temozolomide (150–200 mg/m2 for 5 days
Furthermore, median OS in the per-protocol population was 20.5 during each 28-day cycle). The patients in the radiation plus temo-
months in the tumor-treating fields plus temozolomide group versus zolomide arm had a statistically significant improvement in median
15.6 months in the temozolomide-alone group. The tumor-treating survival compared with patients in the radiation-only arm (P < .001).
fields approach uses electric fields applied to the scalp at 200-kHz At follow-up evaluation at a median of 28 months, the median survival
frequency with four transducer arrays. These fields are applied, after periods in the two arms were 14.6 and 12.1 months, respectively. The
the head has been shaved, with a device contacting the scalp for up 2-year OS rates were 26.5% and 10.4%, respectively. The Stupp
to 22 hours per day with 2 to 3 days off treatment per 4 weeks of protocol (named after lead author Roger Stupp) of maximal safe
treatment. The mechanism of action of these tumor-treating fields resection followed by radiotherapy plus concomitant and adjuvant
is believed to be due to disruption of mitosis through perturbation temozolomide currently remains the standard of care for glioblastoma
of chromosome segregation.279,280 The nature of this mechanism has (Fig. 63.18). Evaluation of these patients confirmed the 5-year durable
been questioned with potential confounding factors for this survival survival benefit of the regimen (Fig. 63.19A).286
benefit. One criticism was the lack of placebo control; there was no Hegi and coworkers287 subsequently evaluated the status of the
sham study group, that may have led to an adherence bias. Another epigenetic silencing of the MGMT DNA repair gene (MGMT) by
criticism was the observation that the tumor-treating fields group promoter methylation in patients enrolled in this trial. In a subgroup
received six cycles of adjuvant chemotherapy compared with four cycles analysis of 206 patients, MGMT promoter methylation occurred in
in the temozolomide-alone group before randomization, which may 45% of these cases and was found to be an independent favorable
also have confounded the results.281 Further follow-up and investigation prognostic factor regardless of treatment. OS of the methylated promoter
into the use of tumor-treating fields may provide insight into the group was 46% at 2 years and 14% at 5 years (P < .001). Patients
mechanisms by which prolonged PFS and OS are conferred. with a methylated MGMT promoter had better survival than those
without such a promoter (see Fig. 63.19B). The median, 2-year, and
5-year survival rates for patients receiving radiation versus radiation
Chemotherapy for Gliomas and temozolomide based on MGMT promoter methylation status
Chemotherapy for Newly Diagnosed High-Grade are shown in Table 63.7. Even among patients with unmethylated
MGMT, a trend toward increased survival is recognized for those
Astrocytomas receiving temozolomide. These data have made irradiation plus adjuvant
Although prior studies demonstrated efficacy for chemotherapeutic temozolomide the standard of care for all patients with glioblastoma
agents such as carmustine282,283 and procarbazine, lomustine, and regardless of MGMT status. The relative contributions of oral daily
vincristine (PCV),284 the introduction of temozolomide has dramatically temozolomide during radiation versus after radiation are not clear,
altered the treatment of high-grade gliomas and currently remains the and the optimal dose of temozolomide to inhibit MGMT activity
standard of care. Temozolomide is an oral alkylating agent whose has not been established and is the subject of ongoing trials. In the
levels are not affected by AEDs or other hepatic enzyme–inducing current RTOG trial, patients with newly diagnosed glioblastoma are
drugs. Toxicity with this drug is relatively mild, and CNS penetration randomized to receive standard chemoradiation therapy followed by
is good. The pivotal study conducted jointly by the EORTC (trial 12 rather than 6 monthly cycles of adjuvant temozolomide at a standard
22981-26981) and the National Cancer Institute of Canada (NCIC) dosage, or, alternatively, dose-intensive temozolomide (21 days on
(trial CE.3) confirmed the usefulness of temozolomide and radiation and 7 days off ). Newly diagnosed glioblastoma treated with radiation

4 weeks
TMZ daily ¥ 42d 5d 5d 5d ¥ 6 cycles
4 weeks

1 6 10 14 18 22 week

RT 30 ¥ 2 Gy

RT; focal radiotherapy, 60 Gy in 6 weeks to tumor volume + 2-3 cm margin


TMZ; Temozolomide
During RT: 75 mg/m2 daily (including weekends) for up to 49 days.
Administration 1-2 hours before RT or in a.m. on days without RT.
Antiemetics: metoclopramide, usually needed only before initial doses.
Maintenance: 150-200 mg/m2 daily ¥ 5, for up to 6 cycles
Antiemetic prophylaxis with 5-HT3 antagonist or metoclopramide

Pneumocystis carinii prophylaxis during continuous TMZ administration only (lymphocytopenia)


Pentamidine inhalations or trimethoprim/sulfamethoxazole 3¥/week

Figure 63.18 • Outline of Stupp protocol, which consists of surgical resection followed by regimen of concomitant chemotherapy and radiotherapy (RT)
followed by maintenance chemotherapy. TMZ, Temozolomide. (From Amouheidari A, et al. The nexus between interdisciplinary approach and extended
survival in CNS tumors. Neuro-Oncology Scientific Club [NOSC] meeting report, 27 December 2012, Isfahan, Iran. Res Cancer Tumor. 2013;2[1]:1–9.)
Cancer of the Central Nervous System • CHAPTER 63 937

100
90 Combined
Radiotherapy
80
70 p < 0.0001

Survival (%)
60
50
40
30
20
10 n = 287
n = 286
0
0 1 2 3 4 5 6 7
A Time (years)
Number at risk
Combined 254 175 76 39 23 14 6
Radiotherapy 278 144 31 11 6 3 0

100
Figure 63.19 • Survival of patients with glioblastoma based 90 Combined
on temozolomide administration or MGMT status. Patients Radiotherapy
enrolled in the European Organisation for Research and Treat- 80
ment of Cancer trial 22981–26981 and National Cancer Institute
of Canada trial CE.3 were randomized to receive radiotherapy 70 p = 0.004
alone or radiotherapy plus temozolomide. (A) Kaplan-Meier
Survival (%)

60
estimates of overall survival according to treatment group. The
hazard ratio for death among patients who received temozolomide 50
compared with those who received radiotherapy alone was 0.63
(95% confidence interval [CI], 0.53–0.75; P < .0001). (B) In 40
a subset of the patients enrolled in the trial in (A) the methylation
status of the O6-methylguanine DNA methyltransferase (MGMT) 30
gene promoter was assessed. The curves in (B) represent Kaplan- 20
Meier estimates of overall survival according to MGMT promoter
methylation status. The difference in survival between patients 10 n = 46 n = 46
with a methylated MGMT promoter and those with an
unmethylated MGMT promoter was highly significant (hazard 0
ratio, 0.49 [95% CI, 0.32–0.76]; P = .001), indicating that the 0 1 2 3 4 5 6 7
MGMT promoter methylation status has prognostic value.
(Modified from Stupp R, Hegi ME, Mason WP, et al. Effects B Time (years)
of radiotherapy with concomitant and adjuvant temozolomide Number at risk
versus radiotherapy alone ion survival in glioblastoma in a Combined 37 35 22 11 6 2
randomized phase III study: 5-year analysis of the EORTC-NCIC Radiotherapy 43 30 11 3 1 0
trial. Lancet Oncol. 2009;10[5]:459–466.)

therapy and temozolomide is now the standard comparison arm for tried. Any benefit from chemotherapy for recurrent disease (Fig. 63.20)
phase II studies seeking novel effective agents. must be measured against the roughly 4- to 5-month median survival
Even though the cited data on temozolomide were generated after repeat surgical removal of recurrent high-grade astrocytic tumors.
in patients with glioblastomas, many oncologists have extrapolated An important recent observation is that MGMT promoter methylation
these findings to anaplastic astrocytomas and have used this agent status can change between the first surgery and second surgery for
concurrently with radiation in this setting. However, some experts have recurrence.287
questioned whether concomitant temozolomide should be considered Some patients with recurrent high-grade astrocytomas are offered
standard therapy in patients with anaplastic gliomas in the absence of reresection with use of either GliaSite (balloon catheter with liquid
randomized data. radiation source) or Gliadel (intracavitary biodegradable BCNU wafers).
A placebo-controlled multicenter study in 22 patients reported a median
Chemotherapy for Recurrent High-Grade Astrocytomas survival of 31 weeks in patients receiving BCNU polymers, compared
Patients with high-grade astrocytomas in whom initial therapy with with 23 weeks in the patients receiving placebo polymers.289 The use
radiation and temozolomide fails to provide benefit are candidates of Gliadel is limited to patients who can have meaningful reresection
for participation in phase I or II trials. In patients for whom some of tumors that are relatively well circumscribed, unilateral, and in
interval has elapsed between initial temozolomide therapy and failure noneloquent brain locations—a situation not typical for a majority
to respond, retreatment with low-dose daily temozolomide may be of anaplastic astrocytic tumors.
938 Part III: Specific Malignancies

Table 63.7 Results of Treatment for Glioblastomas Stratified by MGMT Promoter Status: EORTC Trial
22981-26981 and NCIC Trial CE.3
MGMT PROMOTER STATUS/TREATMENTa
UNMETHYLATED (n = 114) METHYLATED (n = 92)
Radiation Only Chemoradiation Radiation Only Chemoradiation
(n = 54) (n = 60) (n = 46) (n = 46)
Median overall survival (mo) 11.8 12.7 15.3 21.7
2-yr survival (%) 1.8 14.8 23.9 48.9
(P = .06) (P = .007)
5-yr survival (%) 0 8.3 5.2 13.8

a
Either radiation therapy only or chemotherapy plus radiation therapy.
EORTC, European Organisation for Research and Treatment of Cancer; MGMT, O6-methylguanine DNA methyltransferase; NCIC, National Cancer Institute of Canada.
Modified from Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:997; and Stupp R, Mason
WP, Vanden Bent MJ, et al. Effects of radiation therapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma: 5-year analysis of the
EORTC-NCIC trial. Lancet Oncol. 2009;10(5):459–466.

Figure 63.20 • Recurrence just outside the repeat resection margin of a patient with a 5-year survival period after right frontal glioblastoma. Investigational
options at recurrence include dose-intensified temozolomide, CCNU monotherapy, carmustine wafers, stereotactic radiosurgery with or without bevacizumab,
bevacizumab plus a cytotoxic agent, or Novo TTF-100A. Bevacizumab monotherapy is approved for recurrent glioblastoma. Concern with localized treatment
derives from abnormal fluid-attenuated inversion recovery (left) and T1 with gadolinium (right) signal abnormality crossing the corpus callosum.

Nitrosourea (lomustine, carmustine) and carboplatin-based This PFS benefit may be confounded by the fact that bevacizumab
chemotherapy are second line chemotherapeutics agents. Lomustine produces rapid and marked reduction in edema and contrast enhance-
has shown clinical activity as the control arm in several randomized ment because of changes to tumor vasculature. Although initial MRI
trials.290,291 and clinical changes are often dramatic, durable responses average
Bevacizumab, a humanized monoclonal antibody to VEGF, received only about 4 to 6 months. Discontinuing bevacizumab, a very expensive
US Food and Drug Administration (FDA) approval for recurrent drug, may result in rapid rebound of vasogenic edema, and careful
high-grade gliomas in 2009. The approval in the United States was discussion with patients about when to introduce and when to dis-
based on two clinical trials that demonstrated improved response rates continue the agent is necessary.295,296
either with bevacizumab monotherapy or in combination with irino-
tecan in patients who had received chemoradiation and adjuvant Chemotherapy in Addition to Radiotherapy for
temozolomide.292,293 The safety and efficacy of SRS and adjuvant
bevacizumab have been studied in 63 patients whose median PFS was Low-Grade Astrocytomas
5.2 months with adjuvant bevacizumab versus 2.1 months without Chemotherapy for low-grade astrocytomas usually is reserved for
adjuvant bevacizumab. In 2014, a randomized prospective trial of patients whose unresectable tumors are progressively symptomatic.
bevacizumab as a primary agent demonstrated no significant difference Radiation therapy traditionally has been used as a first-line treatment
in OS (15.7 versus 16.1 months); however, a difference was seen in for progression. Potential toxicity of any proposed therapy for low-grade
PFS (10.7 versus 7.3 months).294 Current studies are investigating the gliomas must be balanced against the long natural history. Particularly
role of bevacizumab for recurrent glioblastoma. in younger patients and in those whose tumors have an oligodendroglial
Cancer of the Central Nervous System • CHAPTER 63 939

component, median survival periods may approach 10 years, with OS from 30.6 months to 42.3 months.304 Subgroup analysis by 1p/19q
5-year survival rates of 75% or higher.128 Quality-of-life considerations, status showed that benefit of PCV may be greater in patients with
with particular attention to cognitive status, justify a prolonged “wait codeleted tumors, which is now a diagnostic criterion for oligoden-
and see” policy in asymptomatic or clinically stable patients with droglioma. Similarly, patients with MGMT promoter methylation
suspected low-grade glioma.296 and IDH-mutated tumors had better OS.306
However, more aggressive treatment for low-grade gliomas has The PCV regimen has significant toxicity including myelosup-
become a new paradigm, and recent studies have investigated the role pression, cognitive or mood change, neuropathy, vomiting, hepatic
of the addition of chemotherapy and radiation in the treatment of dysfunction, and allergic rash. Because temozolomide is also effective
low-grade glioma. In 2016 Buckner and colleagues reported that the for anaplastic oligodendrogliomas and it has a more favorable toxicity
addition of chemotherapy (procarbazine, CCNU, and vincristine) profile, oncologists may elect to treat anaplastic oligodendrogliomas
after radiation resulted in both longer PFS (51% versus 21%) and with this drug. In spite of a lack of randomized data with temozolomide
OS (13.3 versus 7.8 years; P = .003) in patients with grade 2 glioma.297 in anaplastic oligodendrogliomas, many oncologists are currently using
The positive survival difference for patients receiving chemotherapy this drug concomitantly with radiation, extrapolating from the experi-
in addition to radiation therapy became apparent only after several ence with glioblastoma.285
years of follow-up. In this study, patients younger than 40 years had
undergone subtotal tumor resection, whereas patients older than 40 Chemotherapy for Recurrent Anaplastic Oligodendroglioma
had undergone either biopsy or resection of any of the tumor. Some Most patients with recurrent anaplastic oligodendrogliomas who received
of the limitations of this study included the fact that molecular PCV at initial presentation are currently being treated with temo-
diagnostic evaluation for mutations such as IDH1 was limited to a zolomide at relapse. Several small trials have shown the safety and
small portion of the patients studied, thus limiting the assessment of efficacy of temozolomide after prior PCV therapy. An open-label
these molecular markers on outcome. phase II trial of temozolomide in 47 patients with anaplastic oligo-
Currently, Baumert and colleagues are investigating the role of dendroglioma or anaplastic oligoastrocytoma who experienced a relapse
temozolomide as adjuvant therapy for low-grade gliomas.298 At 48 after radiation and PCV chemotherapy demonstrated an objective
months, no significant difference in survival has been detected. However, response rate of 43%, a median PFS of 7.5 months, and a 34% rate
given the known long natural history of low-grade gliomas, differences of disease-free survival at 12 months.307 In another phase II multicenter
may be detected with prolonged follow-up. trial that included patients with anaplastic oligodendroglioma or
anaplastic oligoastrocytoma, a response was seen in 26% of patients
Chemotherapy for Low-Grade Oligodendrogliomas who had previously received PCV therapy and in two out of three
chemotherapy-naïve patients.308 A short time to tumor progression
and Oligoastrocytomas appears to be a bad prognostic factor for response to both first-line
Temozolomide has been used in patients with low-grade oligoden- PCV chemotherapy and temozolomide.
drogliomas or oligoastrocytomas that have recurred. The EORTC In a study performed before the reclassification of oligodendro-
conducted one study using temozolomide for patients who had received glioma, Brandes and associates309 reported on 67 patients with recurrent
PCV chemotherapy and irradiation for their initial disease,299 and anaplastic oligodendroglioma or anaplastic oligoastrocytoma who were
another study for patients who had received surgery and irradiation treated with first-time temozolomide at the time of disease progression
without chemotherapy initially.300 These studies showed response rates after prior surgery and radiotherapy. The overall response rate was
to temozolomide of 25% to 53%. Loss of 1p has been correlated with 46% and was higher in patients with anaplastic oligodendroglioma
response to temozolomide for first-line chemotherapy in patients with than in patients with anaplastic oligoastrocytoma (62% versus 25%;
recurrent oligodendrogliomas.301,302 P = .003). Combined 1p and 19q loss (present in 48% of patients)
significantly correlated with response rate (P = .04), time to progression
Chemotherapy for Newly Diagnosed Anaplastic (P = .003), and OS (P = .0001).
Oligodendrogliomas
Therapy for Elderly Patients With Malignant Gliomas
During the past decade, increasing evidence has accumulated to confirm
that anaplastic oligodendrogliomas are special types of malignant Almost half of patients with glioblastoma are older than 65 years, and
gliomas that are sensitive to chemotherapy. Although the molecular the incidence for all primary CNS tumors is highest among those
markers of chemosensitivity, allelic loss of 1p and 19q, have been aged 75 to 84 years, whereas clinical trial participants have a median
shown to have prognostic significance, the reason for oligodendroglial age between 50 and 55 years.310 Elderly patients with malignant gliomas
chemosensitivity remains unknown. have approximately 6-month median survival rates, which is consider-
In the first large study of chemotherapy for new or recurrent ably shorter than those of younger patients. Brandes and coworkers311
anaplastic oligodendrogliomas, Cairncross and the NCIC conducted evaluated the role of surgery, radiation therapy, and chemotherapy
a multicenter phase II trial of intensive PCV chemotherapy using for the treatment of newly diagnosed glioblastoma in patients older
higher doses of the first two drugs than are found in the standard than 65 years. These investigators found that the main predictive
PCV regimen.303 This study reported a 75% overall response rate, factor in evaluating this treatment was perioperative KPS score. The
with 38% complete responses. Time to tumor progression was at least higher the KPS score, the better the patient fared, regardless of the
16.3 months for the entire eligible group and at least 25.2 months treatment. Patients were stratified into group A (surgery with radiation
for patients who had a complete response. therapy to a dose of 59.4 Gy), group B (surgery, radiation therapy,
Two randomized trials have been conducted comparing radiation and PCV chemotherapy), and group C (surgery, radiation therapy,
only with radiation plus PCV chemotherapy in patients with newly and temozolomide). The median time to disease progression was
diagnosed anaplastic oligodendrogliomas or anaplastic oligoastrocy- significantly better in group C patients. OS was better in group C
tomas.304,305 In RTOG 9402, patients were randomized to receive but did not reach statistical significance. Hematologic toxicity was
radiation only or up to four cycles of intensive PCV chemotherapy higher in patients treated with a PCV regimen than in those treated
followed by radiation.305 In EORTC 26951, patients were randomized with temozolomide.
to receive radiation only or radiation followed by six cycles of standard Another study evaluated the role of debulking craniotomy versus
PCV chemotherapy.304 Long-term follow-up has confirmed an OS stereotactic biopsy in the treatment of malignant gliomas in patients
advantage with the addition of PCV. At a median follow-up of 140 older than 65 years; median survival was 171 days after craniotomy
months, EORTC 26951 showed that the addition of PCV increased versus 85 days after biopsy.312 The difference was not statistically
940 Part III: Specific Malignancies

significant but did suggest that patients undergoing a more aggressive and financial burdens of dealing with a terminal disease as patients
craniotomy survived longer. Both groups of patients tolerated the with breast cancer, lung cancer, and other malignancies, but they have
radiation therapy well. A trial of 85 patients older than 70 years random- unique perspectives in that they are uniformly concerned about the
ized participants to radiation therapy (25 fractions of 1.8 Gy) or best potential for damage to the brain and the effect of cognitive function.
supportive care alone and demonstrated improved survival with radiation Measurement of quality of life is important to elucidate the impact
therapy (median survival of 6.8 versus 4.0 months).313 Two randomized of available treatments on the disease: The goal is to improve survival,
phase III trials compared temozolomide alone with radiation therapy but not at the sacrifice of quality of life.323 The most frequent symptoms
alone in older patients with glioblastoma. The Nordic Glioma Trial in the last month of life are headache, lethargy, depression, dysphagia,
found similar efficacy of temozolomide alone compared with radiation delirium, and seizures. Indications for artificial nutrition and parenteral
alone, whereas the German Neuro-Oncology Working Party NOA-08 administration of drugs may require consideration. End-of-life prefer-
trial found that radiation therapy alone was superior to dose-intensified ences should be discussed early in the disease process with a goal
temozolomide.314,315 Iwamoto and colleagues316 reviewed the Memorial toward meeting patient preferences of palliative in-home care or
Sloan-Kettering experience in 394 patients older than 65 years, about acceptable alternatives. In one Italian study, 82% of patients were
one-third of whom received adjuvant temozolomide after radiation able to be at home with good symptom control through a multidis-
therapy, and they reported improved survival with adjuvant chemo- ciplinary home care unit.324
therapy compared with radiation alone.
On the basis of these results, it appears that patients older than New Approaches to Therapy of Gliomas
65 years who have malignant gliomas may still benefit from aggressive
therapy if they have a reasonable performance status. Therefore older The disappointing results with conventional cytotoxic chemotherapy
age alone should not be a criterion for minimizing therapy. An have led to efforts to find more effective and better-tolerated therapies.
abbreviated radiotherapy regimen may be considered in older patients The challenge of neurooncology is to develop new compounds or
with a poor performance status. Roa and associates317 randomized procedures of gene transfer that specifically target the molecular altera-
patients with glioblastoma who were older than age 60 years to receive tions of glioma tumor cells and restore normal gene expression, cell
a standard course of radiation (60 Gy in 30 fractions over 6 weeks) cycle regulation, and apoptosis.325 Significant progress is likely to come
or a shorter course (40 Gy in 15 fractions over 3 weeks). OS times from technologic developments that allow large-scale genomic profiling
measured from randomization were similar at 5.1 months for standard that distinguishes at the molecular level among histologically similar
radiation therapy versus 5.6 months for the shorter course (P = .57). tumors. Therapeutic strategies then could include some combination
The survival probabilities at 6 months were also similar, at 44.7% for of cytotoxic chemotherapy with biologic agents directed at genetic
standard radiation therapy versus 41.7% for the shorter course. The alterations specific to brain tumors of the appropriate subtype. Current
ongoing NCIC/EORTC 26062-22061 trial for patients with glio- pathways of interest include receptor tyrosine kinases (RTKs) EGFR,
blastoma who are older than 65 years randomizes patients to either PDGFR, and VEGFR, in addition to downstream effectors mammalian
short-course radiotherapy (40 Gy in 15 fractions) or temozolomide target of rapamycin (mTOR), Akt, and MAPK. A number of agents
concurrent with and adjuvant to the same radiotherapy regimen.318 targeting the EGFR receptor and the EGFRvIII variant are being tested.
These EGFR-targeted agents have thus far shown limited usefulness
Quality of Life After Therapy for Gliomas with brain tumors in the clinic and are effective only in patients with
tumors that have a susceptible target.326 In one study, patients who
Quality-of-life issues are paramount in the treatment of patients with had received treatment with gefitinib or erlotinib were analyzed for
gliomas. The oncologist’s obligation to disclose prognosis and consider expression of EGFR, the deletion mutant EGFRvIII, and the tumor
palliative care choices occurs very early in the course of a patient’s suppressor gene PTEN. Tumors that coexpressed EGFRvIII and PTEN
illness, and in patients with glioblastoma who have anticipated survival were most likely to respond to EGFR kinase inhibitors.327
that is usually less than 2 years, medical decision-making capacity RTKs such as PDGF and insulin-like growth factor receptors
may be impaired very early on.319 A survey of patients and caregivers regulate cell proliferation and differentiation. Deregulated RTK signaling
on the experience of being diagnosed with high-grade glioma empha- frequently is found in human astrocytic tumors. In addition, gefitinib
sized that in the setting of the initial diagnosis, detailed discussion of and erlotinib, mentioned earlier, and other inhibitors of RTKs, including
diagnosis and prognosis must be carefully balanced with appreciation imatinib mesylate, have been evaluated in patients with malignant
for the emotional state of those concerned.320 As treatment progresses, glioma. Other agents target farnesyl transferase, histone deacetylase,
adverse effects of both disease and its therapy mount. An analysis of rapamycin (mTOR), protein kinase C, and the ubiquitin-proteosome
quality of life was performed in patients participating in an EORTC pathway. In general, the findings of small early trials have been disap-
trial of randomization to either 59.4 or 45 Gy for low-grade gliomas.263 pointing, although the agents were well tolerated. Most are metabolized
This analysis found that patients receiving the higher dose reported through the cytochrome P450-CYP3A4 system, and significant dose
more fatigue and insomnia immediately after radiotherapy and poorer modifications are necessary in patients with brain tumors who are
emotional functioning 7 to 15 months after institution of therapy.321 taking enzyme-inducing AEDs. Tumor resistance may in part be due
Some of the long-term consequences of radiation therapy as appreciated to cancer stem cells. Cancer stem cells are characterized by the ability
at MRI may include diffuse white matter hyperintensities and brain to self-renew and propagate tumor, and they use many of the same
atrophy correlating with cognitive dysfunction, along with cavernous signaling pathways found in normal stem cells such as Wnt, Notch,
angiomata. and Hedgehog.328,329 Integrins, which are transmembrane surface
The EORTC and NCIC performed a quality-of-life study as part proteins that affect several pathways relevant to glioblastoma, promote
of a randomized trial of radiation versus radiation plus temozolomide proliferation, motility, and angiogenesis and represent another potential
for patients with glioblastoma.322 The study investigators found that target for brain tumor therapy. Currently in development are several
health-related quality-of-life measures (e.g., fatigue, overall health, antiintegrins, of which the best studied is cilengitide, an oral agent
social function, emotional function, future uncertainty, insomnia, and that has been evaluated in several completed trials in patients with
communication deficit) did not deteriorate by a clinically meaningful recurrent glioblastoma; little toxicity and several durable responses
amount in either treatment group over time after treatment, and some occurred, leading to a phase III trial of cilengitide for newly diagnosed
parameters even improved. Furthermore, the addition of the temo- glioblastoma.330 Median survival was 19.7 months overall and 30
zolomide regimen to radiotherapy did not negatively affect health-related months in patients with MGMT promoter methylation. The 2-year
quality of life. Notwithstanding these results, patients with malignant survival of 41% exceeded the 27% milestone of the EORTC temo-
gliomas face enormous stresses. They face the same physical, emotional, zolomide trial.331
Cancer of the Central Nervous System • CHAPTER 63 941

Because gliomas produce specific angiogenic peptides such as VEGF consisted of focal tectal gliomas, which affected young adults and had
(see the molecular genetics section earlier in this chapter) to stimulate a favorable outcome.
new blood vessel formation, angiogenesis inhibitors are a logical drug The conclusion that adults with this disease have a better prognosis
development target. Hypoxic areas within gliomas may stimulate than do children also was reached in a study from the Memorial
angiogenesis and promote invasiveness of glial cells. Many of these Sloan-Kettering Cancer Center of 19 adult patients with brainstem
changes in gene expression are mediated by HIF-1. Antiangiogenic gliomas. The investigators found that the median survival period was
agents directed against VEGF, VEGFR, and other targets are in clinical 54 months and the 5-year survival rate was 45%.349
trials. Bevacizumab, a humanized monoclonal antibody against VEGF,
showed a 63% radiologic response rate in patients with recurrent PRIMARY CENTRAL NERVOUS SYSTEM
high-grade gliomas.332
The transfer of genetic material to tumor cells to make them more LYMPHOMA
susceptible to chemotherapy, or to reverse the alterations that sustain Histopathologic Features
the neoplastic phenotype, has been studied in several centers. Gene
therapy strategies include transfecting antioncogenes or drug-activating PCNSL has been known previously by many other names, including
enzymes to glioma cells. The most extensively studied system is the reticulum cell sarcoma, diffuse histiocytic lymphoma, and microglioma.
“suicide gene therapy” in which the herpes simplex virus thymidine Ninety percent of non–HIV-associated PCNSLs are diffuse large B-cell
kinase (HSV-tk) gene is inserted into a replication defective murine lymphomas, with 10% distributed among low-grade, Burkitt, or T-cell
retrovirus. Clinical studies using this approach have been disappointing lymphomas, the last of which represent 1% of all PCNSLs but are
because of poor tumor cell transfection efficiency, although some long more common in Japan. PCNSL arises from late-germinal center
times to tumor progression have been reported.333–335 A prospective or postgerminal center lymphocytes, and CNS tropism is not fully
controlled trial in patients with newly diagnosed glioblastoma showed understood.350 Tumor cells express pan–B-cell markers including CD19,
no improvement in survival or time to tumor progression when CD20, and CD79a, and many also express BCL6 and MUM1.351
intraoperative HSV-tk therapy and standard radiation therapy were A frequent finding is perivascular clusters of lymphocytes, and T-
compared with surgery and irradiation alone.336 Adenoviral and lymphocyte infiltrates are common in immunocompetent patients.
adeno-associated viral vector genes are also being studied. Although the Epstein-Barr virus genome has been found in HIV-
Another strategy involves drugs that inhibit autophagy, such as infected patients with PCNSL, no consistent confirmation of viral
hydroxychloroquine, which in a single-institution phase II trial resulted etiology of PCNSL has been possible among immunocompetent
in more than doubling of overall median survival when added to patients. Unique molecular features distinguish PCNSL from systemic
conventional therapy.337 Biologic response modifiers that stimulate or lymphoma. ECM-related genes are upregulated in PCNSLs, as are X-box
restore the immune system include monoclonal antibodies, interferons, binding protein 1 and STAT6, genes associated with the B-cell growth
and ILs. Inhibitors of TGF-β2, a cytokine that promotes invasion and factor IL-4.352
angiogenesis, have been shown to inhibit glioma growth in vitro.338
Finally, therapeutic targeting of other glioma cell surface receptors Tumor Biology
such as IL-13Rα2 or death receptor DR5 have shown promise in
in vitro and preclinical studies.339–341 Because of the heterogeneity PCNSL arises in the brain leptomeninges, spinal cord, or eyes and
of malignant glial tumors, treatment strategies probably will require rarely spreads outside the CNS. Its predilection for the periventricular
synergistic combinations of cytotoxic agents and noncytotoxic white matter gives rise to the characteristic neuroimaging appearance
specific molecular methods, with the hope that genetic profiling of a hyperdense mass on unenhanced CT or a hypointense appearance
eventually will help pinpoint appropriate choices for individual on long TR-weighted MRI. More than 50% of immunocompetent
patients. patients have a solitary enhancing mass lesion at presentation. After
More recently, treatments directed at immune modulation have administration of contrast material, most bulky masses of PCNSL
become a promising new line of therapy. These include new approaches enhance, most often homogeneously but occasionally in a ringlike
to immune modulation such as vaccines directed at different cell pattern, particularly in immunocompromised patients (Fig. 63.21).
surface receptors such as EGFRvIII based on chimeric antigens and Because of the hypercellularity of PCNSL, diffusion-weighted restriction
dendritic cell vaccines.342 Other work has targeted immune modulators with hypointensity on apparent diffusion coefficient maps may be
such as IL-13.343,344 Furthermore, current research is ongoing into seen.353 PCNSLs, however, are likely to be diffusely infiltrative even
immune checkpoint inhibition in many cancers, including glioblastoma, at the time of presentation. These areas of disease are not visible
using antibodies directed at the PD-1/PD-L1 axis.345,346 on neuroimaging studies because they are behind a relatively intact
blood-brain barrier. When postmortem findings are compared with find-
Adult Brainstem Gliomas ings on MRI performed shortly before death, widespread microscopic
infiltration is seen in areas that appear normal on the MRI scan.354
As discussed previously, brainstem gliomas are much less common in Therefore PCNSL can be classified as stage 1E disease and should
adults than in children; they account for less than 2% of all adult be considered for treatment purposes to be a whole-brain disease.
brain tumors.347 In one study of 48 adults with brainstem gliomas, Until the 1980s, PCNSL was considered a rare tumor, accounting
the overall median survival period was 5.4 years, and the 3-year survival for approximately 2% of CNS malignancies in immunocompetent
rate was 66%.348 The study investigators categorized them into three patients and 1% to 2% of all lymphomas.355 The demographics of
different groups. The most frequent type (in 48% of cases) occurred the disease have changed, however, both among immunocompetent
in young adults and resembled the diffuse pontine glioma of childhood patients and among those with HIV infection and organ transplant
in terms of clinical and radiologic presentation. The overall outcome recipients. The incidence of PCNSL among immunocompetent persons
(median survival of 7.3 years) was much better than that for pediatric had increased 25-fold to 51 cases per 10 million by 2000. Among
diffuse pontine gliomas, however, perhaps because in adults many of immunocompetent patients with PCNSL, median age at diagnosis is
these tumors were low-grade gliomas (in 9 of 11 patients who underwent 55 years, and males outnumber females by 2 to 1. Among patients
biopsy, the tumor was found to have benign histologic features). The with acquired immunodeficiency syndrome (AIDS), the median age
other common tumor type (in 31%), which occurred in elderly patients, is 35 years, and 95% of HIV-infected patients with PCNSL are male.356
showed ringlike contrast enhancement and was associated with a median Among organ transplant recipients, the peak incidence of PCNSL is
survival period of only 11 months. Tumors in this category that were at 6 months after transplantation, a period much shorter than in the
biopsied were found to be high-grade gliomas. The third group (8%) precyclosporine transplantation era.
942 Part III: Specific Malignancies

A B

C D

Figure 63.21 • Central nervous system lymphoma seen on


magnetic resonance images. (A) A T1-weighted postgadolinium
axial image shows a homogeneously enhancing frontal mass compress-
ing the ventricles. (B) A corresponding fluid-attenuated inversion
recovery (FLAIR) image shows extensive peritumoral edema. The
appearance of primary central nervous system lymphoma (PCNSL)
in immunocompromised patients may differ from that in immu-
nocompetent patients. (C and E) T1-weighted postgadolinium
imaging and (D and F) FLAIR imaging of PCNSL in human
immunodeficiency virus–positive patients. Ring enhancement is
shown in (C), and a more diffuse encephalitic pattern appears in
E F (E). The differential diagnosis in immunocompromised hosts should
include toxoplasmosis.

Recent data from the SEER program show an overall decline in Clinical Diagnosis and Staging
total PCNSL incidence rates, from a peak of 102 per million in 1995
to 51 in 1998, a decrease largely attributable to decline in the disease Clinical presentation is variable, perhaps accounting for the 3-month
in men younger than 59 years. The annual rate among patients older diagnostic delay in immunocompetent patients who are eventually
than 60 years has remained unchanged since 1994.357 The decline in diagnosed with PCNSL.360 The most common presenting pattern is
the younger male population with PCNSL reflects the advent of progressive focal symptoms. Less commonly, progressive cognitive
effective antiretroviral therapy and a declining incidence of PCNSL decline without focal symptoms leads to the diagnosis. Several variant
in the HIV-infected population.358 PCNSL should be distinguished clinical presentations are possible in the immunocompetent population:
from secondary spread of systemic lymphoma. In 2% to 5% of primary ocular, meningeal (although isolated meningeal involvement
immunocompetent patients with aggressive diffuse B-cell lymphoma, is more common in secondary CNS lymphoma), or relapsing-remitting
CNS involvement is present at diagnosis or at disease progression or disease. Rare syndromes include intravascular malignant lymphomatosis
relapse.359 This chapter discusses PCNSL and its treatment in the with strokelike onset and neurolymphomatosis with both peripheral
immunocompetent population. and CNS involvement. Considerations in the differential diagnosis
Cancer of the Central Nervous System • CHAPTER 63 943

A B
Figure 63.22 • Central nervous system lymphoma: histologic features. (A) High-grade B-cell lymphoma with dense perivascular lymphocytic cuffing
(hematoxylin and eosin staining). (B) Dense staining with CD20 (a B-cell marker) in the same specimen as in (A).

for PCNSL include high-grade glial tumor, CNS metastases, neuro- CSF for free kappa (κ) and lambda (λ) light chains is a potentially
sarcoidosis, and tumefactive multiple sclerosis. Spinal cord PCNSL useful technique. In one study, 52% of patients with PCNSL had
is very rare. Ocular lymphoma may be the first manifestation of the markedly increased free κ and λ light chain ratios.366 Investigational
disease or its relapse. Patients with ocular lymphoma have a 50% to use of microRNA detection in the CSF determined by real-time
80% chance of developing cerebral lymphoma. PCR has helped to distinguish lymphoma from inflammatory CNS
Initial evaluation of patients with suspected PCNSL should include disease.367,368
a thorough physical examination to exclude possible extraneural sources When the foregoing procedures fail to confirm the diagnosis, the
of lymphoma. Neurologic evaluation is directed at clarification of the surgical procedure of choice is a stereotactic brain biopsy. Aggressive
extent of CNS disease and confirmation that disease is solely in the surgical resection does not improve survival and may cause deterioration
CNS. Although extra-CNS disease is uncommon, one study has because of the deep location of many PCNSLs. It is most important
suggested that whole-body PET-CT localized systemic lymphoma in to try to withhold corticosteroids from patients during initial evaluation
11% of patients who were thought to have PCNSL.361 The International and surgical confirmation. Corticosteroids repair the blood-brain barrier
PCNSL Collaborative Group has established guidelines for the and also have a direct cytolytic effect on B-cell lymphomas. Although
diagnostic evaluation of possible PCNSL. Clinical studies include the patient’s clinical symptoms may abate with early institution of
dilated eye examination with slit lamp evaluation, recording of corticosteroids, with frequent nearly complete resolution of MRI-
prognostic factors (age and performance status), and cognitive function enhancing abnormalities, diagnosis will be compromised, and
(MMSE). Laboratory studies include HIV serology, serum lactate corticosteroids will have to be withdrawn in order to proceed with
dehydrogenase level, CSF cytology, flow cytometry, and immuno- definitive biopsy confirmation. However, a Mayo Clinic series found
globulin heavy-chain polymerase chain reaction (PCR) with 24-hour that the majority of patients did not experience significant radiographic
urine collection for creatinine clearance in persons who will be treated change or require a second biopsy for diagnosis when corticosteroids
with high-dose methotrexate. Imaging studies include contrast-enhanced were used preoperatively, particularly if contrast enhancement persisted
cranial MRI; CT of the chest, abdomen, and pelvis; testicular ultra- after administration of corticosteroids.369 The findings of one study
sonography; and possibly bone marrow biopsy.362 suggest that initial marked diminution in MRI abnormalities in response
Some patients will be able to avoid brain biopsy for diagnosis by to glucocorticoids is a favorable prognostic parameter, with 117-month
either vitrectomy or CSF cytologic confirmation of PCNSL. Oph- survival in responders versus 5.5 months in those whose scans did
thalmologic consultation for slit lamp examination is a critical part not change after administration of glucocorticoids.370
of the workup, because up to 10% to 15% of patients with PCNSL Recent interest has focused on immunohistochemical definition
will have vitreous involvement at the time of diagnosis, and half of of morphologic markers of prognostic significance on the biopsy
these will have no visual symptoms. Lumbar puncture should be material (Fig. 63.22). Braaten and colleagues371 studied the expression
performed if it is not contraindicated because of the intracranial mass of BCL-6 antigen in persons with PCNSL and found that the presence
location(s) with glucose and protein determinations, cell count, of this antigen predicted a median survival of 101 months, compared
cytomorphology, and flow cytometry. Flow cytometry has improved with 14.7 months in patients without this marker. Other groups of
detection rates from 6% to 22% in recent studies, although most investigators, however, have found that the marker correlates with a
studies have involved patients with newly diagnosed aggressive B-cell negative prognosis, and thus clinical decisions cannot be made on the
systemic lymphoma who are at risk for CNS disease.363 The advent basis of early immunohistochemistry studies.372
of molecular diagnosis of PCNSL through the demonstration of
monoclonality by amplification of the rearranged IgH genes by PCR Treatment
to the CDR-III region raises the possibility of definitive diagnosis,
even with few cells present in the CSF.364 In several cases, however, PCNSL is an aggressive disease for which median survival in immu-
the PCR analysis was negative even when conventional cytologic nocompetent patients is only 3 months without treatment. Increasing
findings were suggestive of malignancy. In another study, monoclonal age and poor performance status are important negative prognostic
patterns in the CSF were found in 77% of biopsy-proved cases, with variables. A recently proposed scoring system divides patients into
no false-positive findings among control subjects.365 Examination of three groups: (1) age younger than 50 years with KPS score above
944 Part III: Specific Malignancies

70; (2) age older than 50 years with KPS score above 70; and (3) age up-front combined modality approach in younger patients could lead
older than 50 years with KPS score below 70.373 The goal of treatment to higher cure rates.389
is to eradicate both contrast-enhancing mass lesions and microscopic Most centers currently base their therapy on methotrexate
infiltration of brain, spine, leptomeninges, and vitreous. Treatment regimens. Long-term follow-up of patients who achieved durable
must be designed to maximize efficacy but also to minimize toxicity remissions for more than 1 year suggests that chemotherapy alone
to the brain. Optimal treatment has not been established. The reason is associated with a low risk of neurotoxicity even in elderly persons,
for the lack of standardized treatment protocols is the relatively small although MRI may show significant areas of clinically asymptomatic
number of patients and the absence of a phase III randomized trial.374 leukoencephalopathy.390–394 The neuropsychologic outcome after
In the past 15 years, however, steady increases in median survival have chemotherapy alone for PCNSL is quite good, whereas increasing
been achieved in patients with PCNSL. Five-year survival rates on numbers of studies document progressive dementia, sometimes with
the order of 40% are now being reported, with median survival periods gadolinium-enhancing lesions on MRI, as early as several months after
of 3 to 4 years. combined-modality treatment.381,395,396 Autopsy studies have confirmed
Radiation treatment alone led to a median survival of 18 months in not only widespread “pan-brain” infiltrative lymphoma but also extensive
early clinical trials, with 5-year survival rates of only 3% to 4%.375,376 vascular changes and leukoencephalopathy in patients who received
Chemotherapy was first used as an adjunct to radiation therapy. Twelve radiation with or without chemotherapy.397
studies including more than 450 patients have established the efficacy The following recommendations apply to immunocompetent
of multiple chemotherapeutic regimens. Initially, most were used patients only. The goal of treatment is to achieve a complete response
along with or after a course of WBRT. Extension of median survival while avoiding irradiation of normal brain. Patients with AIDS-related
periods for up to 4 years with combined regimens was achieved in PCNSL require individualized treatment based on their immune status
the mid-1990s. DeAngelis and colleagues reported a 58% complete and presence of concurrent infections.398 Transplant recipients similarly
response rate, with an additional 36% rate of partial responses and require an initial attempt at reducing immunosuppression, and their
progression-free median survival for 24 months.377 A worse prognosis treatment must also be tailored to the extent of organ dysfunction
was associated with age older than 60 years (survival for 50.4 months due to the primary disease process and concurrent therapy.
versus 21.8 months). With improved survival, however, severe delayed No firmly established dose for methotrexate has been confirmed;
neural toxicity developed in increasing numbers of patients. At 18 doses in excess of 3.5 g/m2 have been shown to achieve satisfactory
months, 15% to 50% of patients who had received combined radia- CSF drug levels. Fig. 63.23 presents an algorithm for the diagnosis
tion therapy and chemotherapy were found to have extensive white and management of PCNSL at the Hospital of the University of
matter abnormalities, with consequent severe cognitive decline.378–381 Pennsylvania; the recommended approach to management of these
This complication typically begins from 4 months to several years tumors is summarized in Box 63.2. Patients with PCNSL receive
after treatment.378 Recognition of the serious sequelae of combined methotrexate, 8 g/m2 every 14 days for up to eight cycles. The calculated
radiation therapy and chemotherapy led to attempts to provide dose is diluted in 500 mL of 5% dextrose in water and given intra-
chemotherapy as the sole modality for treatment of newly diagnosed venously over a 4-hour period. Patients achieving a complete response
PCNSL. Initial response rates to chemotherapy vary from 50% to receive two additional doses of methotrexate at 14-day intervals,
100%, with duration of response between 12 and 44 months. Typical followed by 11 monthly doses of methotrexate, 8 g/m2, as long as a
of the successes of many clinical trials are the results of Cher and complete response is maintained. MRI scans are obtained monthly
colleagues,382 who reported complete responses in 17 of 19 patients
receiving methotrexate alone, with an event-free median survival period
of 32 months and OS period of 53 months. Since that publication,
others have confirmed both the efficacy of methotrexate-based regimens
without radiation therapy and the long-term favorable cognitive Box 63.2. MANAGEMENT OF PRIMARY
outcome.383 Therapeutic levels of methotrexate can be achieved in CENTRAL NERVOUS SYSTEM
the CSF after intravenous drug administration, making it possible to LYMPHOMA AT THE UNIVERSITY OF
achieve clearance of malignant cells from the CSF without intrathecal PENNSYLVANIA
administration.384
Attempts to improve on the results of methotrexate have included • Primary central nervous system lymphoma (PCNSL) is a non-Hodgkin
a variety of other multidrug chemotherapy regimens. Standard regimens B-cell lymphoma that arises in the brain, leptomeninges, spinal cord,
effective in the treatment of comparable systemic non-Hodgkin or eyes. It should be regarded as a whole-brain disease. It rarely
lymphomas (cyclophosphamide, hydroxydaunorubicin, Oncovin, spreads outside the nervous system.
and prednisolone [CHOP]; Cytoxan, hydroxydaunorubicin, Oncovin, • Optimal treatment of PCNSL is not yet fully established, but
and Decadron [CHOD]; or methotrexate, leucovorin, Adriamycin, experience acquired during the past decade with various
cyclophosphamide, Oncovin, prednisone, and bleomycin [MACOP-B]) combinations of radiation therapy and chemotherapy suggests that
are not effective for PCNSL. Attempts to disrupt the blood-brain initial induction treatment should be attempted with a methotrexate-
barrier with hyperosmolar intraarterial mannitol do not provide based intravenous chemotherapy regimen and that concurrent
additional survival benefit beyond that achieved with intravenous intrathecal chemotherapy is not necessary. The MATRix regimen of
methotrexate-based regimens.385 methotrexate, cytarabine, thiotepa, and rituximab has been shown to
Many neuro-oncologists have therefore concluded that high-dose result in improved remission rates compared with other combination
methotrexate should be offered to all patients as the first-line agent therapies.
for treatment of PCNSL.386 However, dissenting opinions exist that • Adequate delivery of chemotherapy to microscopically infiltrated
have been discussed in detail elsewhere.387 The role of radiation therapy areas of brain protected by the blood-brain barrier remains a
was investigated in the first large, randomized phase III trial completed significant challenge, and recurrence at multiple cerebral sites
in persons with PCNSL. Omitting WBRT decreased PFS but not remains a problem for which effective therapy remains to be
OS in persons with PCNSL.388 The study provides the evidence base developed.
for the current trend to reserve WBRT for disease recurrence. However, • Retreatment of a prior complete responder at relapse with the
a study reporting outcomes of a chemotherapy-only regimen in young foregoing intensive methotrexate regimen is feasible. Alternatives
patients found reduced PFS in comparison with historical control include whole-brain radiation therapy, cytarabine, and investigational
subjects treated with combined WBRT and chemotherapy, although regimens using temozolomide or rituximab.
OS was similar. The authors questioned whether a more aggressive
Cancer of the Central Nervous System • CHAPTER 63 945

CT/MRI findings
suspicious for PCNSL

Withhold corticosteroids
Chest x-ray, CT: chest, abdomen, pelvis, testicular ultrasound
CBC, HIV, LDH

SLE and lumbar puncture with


cytology and flow cytometry*

+ Cells in vitreous + CSF lymphoma – CSF and SLE

Vitrectomy Brain biopsy

– Lymphoma + Lymphoma + PCNSL – PCNSL

Brain biopsy

– PCNSL + PCNSL

Diagnosis— Liver function tests Diagnosis—


appropriate Creatinine clearance appropriate
therapy Spinal MRI therapy
Assess cognitive function (MMSE)
Figure 63.23 • Algorithm for diagnosis and manage-
ment of primary central nervous system lymphoma (PCNSL) Corticosteroids if necessary
at the University of Pennsylvania. CBC, Complete blood for symptom control
cell count; CSF, cerebrospinal fluid; CT, computed tomog-
raphy; HIV, human immunodeficiency virus; LDH, lactate
dehydrogenase; MMSE, Mini Mental State Examination;
MRI, magnetic resonance imaging; SLE, slit-lamp examina- Definitive treatment of PCNSL
tion. *See text for other CSF investigations.

during induction therapy and at 3-month intervals during maintenance Calcium leucovorin rescue therapy should begin 24 hours after
therapy. The methotrexate protocol involves close clinical monitoring, the start of methotrexate infusion. The dose is adjusted according to
adjustment of intravenous fluid, and calcium leucovorin rescue, in methotrexate levels. Any dose of leucovorin greater than 50 mg should
addition to frequent monitoring of urine pH, renal function, and be given intravenously. If the plasma concentration of methotrexate
methotrexate levels. at 24 hours is greater than 10−5 M, 100 mg/m2 of leucovorin is given
More recent trials have found survival benefits when including intravenously every 6 hours until rescue is achieved and continued at
other regimens in addition to methotrexate. In 2016 the MATRix 25 mg intravenously or orally every 6 hours until the methotrexate
trial demonstrated that the use of methotrexate, cytarabine, thiotepa, plasma concentration is less than 10−7 M. In most patients, the
and rituximab (MATrix) had superior outcomes in patients with methotrexate concentration clears to this level by 72 hours after the
HIV-negative PCNSL compared with treatment with methotrexate infusion.
plus cytarabine alone or with methotrexate plus cytarabine and Patients with an established PCNSL diagnosis may receive concur-
rituximab (complete remission rate of 49% with MATRix versus 23% rent corticosteroid therapy to alleviate symptoms. Concurrent use of
and 30%, respectively).399 salicylates or other nonsteroidal antiinflammatory drugs or sulfonamide
Contraindications to methotrexate therapy include allergy to medications is prohibited for at least 1 week before the initiation of
methotrexate, inability to achieve adequate hydration because of cerebral methotrexate therapy. Many patients with PCNSLs will continue to
edema, or cardiac or pulmonary problems, and concurrent immunosup- take corticosteroids for extended periods. The combination of
pressive treatment. Patients should not have received prior cranial corticosteroids and methotrexate may lead to a low CD4+ count, and
irradiation. Patients with renal dysfunction resulting in a creatinine these patients will be at risk for P. jiroveci pneumonia. Like patients
clearance of less than 50 mL/min or serum creatinine level greater with other brain tumors, patients with PCNSL should receive pro-
than 2 mg/dL should not receive methotrexate. Patients with significant phylaxis with trimethoprim-sulfamethoxazole two or three times per
ascites or pleural effusions may experience delayed methotrexate week, with discontinuation of this drug 1 week before institution of
clearance because of third space accumulation. methotrexate therapy.400 Concurrent AED therapy is acceptable, and
946 Part III: Specific Malignancies

management of nausea associated with chemotherapy does not differ


in this population from that for other patients receiving comparable Table 63.8 Meningiomas: Distribution by Anatomic
regimens. Location
The management of progressive or recurrent PCNSL is not yet FREQUENCY (%a)
well established. Age and performance status of the patient must be
taken into account. In general, greater than 25% enlargement of Tumor Location Series 1a Series 2b
previous areas of gadolinium contrast enhancement, the appearance Convexity 34 21
of new lesions, or the appearance of malignant cells in the CSF, vitreous, Parasagittal 22
or, rarely, elsewhere in the body constitutes treatment failure. Cher
and colleagues382 have reported that for patients who have completed Sphenoid ridge 17 16
their maintenance therapy, return to a more intensive methotrexate Lateral ventricle 5 —
dosage regimen may lead to a second complete response. For patients Tentorium 4 —
who have progressed through therapy, other chemotherapeutic agents Cerebellar convexity, posterior fossa 5 14
such as cytarabine given intravenously or intrathecally may be con- Parasellar 3 12
sidered. At relapse or in the case of failure to achieve a complete
response after eight cycles of methotrexate, many treating physicians Intraorbital 2 2
would consider palliative radiation therapy. The immediate palliation Cerebellopontine angle 2 —
of progressive signs and symptoms is a realistic goal but must be Olfactory groove 3 10
weighed against the possibility that survival will be extended enough Foramen magnum 1 —
to allow emergence of late cognitive neurotoxicity. At most centers, Clivus 1 —
whole-brain irradiation to 40 to 45 Gy in 20 fractions is given over
4 to 5 weeks. Spine — 8
After initial diagnosis, surgical intervention usually has little place Other 1 —
in treatment. The need for intrathecal chemotherapy, however, may a
mandate placement of an Ommaya reservoir. Development of com- Of 179 cases reported by Rohringer and associates404; this series included only
cases of intracranial meningiomas, with none of the spine.
municating hydrocephalus with or after treatment of meningeal b
Of 225 cases reported by Mirimanoff and co-workers.415
lymphoma may require shunting.
Parenchymal treatment failure is the most common pattern of
relapse. Ocular, meningeal, and late rare extra-CNS relapses (e.g.,
breast, abdominal wall, bone, or lymph nodes) have been described on epidemiology).10 Some relationship with breast cancer may exist,
and may be seen with increasing frequency as patients survive longer. because the likelihood of the development of a meningioma after
Temozolomide is under investigation as an agent for treatment of breast cancer, or the development of breast cancer after meningioma,
progressive PCNSL. Rituximab, a monoclonal antibody against the is higher than in the general population.408
B-cell–specific CD20 antigen, has been demonstrated to be effective Deletions of chromosome 22 are frequent in persons with menin-
against various non-Hodgkin lymphomas, but CSF drug levels attained giomas. The likely target of this chromosomal deletion is the NF2
with intravenous therapy are not high.401 Attempts to circumvent this gene, which is mutated in patients with NF2. One of the tumor types
problem have involved intraventricular administration through an commonly seen in these patients is meningioma.409 The NF2 gene
Ommaya reservoir, with some early reports indicating total clearing also appears to have an important role in the pathogenesis of sporadic
of tumor cells with leptomeningeal lymphoma.402 Whether rituximab meningiomas. One study found NF2 mutations in 60% of sporadic
will clear parenchymal masses is not known. Intensive chemotherapy meningiomas, all of which had lost one copy of chromosome 22.410
followed by hematopoietic stem cell rescue using a regimen of cytarabine However, 40% of meningiomas have neither allelic loss of chromosome
and etoposide has been reported to give a complete response rate of 22 nor NF2 gene mutation; therefore a second tumor suppressor gene
70%, with a median PFS period of 3 years.403 Several of the patients probably is linked to the development of meningiomas.
in the reported study, however, had intraocular lymphoma only. In case of non-NF2 meningiomas, through use of whole-genome
techniques, several other genes have been identified that cluster
meningiomas into distinct subtypes: TRAF7, KLF4, AKT1, and SMO.
MENINGIOMA TRAF7 is an E3 ubiquitin ligase, KLF is a transcription factor involved
Clinical and Pathologic Considerations in pluripotency, AKT1 is involved in the PI3 kinase pathway, and
SMO is an activator of the Hedgehog signaling pathway.411 POLR2A,
Meningiomas account for approximately 30% of all intracranial tumors an RNA polymerase, was also subsequently found to have a somatic
(see Table 63.1). The male-to-female ratio is 1 to 2, with the incidence mutation that clusters distinctly from the aforementioned genes.412
increasing with age and reaching a peak in the seventh decade.404
These tumors arise from arachnoidal cells in the meninges, not the Grading of Meningiomas
brain parenchyma; therefore they are extraaxial. They produce signs
and symptoms by compressing normal tissues. The locations of Grossly, meningiomas appear as rounded masses, well circumscribed
meningiomas in two large series are shown in Table 63.8.405,406 The in contour, with a well-defined dural base that can be easily separated
specific signs and symptoms depend on the anatomic location of the from the underlying brain tissue (Fig. 63.24). Often these tumors
meningioma. For example, meningiomas arising from the cerebral extend into the adjacent bone. Meningiomas are graded between grades
convexity can cause headache, altered mentation, and seizures, whereas I, II, and III based on histologic appearance. Histologically, several
those arising from the suprasellar region are likely to cause loss of different Grade I subtypes of benign meningiomas are recognized,
vision, bitemporal hemianopia, and optic atrophy. including syncytial, fibroblastic (fibrous), transitional (mixed), psam-
Numerous potential etiologic agents for meningiomas have been momatous, microcystic, and papillary; however, these distinctions carry
investigated, including radiation, trauma, viruses, occupational exposure, little prognostic significance.218 Of more importance prognostically
diet, and exposure to sex hormones (reviewed by Longstreth and is whether the meningioma is benign, which it is in 90% of the
colleagues)407 Only ionizing radiation, however, has been strongly cases.404 Grade II meningiomas are also known as atypical meningiomas.
implicated in the pathogenesis of these tumors—for example, after Features of atypical meningiomas are focal isolated necrosis, prominence
scalp irradiation for tinea capitis (as discussed previously in the section of nucleoli, and mitotic figures with high cell density. Grade III, or
Cancer of the Central Nervous System • CHAPTER 63 947

Figure 63.24 • Meningioma: gross specimen. A large-convexity meningioma


severely displaces the underlying tissue downward and laterally, creating a
midline shift and resulting in marked ventricular compression. (From Maher
EA, McKee AC. Neoplasms of the central nervous system. In: Skarin AT, ed.
Dana-Farber Cancer Institute Atlas of Diagnostic Oncology. 3rd ed. St Louis:
Mosby; 2003.)

malignant, meningiomas are characterized by invasion into adjacent


normal brain tissue. In one large study based on data from the US
National Cancer Database, the overall 5-year survival rates in patients Figure 63.25 • Meningioma. Axial T1-weighted postgadolinium magnetic
with benign, atypical, and malignant meningiomas were 70%, 75%, resonance image shows an extraaxial enhancing mass that compresses the
and 55%, respectively.413 More recently, analysis of the SEER database underlying brain tissue.
consisting of more than 12,000 patients demonstrated 1- and 3-year
survival estimates of 95.4% and 92.4%, respectively, in patients with
histologically confirmed nonmalignant meningioma, suggesting progress
in the treatment of these tumors.414 Another retrospective study from the University of California at
Meningiomas have a distinctive appearance on radiologic studies. San Francisco reached similar conclusions regarding the improvement
On MRI, most meningiomas are isointense with gray matter on in local control with postoperative radiation therapy in patients who
T1-weighted scans and enhance intensely with gadolinium (Fig. 63.25). have undergone a subtotal resection.417 This series included mostly
Peritumoral edema is evident in 60% of meningiomas, and associated patients with benign meningiomas. A much poorer outcome was
bony changes, either destruction or hyperostosis, are seen in 20%. observed for patients with malignant meningiomas than for those
The bony changes are better visualized on CT scans than with MRI. with benign meningiomas (5-year PFS rate of 89% versus 48%;
Within the tumor may be seen calcifications, central necrosis, or P = .001).
pseudocysts. The foregoing studies all used conventional fractionated radiation
therapy, usually with doses ranging from 50 to 60 Gy of radiation
Surgery and Conventional Radiation Therapy via conventional fractionation and delivery techniques. Most investiga-
for Meningiomas tors today would recommend approximately 54 Gy for benign
meningiomas after incomplete resection and up to 60 Gy for menin-
In a classic article describing the outcome in 225 patients with giomas that have atypical or malignant features.
meningiomas who were treated with surgery as the sole modality at
Massachusetts General Hospital (MGH), Mirimanoff and associates415 Stereotactic Radiation Techniques for Meningiomas
found PFS rates of 93%, 80%, and 68% at 5, 10, and 15 years,
respectively, for persons who underwent a total resection versus 63%, Substantial experience with the use of stereotactic techniques in the
45%, and 9% for persons who underwent a subtotal resection (P < treatment of meningiomas has been accumulated. One of the centers
.0001). In general, lower rates of progression were seen with sites with extensive experience with this technique is the University of
associated with higher resectability rates. For example, meningiomas Pittsburgh. In a long-term follow-up study of 99 patients with
in the convexity, 96% of which underwent total resection, had a meningiomas treated with Gamma Knife SRS from 1987 to 1992
5-year recurrence or progression rate of 3%, versus 34% for lesions with 9 to 25 Gy (median marginal dose of 16 Gy), the total rate of
in the sphenoid ridge, which had only a 28% rate of total resection. failure was 11% at 63 to 120 months after SRS.418 A majority of
These results suggested that surgery alone was inadequate therapy meningiomas showed a decrease in size with time by MRI. At years
for meningiomas if total resection could not be achieved. In a 4 to 6, 69% of cases showed a decrease in size on MRI scan; by years
subsequent study from MGH, Miralbell and coworkers416 reported 8 to 10, this number had increased to 88%.
on 17 patients who underwent subtotal resection of a meningioma The Mayo Clinic and the Na Homolce Hospital in Prague have
followed by radiation therapy. Their 8-year PFS rate was 88%, which also reported rates of local control ranging from 89% to 98%,
was much better than the 48% rate calculated for the cohort of respectively, in benign meningiomas with the use of Gamma Knife
patients with subtotal resections who did not receive postoperative SRS.419,420 Even for meningiomas greater than 10 cm3, authors have
radiation. reported excellent tumor control—99% and 97% at 3 and 7 years,
948 Part III: Specific Malignancies

respectively—albeit with higher complication rates when these lesions


were located in the supratentorial space.421 Follow-up remains short, Box 63.3. MANAGEMENT OF MENINGIOMAS
however, with a median duration of 31 to 60 months. Patients with • Meningiomas are extraaxial tumors that arise from dura; common
atypical or malignant features performed less well. In the Mayo Clinic locations are cerebral convexity, parasagittal falx, and sphenoid ridge.
series, the 5-year local control rates were 93%, 68%, and zero for • A very long natural history is characteristic, mandating prolonged
benign, atypical, and malignant meningiomas, respectively (P < .0001). follow-up.
Despite the goal of delivering an extremely conformal dose, SRS can • More than 90% of these tumors are benign; the remainder exhibit
cause significant complications. Rates of complications, often involving atypical histologic features or frank invasion of brain parenchyma.
cranial nerves, have been reported in the 5% to 13% range.418–420 • Primary treatment is surgical, if feasible.
With the reduction in radiation dose used to treat meningiomas, • Radiation therapy is reserved for tumors that are incompletely
however, this complication rate has decreased. resected, recur after surgery, are inaccessible to surgical resection, or
An extensive experience has been reported regarding the use of have atypical or invasive features.
SRS for treatment of cavernous sinus meningiomas. Because of their • The standard radiation dose has been approximately 54 Gy for
location, surgical resection can cause significant complications, par- benign meningiomas and up to 60 Gy for tumors with atypical or
ticularly cranial nerve morbidity and resulting extraocular muscle invasive features.
paralysis.422 A number of institutions have reported high local control • Stereotactic radiation therapy techniques have been used; however,
rates, in the 91% to 98% range, with use of marginal doses that follow-up is still short in these studies.
typically range from 12 to 18 Gy, but with very short follow-up • Anecdotal reports exist of responses to medical therapy (hydroxyurea
(median, 2 to 3 years).423–425 The current standard dose delivered with and antiestrogen and antiprogesterone agents).
SRS or Gamma Knife for treatment of these tumors is a marginal
dose of 14 Gy. Furthermore, cranial nerve V appears to be sensitive
to single high-dose fractions, because the incidence of trigeminal nerve
dysfunction has ranged from 4% to 11% in these series when higher
doses are used. to interpret because of our limited understanding of the natural history
In an effort to decrease late effects, fractionated stereotactic of meningiomas. Similar modest, and inconclusive, results have been
radiotherapy (FSRT) also has been used to treat meningiomas. At the obtained with interferon-α, tamoxifen, and mifepristone (RU-486).
University of Heidelberg, a mean dose of 56.8 Gy was given in 1.8-Gy Novel pharmacologic agents that target specific signaling molecules
daily fractions and was delivered with a relocatable head frame for and cellular pathways (e.g., PDGFR, EGFR, EGFR, and the MAPK
treatment of large base-of-skull meningiomas.426 At a median follow-up pathway) have been the focus of intense research and may represent
time of 35 months, the 5-year PFS rate was 94% for 180 patients the future of medical therapy for meningiomas. The recommended
with benign meningiomas but 78% for patients with atypical features. approach to management of meningiomas is summarized in Box 63.3.
Another instance in which the fractionated technique has been used More recently, targeted therapy against vascular endothelial growth
instead of conventional SRS is for treatment of optic nerve sheath factor receptor (VEGFR) using the tyrosine kinase inhibitor sunitinib
meningiomas, because of the potential risk of optic nerve injury with has been studied, with evidence of differential effectiveness in VEGFR2
a single large dose of radiation. One group of investigators reported tumors compared with VEGFR2-negative tumors. As of 2017, a study
using FSRT with 50 to 54 Gy in 1.8-Gy fractions to treat these of an anti-PD-1 receptor antibody, pembrolizumab, is ongoing. This
tumors.427 Of 22 optic nerves with vision before FSRT, 20 nerves targeted therapy is a form of immune modulation that relies on
(92%) demonstrated preserved vision, and 42% manifested improve- overexpression of PD-L1 in tumor cells.429
ment in visual acuity and/or visual field at follow-up.
A nonrandomized comparison of conventional (nonstereotactic)
fractionated radiotherapy versus Gamma Knife radiosurgery for PITUITARY ADENOMA
selected patients with cavernous sinus meningiomas was reported by Clinical and Pathologic Considerations
Metellus and coworkers.428 Thirty-eight patients received fractionated
radiotherapy, and 38 underwent Gamma Knife radiosurgery. Both Pituitary adenomas, which make up 10% to 15% of all intracranial
groups fared well, with PFS rates of 94.7% and 94.4%, respectively. tumors, arise from the anterior lobe of the pituitary gland (Box 63.4).
Permanent morbidity was seen in 2.6% of patients in the first group They are extremely common and at autopsy can be found in up to
and in none in the second group. These results suggest that in selected 27% of cases, and in 10% of normal adult volunteers based on MRI
patients, Gamma Knife radiosurgery can be performed with little scanning. Symptomatic pituitary adenomas are much less common.
late morbidity. These tumors can give rise to signs and symptoms through secretion of
In summary, numerous reports have described the use of stereotactic hormones or by compression of nearby structures, causing neurologic
techniques for delivering radiation to meningiomas as an alternative disturbances such as headaches, bilateral bitemporal hemianopia from
to surgery or after subtotal resection. In view of the long natural compression of the optic chiasm, and cranial nerve palsies from invasion
history of these tumors, however, much longer follow-up may be into the cavernous sinus. Adenomas also can cause hypopituitarism
needed to adequately evaluate these outcomes compared with surgical from compression of the pituitary stalk. Pituitary apoplexy, a rapid
resection. and symptomatic expansion of a pituitary adenoma in the context
of hemorrhage or infarction, is a potentially devastating presentation
Medical Therapy for Meningiomas and typically warrants urgent surgical decompression.430–432 Apoplexy
is thought to be uncommon, although it occurred in 21% of patients
In the management of meningiomas, chemotherapy is reserved for with pituitary adenomas who were being followed in one single-
the time after all surgical and radiation therapy approaches have been center series.433
exhausted. A number of phase II studies have examined the potential At one time, pituitary adenomas were classified according to their
benefit of various chemotherapeutic agents (e.g., dacarbazine, Adria- staining characteristics (basophilic, eosinophilic, and chromophobe).
mycin, cyclophosphamide, vincristine, temozolomide, and irinotecan) Today, however, they are classified according to the secretion of
in persons with benign and malignant meningiomas. Unfortunately, hormones. In one series of 684 patients with pituitary adenomas who
preliminary data have demonstrated either a modest response, stability underwent surgery, prolactinomas were the most common (43%),
of disease, or no effect at all. Not only did most of the subjects in followed by nonsecreting tumors (30%)434 (Table 63.9). TSH-secreting
these studies also receive radiation therapy, but these results are difficult tumors were rare in this series (2 of 684), as they are in others.
Cancer of the Central Nervous System • CHAPTER 63 949

Population-based studies on adenoma prevalence have provided similar clinical effects evolve over time and often are ignored early on. Because
results regarding the distribution of adenoma subtypes.435,436 Because the same pituitary stem cell can produce both GH and prolactin,
prolactin-secreting adenomas are primarily treated with dopamine many patients have adenomas that secrete both hormones. Prolactinomas
agonists, prolactinomas represent a smaller percentage of modern in women and ACTH-secreting adenomas usually are microadenomas
surgical series as compared with older surgical literature. Adenomas at diagnosis. Prolactinomas in women commonly cause amenorrhea
also are classified on the basis of size as either macroadenomas (>1 cm and galactorrhea. ACTH-secreting adenomas often cause dramatic
in diameter) or microadenomas. Nonsecreting adenomas are generally clinical signs and symptoms associated with hypercortisolism that
macroadenomas at the time of diagnosis because they usually come rapidly bring patients to medical attention. ACTH-secreting pituitary
to medical attention fairly late, when they are causing neurologic adenomas also can be seen in approximately 25% of patients who
symptoms owing to mass effect. The same is true for gonadotropin- have undergone bilateral adrenalectomies for Cushing syndrome as a
secreting adenomas, which are inefficient producers and secretors of result of loss of negative feedback control by cortisol on the hypo-
hormones. GH-secreting adenomas and prolactinomas in men often thalamus. In a majority of these patients, hyperpigmentation develops
are macroadenomas at the time of diagnosis, probably because their as a result of ACTH hypersecretion, which is termed Nelson syndrome;
in this setting the pituitary tumors usually are very large and often
are difficult to completely resect.437
MRI scans are the best diagnostic imaging test for pituitary adeno-
mas. However, interpretation can be challenging. The pituitary gland
Box 63.4. MANAGEMENT OF PITUITARY and stalk normally are isointense with brain on T1-weighted MRI
ADENOMAS scans. The gland and stalk, however, intensely enhance after contrast
administration because of the absence of a blood-brain barrier. Pituitary
• Pituitary adenomas are extremely common as an incidental finding adenomas generally are seen as hypointense foci on T1-weighted MRI
(in up to 10% of normal volunteers at magnetic resonance imaging scans and do not enhance with gadolinium.405 Other abnormalities
screening). that can appear as a hypointense, nonenhancing lesion in the pituitary
• These tumors may become symptomatic (i.e., the point at which the include pars intermedia cysts, metastases, infarctions, and epidermoid
patient comes to medical attention) because of hormone secretion, cysts and abscesses.
compression of nearby structures causing neurologic symptoms, or Many investigators have studied the genetics of pituitary adeno-
compression of the pituitary stalk, leading to hypopituitarism. mas. Patients with multiple endocrine neoplasia type 1 (MEN1) are
• These tumors are classified by size as microadenomas (≤1 cm in predisposed to the development of pituitary adenomas, in addition
diameter) or macroadenomas. They are also classified by secretion to parathyroid and pancreatic islet tumors (reviewed by Thakker406).
of hormones (most commonly prolactin, growth hormone, or Anterior pituitary tumors occur in 30% of patients with MEN1,
adrenocorticotropic hormone). most commonly prolactinomas but also nonfunctional and GH-
• Initial therapy for most prolactin-secreting adenomas is with a secreting and ACTH-secreting tumors. MEN1 mutations are not
dopamine agonist (e.g., cabergoline or bromocriptine), which usually commonly found in sporadic pituitary adenomas; however, one gene
decreases prolactin levels and shrinks the tumor. that is mutated in a substantial proportion of pituitary adenomas
• Initial therapy for most other pituitary adenomas is transsphenoidal is that encoding the α subunit of the GTP-binding protein Gs.438
surgical resection, which is safe and leads to rapid reversal of Mutation of this gene leads to constitutive activation of the cyclic
neurologic signs and symptoms. Surgery normalizes hormone levels adenosine monophosphate pathway. The mutated form of this
in most patients with microadenomas. gene is called gsp and is present in 10% to 40% of GH-secreting
• Stereotactic radiosurgery or external beam radiation therapy adenomas.438
currently is reserved for treatment of residual disease or recurrence
after surgery, for surgically inaccessible tumors, and for patients who Surgery for Pituitary Adenomas
are not eligible for surgery because of medical reasons or (in patients
with secreting pituitary adenomas) whose status is hormonally The current therapy for most pituitary adenomas, excluding prolac-
uncontrolled after surgical and medical therapy options have been tinomas, involves surgery. For nonsecreting tumors, which tend to be
exhausted. In patients with elevated hormones, normalization of large and manifest with neurologic signs and symptoms, surgery offers
levels after radiation therapy may take years. rapid decompression of the visual pathways. For hormonally active
tumors, surgery leads to a rapid drop in hormone secretion. The

Table 63.9 Pituitary Adenomas: Clinical Presentation by Endocrine Secretion


Frequencya
Type (%) Signs and Symptoms Typical Size at Diagnosis
Prolactinoma 43 Women: amenorrhea, galactorrhea Women: microadenoma
Men: impotence, hypopituitarism Men: macroadenoma
Nonsecreting gonadotropin 30 Hypopituitarism Macroadenoma
GH secreting 17 Gigantism in children Macroadenoma
Acromegaly in adults
ACTH secreting 7 Cushing syndrome Microadenoma
Nelson disease
TSH secreting <1 Hyperthyroidism Can be microadenoma; often macroadenoma owing
to delayed diagnosis

a
Among 684 cases reported by Oruckaptan et al. Pituitary adenomas: results of 684 surgically treated patients and review of the literature. Surg Neurol. 2000;53(3):211-9.
ACTH, Adrenocorticotropic hormone; GH, growth hormone; TSH, thyroid-stimulating hormone.
950 Part III: Specific Malignancies

current preferred technique is the transsphenoidal approach, which Because secretion of prolactin by the lactotroph cells in the anterior
was popularized by Harvey Cushing in the early 1900s. It then fell pituitary gland is negatively regulated by dopamine produced by the
out of favor but regained popularity in the 1960s.439 Transsphenoidal hypothalamus, dopamine receptor agonists such as bromocriptine and
resection is fairly safe, with a mortality rate less than 1%.440 The most cabergoline are used to inhibit prolactin secretion by the lactotrophs.
common complications arising from this surgery are nasal septum Cabergoline has been increasingly preferred over bromocriptine after
perforation, anterior pituitary insufficiency, postoperative diabetes large comparative trials found cabergoline to be equally if not more
insipidus (which usually is transient), and CSF leak, which may result efficacious, better tolerated, and with more convenient administration.454
in meningitis.440 Rarer complications include carotid artery injury Dopamine agonists decrease tumor size and normalize prolactin levels
and loss of vision. A recent variation of this approach has been to use in 70% to 90% of patients. Dopamine agonists also restore ovulation
an endoscope through the nostril to gain access to the pituitary trans- and menses and improve visual fields in a similar percentage of cases.
sphenoidally, thereby eliminating the need for conventional skin Tumor shrinkage and decrease in prolactin levels can take anywhere
incisions, improving intraoperative visualization, and allowing for more from days to weeks to months to occur. Unfortunately, the action of
dramatic resections of macroadenomas that are generally considered dopamine agonists is reversible; therefore when the drug is discontinued,
unresectable through a standard transsphenoidal approach.441–444 The regrowth of the tumor with an increase in the prolactin level is usually
transsphenoidal approach is used in more than 90% of pituitary seen. Therefore lifelong administration is the rule, and many patients
operations. Fibrous pituitary adenomas or those with extension into can tolerate prolonged treatment for years. Both cabergoline and
the middle cranial fossa may require a transcortical approach, as a bromocriptine have been used prophylactically during pregnancy to
transsphenoidal approach may limit adequate resection. reduce symptomatic tumor enlargement, although bromocriptine is
The success of surgery alone in curing adenomas depends on the the best studied. Of note, however, is that up to 20% of patients
size and location of the tumor, especially invasion into the cavernous experience adverse effects such as nausea, vomiting, dizziness, postural
sinus.445 In a large review of a single center series using modern hypotension, and headaches. Patients who have limiting toxicity or
endoscopic endonasal techniques, Paluzzi and colleagues describe a fail to respond to one dopamine agonist may benefit from a trial of
65% cure rate in 359 patients with nonfunctioning adenomas, 82.5% a newer dopamine agonist such as lisuride, pergolide mesylate, or
cure rate in 57 Cushing disease patients, 65.3% cure rate in 49 patients terguride.455,456
with acromegaly, and 54.7% cure rate in 53 patients with prolactinomas. A few drugs are now available for the treatment of endocrine
There was no visual worsening after surgery, and there was a 0.3% hypersecretion in pituitary tumors other than prolactinomas. For
carotid artery injury rate. The degree of cavernous sinus invasion was GH-secreting adenomas, somatostatin analogues such as octreotide
considered to be an important predictor of the ability to resect tumor and lanreotide have been shown to reduce GH levels in more than 90%
completely. Similarly, in a review of transsphenoidal resection for of patients, with almost complete suppression in half of patients.457
GH-secreting adenomas, hormonal normalization was seen in 67% Dopamine agonists such as bromocriptine or cabergoline may also
to 91% of microadenomas but in only 48% to 65% of macroadeno- be used alone or as an adjuvant to somatostatin analogue therapy.458
mas.446 ACTH-secreting adenomas, which generally are smaller than Pegvisomant is a GH receptor antagonist that can provide reduction
1 cm at diagnosis, show a normalization of hormones after resection of GH levels and symptomatic relief in a majority of patients.459
in 75% to 96% of cases in most surgical series (reviewed by Lüdecke In general, surgery remains the first-line treatment for patients
and coworkers447). TSH-secreting adenomas are rare compared with with symptomatic GH-secreting adenomas; however, these drugs
other pituitary adenomas, and when they are diagnosed, they are often provide useful options in patients for whom surgical cure is not
large. Normalization of TSH after resection of these tumors has varied possible.460,461
widely in different series, ranging from 33% to 86%.448 TSH-secreting adenomas often are very large at diagnosis, making
The results of surgery for hormone-inactive adenomas are harder complete surgical resection difficult. Because these tumors express
to document because the criteria for surgical success are less well somatostatin receptors, octreotide and lanreotide can decrease tumor
defined. Clinical improvement can be seen even without total removal size and decrease TSH secretion.457
of the tumor. Series in which CT or MRI scans were performed a
few months after surgery show a wide range of gross complete resection Radiation Options for Pituitary Adenomas
rates for these tumors, from 28% to 84%.449–451 A review of several
surgical series showed that the likelihood of normalization of visual Radiation options can be highly effective in controlling pituitary
fields after surgery ranged from 16% to 53% and that visual field adenomas. Currently, however, radiation therapy is rarely used as sole
improvement occurred in 26% to 70% of cases.452 treatment for newly diagnosed tumors. In contrast with surgery,
radiation therapy will not result in a rapid reversal of neurologic signs
Medical Therapy for Prolactinomas and symptoms or a rapid drop in hormonal secretion. Therapeutic
radiation for pituitary adenomas can be delivered either as single-session
The primary treatment for prolactinomas is medical because of the SRS or as fractionated radiation therapy. SRS is generally the preferred
availability of drugs that can suppress prolactin secretion and shrink technique because this treatment is as efficacious as fractionated therapy
these tumors. Surgical resection and radiotherapy are still used for and is more convenient for patients. Fractionated radiation therapy
the treatment of prolactinomas in the minority of patients who fail is indicated when treating a tumor that is near radiation-sensitive
to respond to drugs or who cannot tolerate them.453 normal tissue, such as the optic chiasm. Indications for radiation for
Clinical manifestations of prolactinomas differ between the sexes. a nonfunctioning pituitary adenoma include medically inoperable
In premenopausal women, oligomenorrhea or amenorrhea and galactor- patients, tumor recurrence or progression after surgery, or a surgically
rhea are extremely common. Infertility also may be the presenting inaccessible tumor. Indications for radiation for a functioning pituitary
sign, and women often have decreased libido. Because estrogens have adenoma include a hormonally uncontrolled tumor after maximal
a marked stimulatory effect on prolactin synthesis and secretion, surgical resection and optimal medical management, tumor growth
pregnancy can stimulate the growth of these tumors. In men and in a poor surgical candidate, tumor recurrence, or a surgically inac-
postmenopausal women, these tumors generally are asymptomatic cessible tumor.462
until they are large enough to compress nearby structures, causing
signs and symptoms such as visual deficits, headaches, and panhypo- Stereotactic Radiosurgery for Pituitary Adenomas
pituitarism. Chronic hyperprolactinemia leads to decreased libido and
impotence in 90% of men. Galactorrhea is uncommon in men but The largest multicenter report regarding single-session, Gamma Knife
can occur in 10% to 20% of cases. radiosurgery for nonfunctioning pituitary adenoma was published by
Cancer of the Central Nervous System • CHAPTER 63 951

Sheehan and colleagues in 2013.463 In this study, nine centers pooled Late Effects After Pituitary Irradiation
outcome data on 512 patients, and an overall tumor control rate of
93.4% at last follow-up was achieved. Ten-year actuarial control was In the series just discussed, the median doses ranged from 45 to
85%, with predictors of control being smaller adenoma volume and 50 Gy.468–471 In the University of Heidelberg study, a statistically
absence of suprasellar extension. Side effects of Gamma Knife radio- significant dose-response relationship was found in favor of a dose of
surgery included new hypopituitarism in 21% of patients and new 45 Gy or less.470 One of the worrisome potential late effects of use
optic nerve complications in 6%. of higher total doses is the possibility of radiation-induced optic
SRS for functioning adenomas has delayed benefit, usually years neuropathy. In some series, visual problems develop in a few patients
after the procedure. In addition, its efficacy for Cushing disease, after radiation, presumably as a result of optic nerve damage, but such
acromegaly, and prolactinoma is variable. Nevertheless, success can complications are uncommon, ranging in frequency from 0.7% to
be seen in many series. In one of the larger series of almost 100 2%.468,469 As discussed earlier (in the section on adverse effects after
patients with refractory Cushing disease, Sheehan and colleagues irradiation of the brain or spine), the risk of optic nerve and chiasm
describe remission of Cushing disease in 70% of patients with a median injury is dependent on both total dose and dose per fraction. At the
time to remission of 16.6 months.464 doses commonly used to treat pituitary adenomas (45–50 Gy), the
Publications and data on fractionated radiation therapy are more risk of radiation-induced optic neuropathy with standard fractionation
difficult to assess because technology, dosing schemes, techniques, (1.8–2 Gy per day) is very low. When doses of 45 to 50 Gy were
accuracy have all changed over time. Often recommended doses for given in fractions of less than 2.0 Gy, the risk of deterioration of
fractionated radiation therapy are on the order of 45 to 50.4 Gy in vision was less than 1%.474
1.8-Gy daily fractions for nonfunctioning pituitary adenomas and Doses of 45 to 50 Gy to the pituitary gland carry a substantial risk
50.4 to 54 Gy in 1.8-Gy daily fractions for functioning pituitary of causing hypopituitarism. In patients who did not have hormonal
adenomas.462 Radiotherapy is rarely used to treat prolactinomas because deficiencies at the start, the risk of developing insufficiency of a given
medical treatment with dopamine agonists is so effective.465 Of note, hormone ranged from 10% to 30% in the series discussed earlier.468–472
it has been proposed that hormone-suppressing medications used to It is likely that in half of all patients who received radiation doses of
treat pituitary adenomas, such as somatostatin analogues and dopamine 45 to 50 Gy, deficiency of at least one pituitary hormone will develop
agonists, may exert radioprotective effects,466,467 and therefore these 5 years after radiotherapy. Because this risk is ongoing after radiation
medications are often discontinued before radiotherapy. However, therapy and can occur many years later, patients must be monitored
data to support this effect are limited. The overall 10-year control indefinitely for this complication. GH is often the most sensitive to
rate using fractionated radiation therapy is on the order of 85% to radiation damage. Otherwise, the sensitivity of the different hormonal
95%, based on a number of large retrospective series.468–472 In a study axes (luteinizing hormone, follicle-stimulating hormone, ACTH, and
from the University of Heidelberg, in 138 patients with pituitary TSH) varies with many factors including dose, fractionation, age,
adenomas who received radiation as initial therapy or after recurrence, and sex. Panhypopituitarism occurs in 5% to 10% of patients at 5
the overall local control rate was 95% with a mean follow-up time years from treatment. Surgical manipulation before radiation therapy
of 6 years.470 Likewise, in a study from the University of Florida is also associated with increased risk of hypothalamic-pituitary axis
with a median follow-up time of 9.2 years, the overall local control dysfunction.462
rate at 10 years was 93%.468 Ninety-eight patients in this series had Second malignant neoplasms are always a concern in patients who
surgery and radiotherapy as initial therapy, and their 10-year local receive radiation and who are expected to be long-term survivors. In
control rate was 95%. This rate was comparable to the 10-year local an analysis of 426 patients with pituitary adenomas treated at the
control rate of 90% for the 23 patients who received radiotherapy Royal Marsden Hospital with surgery and radiation therapy (median
alone for newly diagnosed adenomas, but better than the 80% control dose of 45 Gy), a secondary brain tumor (two astrocytomas, two
rate seen in 20 patients who received radiation for a recurrence after meningiomas, and one meningeal sarcoma) developed in five patients,
their initial surgery (P = .03). Similar findings regarding improved for an actuarial risk of 2.0% at 10 years and 2.4% at 20 years. The
local control in patients undergoing surgery and radiation “up-front” relative risk for developing a second tumor compared with the incidence
compared with those receiving radiation at recurrence were obtained in the normal population was 28.6-fold in patents 20 years after
from the Princess Margaret Hospital study, which examined 160 radiation.475
patients with hormonally inactive pituitary adenomas.469 Pituitary
adenomas can occur in the pediatric population, although they are
much less common than in adults. In one study in 11 patients aged ACOUSTIC NEUROMA
19 years or younger with pituitary adenomas, treatment consisted Clinical and Pathologic Considerations
of surgery plus radiation or radiation only.473 At a median follow-
up time of 15.6 years, only two patients had failed to respond The term acoustic neuroma, although widely used, is actually a misnomer.
to treatment. The tumor is not a neuroma, nor does it arise from the acoustic
In the aforementioned studies, local control refers to lack of disease (cochlear) nerve. It is a schwannoma derived from the myelin-generating
progression, clinically and radiologically. In many patients who have Schwann cells, and its origin is the superior or inferior vestibular
hormone elevations at the outset, however, complete normalization nerve. Schwannomas can affect other cranial nerves, such as the tri-
may not be achieved after radiation therapy. In the University of geminal nerve and the lower cranial nerves in the jugular foramen
Heidelberg series, of 68 patients with hormonally active pituitary region; however, these tumors are much less common than acoustic
adenomas, 52% of patients showed some reduction in their hormonal neuromas.476,477
overproduction, but only 38% demonstrated complete normalization.470 Acoustic neuromas account for 8% to 10% of all primary intracranial
Furthermore, in patients who had a response, it often took years, and tumors, and in general affect people in the fifth decade of life. These
in some cases up to 9 years. A study from the Princess Margaret tumors characteristically grow very slowly. Early on, they are
Hospital specifically analyzed data from 145 patients who received asymptomatic, but with enlargement they lead to progressive hearing
radiation for hormonally active pituitary adenomas.471 The PFS rate loss and tinnitus.478 The hearing loss typically is in the conversational
was 96% at 10 years; however, the actuarial long-term biochemical range. As the tumor expands and compresses cranial nerve VIII, it
remission rate was only 40%. Radiation is thus highly effective in may cause vertigo and unsteadiness of gait. With growth into the CP
preventing pituitary adenomas from growing; however, it is far less angle, cranial nerves V and VII may be compressed, resulting in otalgia,
effective in normalizing hormone levels in patients with hormonally facial numbness, change in taste, and only very rarely facial palsy.
active tumors. With continued growth, brainstem compression and obstruction of
952 Part III: Specific Malignancies

the fourth ventricle may develop, causing hydrocephalus. Because results, the investigators at the University of Pittsburgh changed their
signs and symptoms can arise insidiously, a progressive hearing loss policy by decreasing the marginal dose and using MRI scans for
and gait unsteadiness may evolve over years in some patients before treatment planning purposes.490 From 1992 to 1997, 190 patients
the tumor is diagnosed. underwent Gamma Knife radiosurgery with delivery of 11 to 18 Gy
Patients with NF2 often have bilateral acoustic neuromas; this to the margin of the tumor (median dose, 13 Gy). With a median
finding is diagnostic for NF2.409 Acoustic neuromas in patients with follow-up time of 30 months, the 5-year actuarial tumor control rate
NF2 contain mutations in the NF2 gene and chromosome 22 deletions. was 97%, and 71% of patients retained useful hearing. The 5-year
Sporadic unilateral acoustic neuromas arising in patients without NF2 rates for development of facial weakness and facial numbness were
are also associated with chromosome 22 deletion and NF2 mutation.479 1% and 2.7%, respectively.490 The tumor size and the prescription
Interest in chemotherapeutic options for patients with NF2 whose dose correlate with cranial neuropathy.491
tumors cannot be controlled with surgery and/or radiotherapy is Linear accelerators can also be used to perform SRS in a single
increasing. Bevacizumab has shown promise in this population in session. Based on a series of 390 patients treated with linac-based
some patients, although its role at this point remains experimental or radiosurgery at the University of Florida, investigators determined
palliative.480 that each additional cubic centimeter of volume increased the odds
of facial weakness by 17%. The same series showed that each 250-cGy
Surgery for Acoustic Neuromas dose increase resulted in 8.14 times the risk of facial weakness.492 The
range of hearing preservation in single-institution series is 32% to
Because acoustic neuromas are benign tumors without metastatic 71%. Long-term follow-up is required for these patients, because
potential, they have been traditionally treated with surgical resection. hearing can continue to decline for many years after SRS.491
A number of different approaches can be taken, including the trans- To potentially reduce late effects, a number of institutions have
labyrinthine approach, the suboccipital (retrosigmoid) approach, and fractionated their dose. Fractionation schemes have varied widely.
the middle fossa approach.481 Some centers strongly favor one approach Both hypofractionation (25 Gy in 5-Gy fractions, 30 Gy in 3-Gy
over the others, but many surgeons make a decision on a case-by-case fractions, or 21 Gy in 7-Gy fractions)493 and conventional fractionation
basis. The translabyrinthine approach is the only procedure that with 54 to 58 Gy in 1.8- to 2-Gy fractions,494,495 50 Gy in 2-Gy
inherently sacrifices hearing in the course of the procedure, and thus fractions,496 or 36 to 44 Gy in 1.8-Gy fractions497 have been used.
it typically is not used in patients who have some residual useful Because of the variability in fractionation, dosage and administration,
hearing.482 Hearing preservation is possible but not guaranteed with and treatment volume, it is very difficult to assess outcomes measures
either the suboccipital or middle fossa approach. In one series of and even more difficult to draw meaningful conclusions across therapies.
patients who underwent a resection with the suboccipital approach Although the follow-up is very short in some of these series, the
with an attempt to preserve hearing, 18 of 46 patients (39%) who control rates have ranged from 96% to 100%, with a useful hearing
had “functional” preoperative hearing maintained this hearing after rate ranging from 72% to 85% and low rates of cranial nerve V and
surgery.483 The middle fossa approach is indicated in patients who VII dysfunction. In a series from the University of Heidelberg, the
have useful hearing and have tumors entirely contained within the actuarial useful hearing preservation was 94% at 5 years. The diagnosis
internal auditory canal. In one series using this approach, total tumor of NF2 was a significant factor in hearing preservation; persons with
removal was achieved in 98% of the cases, with hearing preservation the diagnosis had a hearing preservation rate of 64% compared with
in 59% of patients.484 Approximately 89% of patients maintained 98% for those without NF2.498
normal or near-normal facial nerve function. The Heidelberg group compared their experiences in treating 200
Catastrophic complications such as brainstem stroke, postoperative patients who had acoustic neuroma with either SRS (a median of
cerebellar hemorrhage, or death are rare after surgery for acoustic 13 Gy prescribed to the 80% isodose) or FSRT (a median of 57.6 Gy
neuromas.485 Two other serious complications that are more common in 1.8-Gy fractions).499 In general, tumors treated with SRS were
are CSF leak, which can lead to meningitis, and cranial nerve VII smaller. At a median follow-up of 75 months, no difference in local
palsy. Some surgical teams believe that CSF leak and damage to the control was found. No difference in hearing preservation occurred
facial nerve are more likely with the suboccipital approach than the between patients treated with SRS who received 13 Gy or less and
translabyrinthine approach; however, both complications have been those treated with FSRT. However, when patients treated with SRS
described with both approaches.482,486 In an effort to decrease damage who received more than 13 Gy are included, preservation of useful
to the facial nerve, many surgeons routinely perform intraoperative hearing was more likely in patients treated with FSRT.499
electromyographic monitoring.485 This group reported rates of trigeminal nerve damage of 7% for
patients treated with SRS and 3% for patients treated with FSRT.
Radiation Treatment Options for Facial nerve toxicity was observed in 17% of patients treated with
Acoustic Neuromas SRS and 2% of patients treated with FSRT. For patients treated with
SRS doses of 13 Gy or lower, the rate of facial nerve damage was 5%,
Radiation can be used to treat both recurrent and residual acoustic and no patients experienced trigeminal nerve damage.499 A group at
neuromas in addition to primary acoustic neuromas. the University of California–Los Angeles reported on their experience
The prototypical method of delivery of radiation to treat acoustic in treating 400 patients with SRS or FSRT.500 No significant difference
neuromas has been through the widespread adoption of Gamma Knife in hearing preservation was found between the two modalities for
cobalt-60 units by neurosurgeons to treat acoustic neuromas. The patients with tumor volumes less than 3.0 cm3, but FSRT resulted
University of Pittsburgh has accumulated one of the most extensive in better hearing preservation for patients with larger tumors.500
experiences with this approach. An analysis of the first 5 years of use As indicated by the foregoing findings, radiation therapy can be
of Gamma Knife radiosurgery at the University with a 12- to 20-Gy a useful alternative to surgery to control the growth of acoustic
marginal dose showed a 98% local control.487 Of patients with normal neuromas. Undoubtedly, however, debate will continue regarding the
function of cranial nerves VII and V before radiosurgery, however, merits of one treatment over the other, and the optimal radiotherapy
15% and 16%, respectively, experienced some dysfunction afterward. technique. Although the previous discussion centered on the treatment
Furthermore, of patients with useful hearing before radiosurgery, only of acoustic neuromas, some evidence indicates that SRS is effective
47% maintained the same level of hearing. Other institutions have in controlling the growth of trigeminal neuromas and alleviating
also found high local control rates, along with high rates of cranial trigeminal neuralgia, as either an alternative or an adjunct to surgery.501
nerve V and VII dysfunction, after single large stereotactic doses with One recent development for patients with an inheritable form of
either the Gamma Knife488 or a linac.489 On the basis of their initial acoustic neuroma due to NF2 is the use of bevacizumab to control
Cancer of the Central Nervous System • CHAPTER 63 953

tumor growth and to improve hearing. Plotkin and colleagues published less than this dose. In four of the six patients with microscopic residual
initial results with this technique in 2009, and ongoing studies have disease, this protocol achieved local control. For all 27 patients, the
shown favorable results.480 In 31 NF2 patients, tumor shrinkage and overall 15-year survival rate was 58%, and the relapse-free survival
hearing improvement were seen in more than 50% of vestibular rate was 42%. Sung and associates510 also found that patients who
schwannomas, and stable or improved hearing was retained in the received a higher dose (40–55 Gy) had a superior survival compared
majority of patients.502 The challenge, however, is that patients need with those who received 20 to 36 Gy. On the basis of these results,
to stay on this drug for an extended period of time, and complications it would be reasonable to use approximately 50 Gy in patients requiring
include a 58% risk of hypertension and a 62% risk of proteinuria.503 radiation therapy. A series from two Canadian institutions reported
Thus long-term outcomes remain to be defined. on the outcome of 18 patients treated with external beam radiotherapy,
50.0 to 55.8 Gy in 1.8- to 2.0-Gy daily fractions, for cerebellar or
spinal hemangioblastomas.511 Outcomes differed according to VHL
CEREBELLAR HEMANGIOBLASTOMAS status. The 5-year OS was 100% for those with VHL-associated and
Clinical and Pathologic Considerations 55% for those with non–VHL-associated hemangioblastomas. The
5-year disease-free survival was 80% for patients who had VHL
Hemangioblastomas are low-grade vascular tumors that constitute syndrome compared with 48% for patients who did not have VHL
1% to 2% of intracranial tumors. They usually occur in the cerebellar syndrome.511
hemispheres and vermis, although they can involve the pons, medulla, Radiosurgery has been used for cerebellar hemangioblastomas. In
and spinal cord. Most cases occur sporadically, but 20% occur as part a study from three institutions, 38 hemangioblastoma lesions in 22
of the familial von Hippel-Lindau (VHL) syndrome.504 Patients with patients were treated stereotactically, with single fractions ranging
this syndrome have a germline mutation in the VHL gene. Tumors from 12 to 20 Gy (median, 15.5 Gy).512 The vast majority of these
that develop in these patients have sustained a mutation in the second tumors had not undergone gross total resection. With a median
VHL allele, leading to loss of VHL protein function and resulting in follow-up time of 24.5 months, the 2-year actuarial OS rate was 88%
stabilization of the HIF-1α protein under normoxic conditions. and the 2-year PFS rate was 86%. A prospective evaluation of the
Stabilization of HIF-1α leads to constitutive high levels of expression natural history of hemangioblastomas in 44 patients with VHL after
of target genes, including VEGF (reviewed by Kaelin505). For this treatment with SRS (median dose 18.9 Gy at the tumor margin)
reason, the tumors seen in patients with VHL syndrome are highly showed that the local control rates at 2, 5, 10, and 15 years after
vascular. Other abnormalities seen in these patients include retinal treatment were 91%, 83%, 61%, and 51%, respectively.513
angiomas, renal cell carcinomas, and cysts involving many organs
such as the pancreas, kidney, lungs, and liver. Pheochromocytomas CHORDOMAS AND CHONDROSARCOMAS
may also develop in these patients, who often display erythrocytosis
as a result of increased erythropoietin production. Sporadic heman- INVOLVING THE BASE OF THE SKULL
gioblastomas, which occur in patients who do not have VHL syndrome, Clinical and Pathologic Considerations
also contain mutations in the VHL gene in at least 20% of cases.506
Cerebellar hemangioblastomas often manifest in the third decade Chordomas are rare tumors with an incidence of less than 0.1 per
of life. Presenting manifestations stem from cerebellar dysfunction, 100,000 population per year.514,515 Originating from the remnant of
increased ICP, and involvement of nearby cranial nerves. These signs embryonal notochord, such lesions can occur along the spinal axis,
and symptoms include headaches, nausea, vertigo, diplopia, tinnitus, including the base of skull (dorsum sellae) to the coccyx.516 Fifty
ataxia, and poor coordination.507 The lesion is well visualized at CT percent of chordomas arise from the sacrococcygeal region (most
scanning or MRI. Angiography, which typically is performed before commonly in the fifth or sixth decade of life), with a third arising
surgery, shows the highly vascular nature of these tumors. from the base of the skull, most commonly the clivus but occasionally
Histologically, hemangioblastomas show numerous capillary and the petrous bone; the remainder arise from the cervical region (with
sinusoidal channels lined with endothelial cells. A cystic component a younger age distribution in the 30s to 40s). Chordomas rarely
is often present; the cyst is filled with xanthochromic, proteinaceous metastasize, although there have been documented cases; rates of
fluid, with a vascular nodule in the cyst wall. In one series, 6 of 19 metastasis range from 10% to 25%, with the most common sites
patients (32%) had a solid lesion without a cystic component.508 being lung, liver, and bone.517 Grossly, they are extradural, often
Lesions of this type are more likely to originate in the brainstem. multilobulated, and pseudoencapsulated.518 They may have a consistency
that can range from extremely soft to woody or cartilaginous. Histologi-
Therapy for Cerebellar Hemangioblastomas cally, nests and cords of large vacuolated epithelioid cells can be seen
within a myxoid stroma with evidence of the physaliphorous cell.
The primary therapy for most patients with these tumors is surgical Three subtypes exist: (1) conventional, (2) chondroid, and (3) dedif-
resection, if surgery can be performed safely. If a cyst is present, it is ferentiated, with the last subtype having a poor prognosis.519,520
drained; then the solid component is dissected and removed. If the Low-grade chondrosarcomas often are included in reports and
tumor involves the brainstem, however, surgery may be extremely studies with chordomas given the low incidence of both tumors.
risky, with the potential for massive hemorrhage or extensive postopera- Chondrosarcomas arise from primary mesenchymal cells or embryonic
tive edema leading to death.508 Surgical resection historically has been rests of cartilaginous matrix. Chondrosarcomas are composed of either
associated with high local control rates. In one series, recurrence was hyaline cartilage or myxoid cartilage or a mixture of the two. The
noted in only 13 of 112 patients (12%) after surgery.507 In 6 patients, myxoid variant may be confused with chordoma. In one large series
disease recurred in the original tumor bed, after incomplete removal, of chondrosarcomas of the base of the skull, 66% arose from the
and in 10 patients, recurrence was in a different site or in the same temporo-occipital region, 28% from the clivus, and 6% from the
site after gross total resection. sphenoethmoid complex.521
For patients with unresectable or incompletely resected heman- In a report from the Mayo Clinic, diplopia was the most common
gioblastomas or for those whose tumors are medically inoperable, symptom, followed by headaches, ptosis, retro-orbital pain, and neck
radiation therapy usually is provided. In a series from the Mayo Clinic, pain. Cranial deficits were very common, especially deficits of cranial
27 patients received radiation treatment, six because of microscopic nerve VI, although deficits of cranial nerves III, IV, V, IX, X, and XII
positive margins after surgery and 20 for gross residual disease.509 In were also seen.521 In the same study, MRI was shown to be the best
patients with gross residual disease, the rate of local control was 57% modality for demonstrating the entire extent of cranial chordomas
for those who received 50 Gy or more but only 33% for those receiving and the involvement of adjacent structures. These tumors typically
954 Part III: Specific Malignancies

appear hypointense (black) on T1-weighted images and hyperintense


(white) on T2-weighted images. Other possibilities to be considered
in the differential diagnosis for a clival lesion are metastasis, myeloma,
osteochondroma, meningioma, and chondrosarcoma.
Chordomas and low-grade chondrosarcomas have a similar radiologic
appearance. Together they account for almost all primary malignant
bone tumors arising from the base of the skull. They can result in
significant morbidity from local invasion and can be fatal. Even though
they are treated similarly, low-grade chondrosarcomas have a better
prognosis than do chordomas, as discussed next.

Therapy for Chordomas and Chondrosarcomas


Involving the Base of the Skull
Optimal treatment consists of maximal surgical resection (to establish
a diagnosis and to alleviate symptoms) followed by local radiation
therapy. Given the increased experience with endoscopic skull-based
techniques, improved instrumentation and, when necessary, the use
of arterially based mucosal flaps for CSF leak closure, the ability to
approach resections of these lesions has significantly improved. However,
obtaining a complete surgical resection, given the anatomic constraints
and infiltrative nature of the lesion, is very challenging. The surgery
often is performed jointly by a neurosurgeon and an otolaryngologist.
Complete resection can be curative, but this outcome is often not
possible because of the extent of disease. In the mobile spine, data have
shown that there is a 20% recurrence rate with negative margins, and Figure 63.26 • Dosimetric proton therapy treatment plan for clival
this rate rises to 70% in patients with positive margins.522 In a series chordoma. Dosimetric treatment planning for clival chordoma aims to maximize
of 60 patients (46 with chordomas and 14 with chondrosarcomas) dose to the target volume while minimizing dose to surrounding critical
from the University of Pittsburgh, the tumors were treated primarily structures as shown in color wash representing radiation dose distribution.
with surgery.523 Approximately 67% underwent total or near-total (From Walcott BP, Nahed BV, Mohyeldin A, et al. Chordoma: current concepts,
resection, and 20% were treated with postoperative radiation because management, and future directions. Lancet Oncol. 2012;13:2.)
of radiologic evidence of residual tumor. The 5-year recurrence-free
survival rates were 65% for patients with chordomas and 90% for
patients with chondrosarcomas (P = .09). Two common problems 5-year survival rates were 100% for patients with chondrosarcoma
after surgery were the development of CSF leakage and new cranial and 79% for patients with chordoma. Grade 3 and 4 late toxicities
nerve deficits, which occurred in 30% and 80% of cases, respectively. (those requiring increased medical intervention or hospital admission)
Early treatment of chordomas with radiation (about 1950 to about were observed in four patients (7%) and were symptomatic in three
1980) consisted of photon radiation therapy delivered to a total dose patients (5%). Other radiation techniques are also in use, including
of 55 Gy with use of conventional two-dimensional techniques.517 In SRS, but the data are limited. Finally, although some reports have
a 1988 report by Fuller and Bloom, the 25 patients treated had a suggested anecdotal efficacy of chordoma to alkylating agents and
median duration of freedom from local progression of 32 months, camptothecin analogues, systematic reviews of the literature have
with OS at 5 and 10 years of 44% and 17%, respectively. The unfortunately found chordomas to be generally refractory to conven-
importance of a maximal safe resection was documented in a study tional chemotherapies.529
of 21 patients with chordoma from the Mallinckrodt Institute. Although
disease-free survival was not different among the groups reviewed, GLOMUS TUMORS OF THE BASE OF
those treated with surgery alone versus biopsy and radiation had an
improvement in 10-year survival.524 The Mayo Clinic reviewed the THE SKULL
role of postoperative radiation therapy after various surgical modalities Clinical and Pathologic Considerations
and found an increase in disease-free survival and an increase in OS
dependent on the extent of surgical resection (55% resection versus Glomus tumors are low-grade tumors of neural crest origin arising
36% for biopsy only).525 from the paraganglionic (glomus body) cells and therefore are also
Given the poor outcomes with photon therapy alone in the 1960s referred to as nonchromaffin paragangliomas. Because they are thought
and 1970s, proton therapy was evaluated because of the potential to be associated with chemoreceptor tissue, they are also known as
dosimetric advantages of higher dose deposition into tumor with the chemodectomas. Glomus tissue is found along the vagus nerve, the
ability to limit radiation dose to the surrounding tissue (Fig. 63.26). glossopharyngeal nerve, and the jugular ganglion. Glomus tumors are
MGH reported on the use of fractionated proton radiation therapy often divided into those arising in the soft tissue of the neck (glomus
in 1989 with 68 patients who received postoperative radiation therapy vagale and carotid body) and those involving the temporal bone or
to a median dose of 69 cobalt Gy equivalents (CGE) with a 5-year base of the skull (glomus jugulare and glomus tympanicum). The
actuarial local control rate of 82% and disease-free survival of 76%.526 focus of this section will be the skull base glomus tumors.
A review of cases at MGH published in 1999 showed a 10-year local Glomus jugulare tumors originate from the jugular bulb in the
control rate of combined proton-photon therapy that was highest for skull base, whereas glomus tympanicum tumors arise from the middle
chondrosarcomas, intermediate for male chordomas, and lowest for ear cavity along the nerve of Jacobsen (cranial nerve X) or Arnold
female chordomas (94%, 65%, and 42%, respectively).527 A similar (cranial nerve IX). In one series, 57 of 75 patients (76%) with glomus
review at Loma Linda University Medical Center with 58 patients tumors of the head and neck region had glomus jugulare tumors,
who had skull base chordomas and chondrosarcomas and were treated whereas 11 of 75 (15%) had glomus tympanicum tumors.530 In this
with total doses between 64.8 and 79.2 CGE showed local control series, otologic signs and symptoms were common in patients with
rates of 92% for chondrosarcoma and 76% for chordomas.528 Actuarial both of these tumors. Conductive hearing loss and/or tinnitus occurred
Cancer of the Central Nervous System • CHAPTER 63 955

in the majority of these patients. More than a third had bleeding from Japan, however, they account for 3% of all intracranial tumors. Tumors
the ear, ear pain, a polyp visible in the ear canal, and/or a mass behind in this location are much more common in childhood and account
the tympanic membrane. Cranial nerve impairments were also very for 3% to 11% of intracranial tumors in this age group.535
common, specifically cranial nerve VII in patients with glomus In series from the United States and Europe, roughly a third of
tympanicum tumors and cranial nerves V, VI, VII, VIII, IX, X, XI, all pineal region tumors are germ cell tumors, a majority of which
and XII in patients with glomus jugulare tumors. are germinomas.534,536 In Japan, germ cell tumors constitute a larger
CT and MRI often are diagnostic with these tumors. Cerebral percentage of all pineal region tumors because germinomas are much
angiography will demonstrate the extremely vascular nature of these more common than in the West. Because germ cell tumors occur
tumors and in general is performed immediately before surgery. most commonly in the second decade of life, they are discussed in
the section on childhood brain tumors. Approximately a third of
Therapy for Glomus Tumors of the Base of the Skull pineal region tumors are of glial origin, mostly astrocytomas, but
some are glioblastomas, oligodendrogliomas, and ependymomas.
The treatment of glomus tumors is a very controversial topic, with These tumors are managed in a similar manner to management of
some experts strongly advocating surgery and others strongly advocating their counterparts in other parts of the brain, as discussed elsewhere in
irradiation. When surgery is performed, it generally is preceded by this chapter.
embolization to reduce the subsequent operative time and blood loss. Pineal parenchymal tumors (PPTs) account for slightly less than
Surgery for glomus tumors involving the base of the skull usually is a third of all pineal region tumors. A little less than half of PPTs are
performed jointly by a neurosurgeon and a head and neck surgeon. pineocytomas; the other half are pineoblastomas. The papillary tumor
The results with surgery in the modern era have been excellent, with of the pineal region was introduced in the WHO 2007 classification.
local control rates ranging from 83% to 95%.531 The main complications Pineocytomas are histologically benign neoplasms composed of
after surgery have been cranial nerve palsies, especially the facial nerve, well-differentiated pineal parenchymal cells. In general, these tumors
and CSF leak. affect young adults and rarely disseminate. In one study the 5-year
An extensive amount of literature regarding radiation treatment actuarial survival rate for nine patients with pineocytomas was 86%.537
for glomus tumors has been accumulated. In a series of 46 patients After resection, all of these patients were treated with local field
with glomus tumors treated with radiation doses ranging from 35 to radiotherapy, receiving a dose greater than 50 Gy without craniospinal
66 Gy at the Royal Marsden Hospital, the 10-year local control rate irradiation.
was 90%, with a median follow-up time of 9 years.532 Some late Pineoblastomas, which consist of embryonal cells indistinguishable
relapses occurred, and thus the 25-year local control rate dropped to from those that are characteristic of PNETs in other CNS sites, tend to
73%. In two patients, both of whom had received 64 Gy or more, a disseminate through the CSF and have a much poorer prognosis than
facial nerve palsy developed as a late complication. In a series from do pineocytomas. Pineoblastomas are rare in adults538 and typically
the University of Florida, 53 patients with temporal bone glomus occur in the first two decades of life. A report reviewed the outcomes of
tumors (46 with jugular tumors and 9 with tympanicum tumors), 299 patients with pineoblastoma from 109 studies in the literature.539
most of whom had received no prior therapy, underwent radiation At a mean follow-up of 31 months ± 1.9, the OS was 54%, with
therapy at doses ranging from 37.7 to 60 Gy (median dose, 45 Gy).531 patients diagnosed at 5 years of age or younger having poorer survival
The 10-year local control rate was 92%, with a median follow-up than patients diagnosed at an older age. Furthermore, multivariate
time of 15 years. On the basis of their experience, the investigators analysis revealed that patients for whom gross total resection was
recommended a total dose of 45 Gy in 1.8-Gy fractions when radiation unable to be achieved had worse survival rates than did patients for
was used. Their review of the literature showed local control rates whom gross total resection was achieved.539 The addition of adjuvant
ranging from 83% to 100% in series using radiation therapy for radiation therapy resulted in a survival benefit for patients who had a
temporal bone glomus tumors. subtotal resection, but not for patients for whom gross total resection
A recent literature review sought to compare surgery with radiation was obtained.539 Current standard treatment includes maximal safe
(either fractionated or SRS) in jugular and vagal glomus tumors.533 surgical resection followed by adjuvant cranial-spinal irradiation with
Tumor control failure, major complication rates, and the number of a boost to the postoperative bed, along with systemic chemotherapy.539
cranial nerve palsies after treatment were significantly higher in surgical A small percentage of PPTs (less than 10%)—mixed pineocytoma-
than in radiotherapy series. No differences could be noted in tumor pineoblastoma tumors or PPTs of intermediate differentiation—do
control between fractionated or stereotactic treatment, and therefore not fit either of the two categories.540 These tumors, like pineoblastomas,
consideration for SRS may be an option in certain patient populations. have the capacity to seed the CSF, and thus some experts have recom-
Although the evidence is based on a retrospective review, the authors mended craniospinal irradiation for management in such cases.540 In
emphasize the importance of individualized and multidisciplinary care one study the 5-year survival rate for patients with PPTs excluding
in the management of these tumors. Considerations include size, age pineocytomas (15 pineoblastomas, 2 mixed PPTs, and 4 PPTs with
of the patient, evidence of malignancy, genetic and family history, intermediate differentiation) was 49%.537 A multicenter retrospective
risk of multiple tumors, and morbidities of treatment (usually neu- study examining adults with PPTs found that persons with tumors
rologic), which, most important, include cranial nerve palsies and of intermediate differentiation had a better outcome than did persons
intracranial mass effect. with pineoblastomas, with 10-year survival rates of 72% versus 23%,
respectively (P = .001), and rates for control of spinal disease at 10
PINEAL REGION TUMORS years of 81% and 50%, respectively (P = .04).538

The pineal gland is located adjacent to the cerebral aqueduct and


brainstem, and therefore tumors in this location frequently obstruct TUMORS OF THE SPINAL AXIS
the posterior aspect of the third ventricle and aqueduct of Sylvius, Clinical and Pathologic Considerations
causing acute hydrocephalus with headaches, papilledema, nausea,
vomiting, diplopia, and lethargy. As tumors grow anteriorly, the Tumors of the spinal cord, which are far less common than intracranial
midbrain tegmentum and quadrigeminal plate are compressed, resulting tumors, account for only 15% of all CNS tumors. The distribution
in Parinaud syndrome: paralysis of upward gaze, diminished pupillary of histologic types of tumors involving the spinal axis is different from
response to light, and refractory or convergence nystagmus. that of tumors affecting the brain (Table 63.10).541 For example,
In the United States and Europe, tumors of the pineal region schwannomas (neuromas) account for almost one-fourth of all spinal
account for fewer than 0.5% to 1% of all intracranial tumors.534 In axis lesions but are very uncommon in the brain. Most primary spinal
956 Part III: Specific Malignancies

20% at 10 years; patients with nonclival disease did just as poorly as


Table 63.10 Primary Spinal Axis Tumors: patients with clival disease.
Distribution by Histologic Type Patients with chordomas of the sacrum or spine who are able to
undergo a radical resection have a reasonable chance for long-term
Tumor Location Frequency (%)
local control and cure. In a series from the Sahlgrenska University
Meningioma Intradural; extramedullary 42 Hospital in Sweden, 39 patients (30 with tumors involving the sacrum
Schwannoma Intradural; extramedullary 22 and 9 with tumors involving the mobile spine) underwent surgery as
Ependymomaa Intradural; intramedullary 15 the primary treatment.544 Most patients had pain at presentation, but
Astrocytoma Intradural; intramedullary 11
many also had neurologic symptoms. En bloc surgical resection was
performed in 35 cases. The final surgical margins were negative for
Other 6 tumor in 23 patients but only marginal or positive in 16 patients.
a
With a mean follow-up time of 8.1 years, 23 patients (59%) were
Ependymomas of the spinal cord are intramedullary, but myxopapillary
ependymomas of the cauda and filum terminale are intradural but extramedullary.
found to be disease free at the time of last follow-up evaluation. Local
Data from Los Angeles County, 1972–1985 (reported by Preston-Martin541). recurrence was seen in 17 patients (44%), and distant metastases
occurred in 11 patients (28%). The estimated 10- and 15-year survival
rates were 64% and 52%, respectively.

Spinal Meningiomas
axis tumors are intradural, although chordomas are extradural (see
Table 63.10). Clinically these tumors manifest in one of three ways: Meningiomas are the most common spinal axis tumor (see Table
(1) radicular pain as a result of compression or infiltration of spinal 63.10). They are associated with NF2, as are their intracranial coun-
cord roots, causing a knifelike sensation along the nerve distribution; terparts.409 The prognosis with surgery is excellent. In a large series
(2) sensorimotor deficits dependent on the level of the tumor, character- of meningiomas from MGH treated with surgery alone, 18 (8%)
ized by muscle weakness and paresthesias (pain and temperature involved the spine (see Table 63.10).415 No failures were observed at
abnormalities contralateral to the side of muscle weakness); and (3) 5 years, and a 13% recurrence rate was noted at 10 years. In a report
central syringomyelia with destruction of the central gray matter causing from the Milan Neurologic Institute, the crude recurrence rate was
motor neuron destruction, muscle wasting, and loss of pain and 6% in 150 patients with spinal meningiomas who had a complete
temperature sensation with preservation of touch. tumor resection versus 17% in the 6 patients who underwent subtotal
The imaging modality of choice for spinal tumors is MRI.542 Both resection.545 Of 80 patients who had undergone a total resection at
meningiomas and schwannomas are intradural but extramedullary. the Cleveland Clinic, only one experienced a relapse, at 8 years.546
The signal intensity of meningiomas on T1- and T2-weighted images Even the seven patients who had a subtotal resection did well, with
is similar to that of the normal cord, whereas for neuromas, the signal only two relapsing, one at 13 years and one at 16 years. These findings
is increased on T2-weighted images. Meningiomas typically enhance indicate that patients with these tumors do very well after total resection.
with gadolinium. Spinal cord ependymomas and astrocytomas generally In the event of subtotal resection or if surgery is contraindicated
are intramedullary tumors. The exception is ependymoma involving because of medical comorbidities or multifocality of lesions, radiation
the conus medullaris, which is not actually an intrinsic tumor of the therapy should be considered, as in the case of intracranial meningioma.
spinal cord. Ependymomas and astrocytomas of the spinal cord have SRS has been used in one series of 51 patients with benign spinal
a similar appearance on MRI. T1-weighted images show an enlarged lesions, including meningiomas, who were not considered surgical
spinal cord extending for several vertebral body segments. Both candidates, with a majority of patients demonstrating stability or
astrocytomas and ependymomas may have cysts that are visible on reduction in tumor size.547
MRI. T2-weighted images show increased signal intensity in the region
of the tumor, with accompanying adjacent edema. Both tumor types Spinal Schwannomas
typically enhance with gadolinium, although there are rare exceptions.
The gadolinium enhancement in ependymomas tends to be very Schwannomas (neuromas) arise from the Schwann sheath, which covers
homogenous with clear demarcation of the upper and lower extent, the extramedullary axons of the nerve roots. They are evenly distributed
unlike astrocytomas, in which the true extent often is underestimated throughout the cervical, thoracic, and lumbar regions but are uncom-
by MRI. Because they arise from cells in the central canal, ependymomas mon in the sacral region. The lesions are benign and well encapsulated;
are centrally located and expand circumferentially. In contrast, therefore total surgical resection usually is possible and is curative.
astrocytomas can originate from anywhere in the spinal cord. The
differential diagnosis of these tumors includes various nonmalignant Spinal Cord Ependymomas
conditions such as multiple sclerosis, transverse myelitis, and infarction
of the cord. Hemangioblastomas are much less common than astro- Although spinal cord ependymomas are less common than meningiomas
cytomas or ependymomas. They show enlargement of the spinal cord and schwannomas involving the spinal axis, they account for 60% of
with multiple cysts. One or more intensely enhancing nodules may all intramedullary tumors. They occur more frequently in the middle
be present in the cyst wall. adult years and are rare in children. Because of their central location
within the spinal cord, ependymomas often manifest with dysesthesias
Chordomas Involving the Spinal Axis followed by progressive motor dysfunction but without objective
evidence of sensory dysfunction.548 A variety of histologic subtypes
Chordomas involving the base of the skull were discussed earlier; have been described, including cellular (the most common), epithelial,
however, these tumors also can involve the sacrum or the mobile fibrillar, malignant, and myxopapillary. The last subtype is seen only
spine. Unlike the other histologic tumor types listed in Table 63.10, in the filum terminale or the conus medullaris and therefore is techni-
chordomas are extradural tumors. Radiation therapy has been used, cally not an intramedullary tumor.
but results have been poor. In a series from the Princess Margaret Spinal cord ependymomas have a more favorable outcome than
Hospital, 48 patients with chordoma (23 of the sacrum, 5 of the that noted for intracranial ependymomas. The primary treatment for
mobile spine, and 20 of the base of the skull) were treated with radiation these tumors is surgery. Although not usually encapsulated, benign
therapy, most immediately after the initial diagnosis but some after spinal cord ependymomas generally do not infiltrate adjacent normal
local failure after surgery.543 Survival rates were 54% at 5 years and tissue. Therefore the surgeon usually can find a plane between tumor
Cancer of the Central Nervous System • CHAPTER 63 957

and normal tissue, allowing for gross total resection, which is associated As with spinal ependymomas, it is difficult to conclusively dem-
with a very low rate of recurrence. In one series of 38 patients with onstrate that postsurgical radiation improves outcome; however, in
spinal cord ependymomas who underwent a gross total resection, general it has been given to patients who have had a subtotal resection.
none had recurred after a mean follow-up time of 24 months.548 In Most investigators have used 45 to 50 Gy to local fields for low-grade
multiple single-institution series, all with fewer than 100 patients, astrocytomas. High-grade astrocytomas have been known to recur
gross total resection is consistently associated with longer PFS.549–553 with CNS dissemination, leading some persons to recommend cra-
However, two studies with 10-year follow-up data suggest that even niospinal irradiation. However, in spite of such aggressive therapy,
after gross total resection, recurrence can occur in up to 45% of these tumors still recur.
patients.552,553 No strong data exist to support the use of chemotherapy for spinal
It is difficult to assess the role of postoperative radiation for this cord astrocytomas. However, nitrosoureas and other agents used in
tumor because of the limited number of retrospective studies with intracranial astrocytomas have been used with anecdotal reports of
small numbers of patients. In general, postoperative radiation has efficacy (reviewed by Balmaceda564).
been given to patients who underwent a subtotal resection. In a review
of 11 series using surgery followed by postoperative radiation for Miscellaneous Intramedullary Tumors
spinal ependymomas, both the 5- and 10-year survival rates ranged
from 60% to 100%, with most of the series showing 5-year survival Of the 10% of intramedullary spinal cord tumors that are not
rates in the 80% to 90% range and local relapse rates of 13% to ependymomas or astrocytomas, a mixture of uncommon tumors is
33%.554 In two of the largest series from the Princess Margaret found, including hemangioblastomas and gangliogliomas (reviewed
Hospital555 and the Royal Marsden Hospital/Atkinson Morley’s by Miller and McCutcheon565). Although exceedingly rare, primary
Hospital,556 patients with high-grade tumors had a much higher relapse intramedullary germ cell tumors and PNETs have been reported.564,566
rate than those with low-grade tumors. In both of these series, when Hemangioblastomas are benign vascular lesions associated with
failures occurred, they were usually local. VHL disease in 10% to 30% of cases, as is the case for their cerebellar
Because available data are limited, it is difficult to make strong counterpart (discussed in the section on cerebellar hemangioblastomas).
recommendations. However, patients who have benign ependymomas They typically occur in men in their fourth decade of life. Most of
that are totally resected do not appear to need postoperative radiation. these tumors appear as an enhancing tumor nodule within a cyst or
For incompletely resected benign ependymomas, it would be reasonable syrinx. Because they have well-defined margins, surgical resection
to administer 45 to 50 Gy to the tumor bed. For high-grade epen- usually provides a cure, although care must be taken to avoid excessive
dymomas, some persons would advocate giving all patients postoperative bleeding of these vascular tumors.567
radiation, regardless of the extent of surgical resection. The actual Subependymomas are benign, well-circumscribed lesions that affect
volume that should be irradiated is unclear. High-grade spinal epen- men between 30 and 60 years of age. Subependymomas typically are
dymomas have been reported to recur intracranially, which has led avascular and well demarcated from the normal cord, enhancing the
some persons to advocate craniospinal radiation,555 although little feasibility of complete surgical resection. In general, gangliogliomas
evidence exists that the addition of cranial irradiation adds any benefit.556 are benign tumors that have neuronal differentiation. They usually
If one considered craniospinal irradiation for a high-grade spinal are seen intracranially, but tumors in the spinal cord have been reported.
ependymoma, a reasonable dose would be 36 Gy followed by a boost The primary treatment for these tumors is complete surgical resection;
to the primary tumor bed to a cumulative dose of 50 to 54 Gy. however, this approach is associated with a significant risk of recurrence.
In one series of 30 spinal gangliogliomas, the 5-year actuarial survival
Spinal Cord Astrocytomas rate was 84%, but the 5-year event-free survival rate was only 36%.568

Astrocytomas are slightly less common than ependymomas in the CHILDHOOD BRAIN TUMORS
spinal cord, accounting for approximately 40% of all intramedullary
spinal tumors. Most spinal astrocytomas are low grade; in adults, only Primary CNS tumors are the most common solid tumors and the
10% to 15% are high grade. In children, high-grade astrocytomas leading cause of cancer-related morbidity and mortality in children.569
are even less common, but pilocytic astrocytomas are seen. As with CNS neoplasms constitute more than one-quarter of all malignancies
ependymomas, the initial therapy for astrocytomas of the cord is in children younger than 14 years in the United States.570 More than
surgical resection. Astrocytomas tend to be more infiltrative than 3000 new cases are reported each year. Data collected by the Central
ependymomas, however, making it difficult to find a plane of resection Brain Tumor Registry of the United States from 2007 to 2011 showed
between tumor and normal cord in order to perform a total resection. that age-adjusted incidence rate of CNS tumors was highest in infants,
Spinal astrocytomas have a poorer prognosis than do spinal who had an overall incidence rate of 6.22 per 100,000. Children aged
ependymomas. In a study from Hokkaido University of 13 patients 1 to 4 years had the second highest rate at 5.52 per 100,000, followed
with astrocytomas and 22 patients with ependymomas, the 5-year by children aged 5 to 14 years with a slightly lower age-adjusted
actuarial survival rates for the two groups were 50% and 96%, incidence of 5.00 per 100,000. During the period 1973 through
respectively (P = .007). The histologic grade of the astrocytoma has 1994, the reported incidence of primary malignant brain tumors among
a significant influence on prognosis.557 In a series from the Mayo children in the United States increased by 35%.571 This apparent
Clinic, of 43 pilocytic astrocytomas and 25 diffuse fibrillary astrocy- increase may have been due to improved detection and reporting
tomas, the 10-year survival rates were 81% and 15%, respectively.558 facilitated by the availability of high-resolution neuroimaging.572 SEER
In a series from the University of California at San Francisco, 12 data limited to infant brain tumors confirmed an increase in reported
patients with low-grade spinal astrocytomas had a relapse-free survival cases between 1975 and 1985 but no significant increase in tumor
rate of 53%, whereas three patients with high-grade tumors all died incidence between 1985 and 2005.573
within 8 months.559 The 5-year survival rate for patients with low-grade Reports of increased brain tumor incidence rates raise concern that
astrocytomas has been in the 55% to 79% range in other series.560,561 environmental factors may play a causative or contributory role in
For high-grade astrocytomas, the prognosis is significantly worse, with childhood brain tumors. Despite these concerns, epidemiologic studies
case series of grade III and IV astrocytomas having survival period of investigating maternal nutritional intake, childhood diet, childhood
1 year or less.561,562 Another series of 27 patients with anaplastic exposure to electromagnetic fields, air toxins, and parental occupational
astrocytoma and 8 patients with GBM found 1- and 5-year survival exposure have not established direct links between these factors and
rates to be 85% and 59% for anaplastic astrocytoma and 31% and the development of childhood brain tumors.574–578 As noted earlier in
0% for GBM, respectively.563 the section on epidemiology, however, strong data support a connection
958 Part III: Specific Malignancies

between cranial irradiation in childhood and the subsequent develop- Research efforts to diagnose posterior fossa tumors preoperatively have
ment of brain tumors. combined computer-based neural networks with data from advanced
Hereditary factors are estimated to be primarily responsible for neuroimaging studies (Fig. 63.27), and patient characteristics have
approximately 2% of childhood brain tumors.29 Nearly 70% of all been studied. In a series of 33 children with posterior fossa tumors,
optic pathway gliomas occur in patients with NF1,580 and almost an experienced neuroradiologist was able to correctly predict the tumor
all childhood vestibular schwannomas occur in patients with NF2. type in 73% of cases, whereas the neural networks using different
Hereditary immunosuppression disorders, as seen in Wiskott-Aldrich datasets had 95% accuracy.591
syndrome and ataxia-telangiectasia; treatment-associated immunosup- The 2016 WHO classification of medulloblastomas is based heavily
pression, as in organ transplant recipients; and exogenous immunosup- on molecular characterization. Histopathologic criteria are still used
pression, as in HIV infection, are known to be associated with an in conjunction with the molecular subtyping, which allows for the
increased risk of PCNSLs.581–583 The risk of a variety of childhood most precise prognostic information. Medulloblastomas are categorized
primary brain tumors is elevated by germline p53 mutations (Li- into four subgroups based on a several large-scale genetic profiling
Fraumeni syndrome),584 whereas PTCH gene mutations (Gorlin studies: WNT (10%), Sonic Hedgehog (30%), and group 3 (20%)
syndrome) predispose to medulloblastoma585,586 and mutations causing and group 4 (40%).592–594 WNT and Sonic Hedgehog tumors have
defects in DNA mismatch repair (Turcot syndrome) predispose one of many identified activating mutations along their respective
to medulloblastoma and malignant glioma during childhood and pathways. Groups 3 and 4 are less well defined, with MYC amplification
adolescence.587 and chromosome 17 instability as identifiable molecular features. These
two latter groups have the highest potential for metastases to the
Embryonal Brain Tumors in Childhood leptomeninges and spinal cord. The histopathology of medulloblastomas
can be one of the following: classic, desmoplastic/nodular, extensive
Embryonal tumors in childhood are defined as tumors that arise nodularity, and large cell/anaplastic. Patients with nodular tumors
from pluripotent uncommitted neuroectodermal precursors cells of have the best outcome among this group, whereas those with large
the brain and are typically highly proliferative, with WHO grade IV cell/anaplastic tumors have the worse outcome.595
designation. The older 2007 WHO classification system of embryonal Preoperative management of medulloblastoma includes the assess-
tumors included medulloblastoma, atypical teratoid rhabdoid tumor ment and treatment of increased ICP. Patients with papilledema and
(ATRT), and PNET. PNET has been eliminated from the current significant visual impairment may require emergency placement of
2016 classification system, and in its place are embryonal tumors an external ventricle drain, followed as soon as feasible by tumor
with multilayered rosettes with or without chromosome 19 (C19MC resection. Prolonged delay between ventricular drainage and tumor
region) amplification. Other embryonal tumors in this new classifi- resection significantly increases the risk of transtentorial upward hernia-
cation include medulloepithelioma, CNS neuroblastoma, and CNS tion. For patients with less severe signs and symptoms of increased
ganglioneuroblastoma. ICP, corticosteroids and acetazolamide may be used to relieve symptoms,
reduce tumor swelling, and permit further surgical planning.
Medulloblastoma Surgical treatment of medulloblastoma has three objectives. First,
sufficient tissue must be obtained to permit accurate histopathologic
The most common CNS embryonal tumor is medulloblastoma, which diagnosis. Second, tumor removal should be complete or nearly
comprises approximately 10% of all pediatric brain tumors.570 Medul- complete, because complete tumor removal favorably influences
loblastomas arise in the cerebellum or dorsal brainstem and can be prognosis. However, efforts to remove residual tumor adherent to or
seen growing into the fourth ventricle. The clinical presentation of a invading the brainstem may cause considerable postoperative neurologic
medulloblastoma is dominated by signs and symptoms of obstructive deficits, and a small residual tumor burden (e.g., <1.5 cm3) has no
hydrocephalus and increased ICP that include headache, nausea and apparent adverse effect on survival outcomes.596 Third, efforts should
vomiting, drowsiness, and ataxia. These clinical characteristics often be made to reestablish normal CSF flow. A majority of children with
are indistinguishable from those of other posterior fossa tumors, medulloblastoma will not need a permanent ventriculoperitoneal shunt.
including ependymoma and cerebellar astrocytoma. CT scans typically Furthermore, the increased use and success of endoscopic third
show evidence of hydrocephalus and relatively homogenous contrast ventriculoscopy in which the floor of the third ventricle is fenestrated
enhancement with little or no evidence of calcification. MRI scans and CSF is diverted from the third ventricle to the prepontine cistern
typically show a slightly increased T1 signal, homogenous contrast has significantly reduced the need for ventriculoperitoneal shunts and
enhancement, and diffusion restriction due to hypercellularity.589,590 their hardware-related higher infection risk.597 The incidence of tumor

A B C
Figure 63.27 • Posterior fossa tumors. Sagittal T1-weighted postgadolinium magnetic resonance images of three different posterior fossa tumors. (A) A
medulloblastoma shows homogeneous contrast enhancement without evidence of cyst formation. (B) A cerebellar pilocytic astrocytoma shows prominent cyst
or multicyst formation, with one or more contrast-enhancing mural nodules. (C) An ependymoma arising from the floor of the fourth ventricle shows a
heterogeneous contrast enhancement pattern and extends inferiorly to the upper cervical spinal cord. Note that the pilocytic astrocytoma shows intense
enhancement despite being a low-grade glioma (World Health Organization grade I).
Cancer of the Central Nervous System • CHAPTER 63 959

dissemination to the abdomen by ventriculoperitoneal shunt is extremely evaluation by spinal MRI (preoperative when feasible) to identify
low, possibly because the peritoneal environment is not conducive to metastatic tumor aggregates; (2) CSF cytologic examination (within
neuroectodermal tumor growth, or possibly because concomitant 14 days after surgery) to identify leptomeningeal tumor spread; and
systemic chemotherapy may effectively prevent tumor implantation (3) postoperative neuroimaging to assess residual tumor.606 Based on
or growth.598 the results of these studies, patients with medulloblastomas are classified
Two postoperative syndromes may complicate the clinical course into two risk-for-recurrence groups—standard risk and high risk.
of patients with medulloblastomas and other posterior fossa tumors. Standard-risk patients must have no evidence of metastatic disease,
Aseptic meningitis may occur in up to 5% of patients undergoing have 1.5 cm or less of residual tumor, be older than 3 years at diagnosis,
posterior fossa surgery. Fever and meningismus, ranging in intensity and have primary tumor located in the posterior fossa only.607 High-risk
from mild to severe, develop 5 to 10 days after surgery. Although this patients have one or more of the following conditions: evidence of
complication may occur more frequently in patients with large leptomeningeal tumor spread; more than 1.5 cm of residual tumor;
postoperative pseudomeningoceles under tension, no clinical features age younger than 3 years at diagnosis; or primary tumor location
have been found to reliably distinguish this presumed chemical outside the posterior fossa. The prognostic significance of malignant
meningitis from bacterial meningitis, and thus CSF analysis and culture CSF cytologic findings in the absence of neuroimaging abnormalities
are essential. If no infectious cause is identified, this complication (i.e., M1 stage) has been the subject of debate for more than a decade.
may be effectively treated with corticosteroids. A second syndrome However, multiple investigators have clearly demonstrated that M1
of cerebellar mutism after resection of posterior fossa tumors was first status has substantially poor prognostic implications and is sufficient
described by Hirsch and colleagues599 in 1979. More recently, these to warrant classification of the patient as high risk.608
deficits have been attributed to surgical disruption of the dentate- Radiation treatment approaches for medulloblastomas are deter-
thalamo-cortical pathway.600 This condition is far more common than mined by assignment of the patient to either a standard- or a high-risk
was originally reported and occurs in 15% to 25% of children with category. High-risk patients receive 3600 cGy of radiation to the
large midline cerebellar tumors.601,602 Complete or near-complete loss craniospinal axis and chemotherapy, followed by a boost to the posterior
of speech often is accompanied by severe lower cranial nerve, cerebellar, fossa tumor site to a total dose of approximately 5580 cGy. Standard-risk
and motor abnormalities and visual disturbances.603 Cerebellar mutism patients typically receive a lower craniospinal dose (e.g., 2340 cGy),
typically manifests 1 to 4 days after surgery and may be more frequent followed by a posterior fossa boost to 5580 cGy. Radiation doses to
in cases of aggressive surgical pursuit of medulloblastoma adherent the primary tumor site of less than 5000 cGy increase the risk of
to or invading the brainstem.602 Most patients with this syndrome treatment failure.609 The lower craniospinal radiation therapy dose for
recover functional speech during a period of several weeks to months standard risk is intended to decrease the risk of neurocognitive and
from onset, although it is common for them to have significant residual other radiation-associated toxicities. Infants and children younger than
dysfunction in speech, lower cranial nerve conduction, and motor 3 or 4 years of age may be given intensive chemotherapy alone to
coordination. In a prospective study of cerebellar mutism by the postpone or avoid the neurotoxic effects of radiation on the developing
Children’s Oncology Group, the level of initial severity of signs and brain.610 Ten-year OS in these patients has been reported to be as
symptoms directly correlated with coordination, speech and language high as 60%.611
function, and intellectual function 1 year after onset, and these deficits Treatment with craniospinal irradiation (to a dose of 36 Gy) and
persisted in a significant fraction of patients.602 local boost radiotherapy (for a total dose of 54 Gy) without adjuvant
Postresection adjuvant therapy in persons with medulloblastoma chemotherapy typically yields long-term disease control in approximately
is determined by postoperative staging for prognostic risk factor 60% of children with medulloblastoma; however, 5-year survival rates
assessment. The most important clinical prognostic factor is metastatic approaching 90% after radiation therapy alone have been reported.
stage, followed by postoperative residual tumor volume, tumor location, After whole-brain radiotherapy, however, many children will have
and the patient’s age at diagnosis.596,604,605 Approximately 20% to 30% significant long-term neurocognitive sequelae, including a demonstrable
of medulloblastoma patients have dissemination at time of diagnosis. drop in overall intelligence. This decline in intelligence is influenced
Medulloblastoma is commonly associated with seeding of the spinal by age at irradiation and the dose used. Silber and colleagues reported
cord (Fig. 63.28). Accordingly, three tumor staging studies prior to that patients who received a dose of 36 Gy to the whole brain scored
treatment are important for medulloblastoma: (1) neuraxis staging 8.2 points lower on IQ testing than did those who received 24 Gy

A B
Figure 63.28 • Disseminated medulloblastoma. (A) “Studding” of caudal nerve roots from spinal arachnoid spread of tumor. (B) Malignant cells identified
at cytologic examination of cerebrospinal fluid. (From Maher EA, McKee AC. Neoplasms of the central nervous system. In: Skarin AT, ed. Dana-Farber Cancer
Institute Atlas of Diagnostic Oncology. 3rd ed. St Louis: Mosby; 2003.)
960 Part III: Specific Malignancies

and 12.3 points lower than those who received 18 Gy.612 Older age in a study that included 42 patients with recurrent medulloblastomas
at the time of irradiation was associated with less decline in subsequent or supratentorial embryonal tumors (PNET). Median OS rates at
IQ score. 6 and 12 months were 42.5% and 17.5%.623 At the current time,
Serious long-term adverse effects of radiotherapy on the developing it is clear that very few patients with relapsed medulloblastomas
nervous system prompted efforts to reduce the dose of craniospinal have PFS in excess of 5 years. The majority of patients with recurrent
radiation therapy in nonmetastatic medulloblastoma. The lowest medulloblastomas are candidates for phase I and phase II trials.
craniospinal radiotherapy doses reported, 18 Gy in 10 fractions, with Box 63.5 summarizes the recommended approach to management of
50.4 to 55.8 Gy to the posterior fossa tumor bed, has been used in medulloblastomas.
combination with vincristine during irradiation and subsequent
vincristine, CCNU, and cisplatin.613 Ten patients aged 18 to 60 months Atypical Teratoid Rhabdoid Tumors
and without evidence of tumor dissemination received treatment
according to this approach. With a median follow-up time for living ATRTs are highly malignant embryonal tumors that mainly affect
patients of 6.3 years, the survival rate at 6 years was 70% ± 20%. children younger than 3 years. They make up about 10% of CNS
The three patients in whom relapse occurred all were found to have tumors in infancy and have a predilection for developing in the
spinal metastases, in association with brain or posterior fossa recurrence. brainstem in this age group. ATRT’s cell of origin is most likely a
These data and others provided the foundation for a Children’s pluripotent fetal cell because immunohistochemical analysis shows
Oncology Group prospective clinical trial for children with medul- that they express neural, epithelial, and mesenchymal markers. Because
loblastomas who are older than 3 years at diagnosis in which the they are highly cellular and densely packed, they appear on MRI with
craniospinal dose in two cohorts is randomized between 18 Gy (with increased diffusion restriction, similar to medulloblastomas.624 Unlike
reduction in boost volume) and standard 23.4 Gy (with posterior medulloblastomas, they are less likely to have a heterogeneous appear-
fossa radiotherapy). Preliminary results from this trial (ACNS0331) ance from hemorrhage, cysts, and calcifications. Molecular analysis
showed that decreasing radiation boost volume to the primary site of ATRT has identified a mutation in INI1 that now is a part of the
was not inferior to standard posterior fossa radiotherapy, but nonin- diagnosis of ATRT. The absence of INI1 staining is essential for the
feriority could not be established for decreased 18-Gy craniospinal identification of the ATRT. INI1 (also called SNF5 or SMARCB1)
dose.614 Prospective studies have demonstrated that chemotherapy has encodes a protein that is a part of the SI/SNF complex, which mobilizes
an important role in the treatment of high-risk medulloblastoma. nucleosomes and remodels chromatin to regulate transcription of
Medulloblastomas are responsive to a variety of chemotherapeutic target genes.625 Two comprehensive epigenetic analyses found three
agents, including cisplatin, cyclophosphamide, vincristine, CCNU, distinct patterns of methylation and gene expression patterns in tumors
and busulfan.615 Incorporation of cisplatin, CCNU, and vincristine classified as ATRT.626,627 The variable tumor response to treatment
into a postirradiation chemotherapy regimen for high-risk patients was the impetus for these investigations; however, no studies have
resulted in 5-year survival rates in excess of 80%.616 Of note, this yet been conducted to determine the treatment outcome of these
survival rate was significantly higher than survival rates for standard-risk subgroups.
patients treated with radiation therapy alone (i.e., historical control Prognosis for patients with ATRT is extremely poor, and treatment
subjects). This study and other single-institution studies have provided is intensive and multimodal. In general, 5-year OS with radiation
strong support for the use of effective adjuvant chemotherapy for all and chemotherapy approaches only 30%, with the worst prognosis
patients with medulloblastoma. Efforts to increase the efficacy of
chemotherapy for clinically high-risk patients have focused on increasing
dose and dose intensity of the active agents previously noted through
the use of serial nonmyeloablative treatment cycles, each with stem
cell support. Gajjar and colleagues reported a 5-year event-free survival Box 63.5. MANAGEMENT OF
of 83% for standard-risk patients and 70% for high-risk patients with MEDULLOBLASTOMAS AND
medulloblastoma who were treated with craniospinal radiation followed PRIMITIVE NEUROECTODERMAL
by four cycles of cyclophosphamide-based, dose-intensive chemo- TUMORS
therapy.617 For patients with supratentorial embryonal tumors, the
event-free survival rates were 75% for standard-risk and 60% for • Medulloblastoma (MB) and related primitive neuroectodermal tumor
high-risk patients with use of a similar treatment strategy.618 (PNET) constitute the most common type of malignant childhood
Effective treatment of recurrent medulloblastomas, in either cerebel- brain tumor.
lar or supratentorial sites, remains difficult. Whereas high-dose che- • Surgical objectives include gross total resection to establish
motherapy with peripheral blood stem cell rescue is a therapeutic diagnosis, restore cerebrospinal fluid (CSF) flow, and improve survival.
strategy that has produced durable responses in selected patients,619 • Postoperative staging should be undertaken in all patients and
the effectiveness of this strategy as a curative therapy remains contro- should include magnetic resonance imaging (MRI) of the brain within
versial. For children with recurrent medulloblastoma, Kalifa and 24 hours of surgery, an MRI study of the entire spine 10–14 days after
associates620 used a high-dose busulfan-thiotepa regimen. Of 28 patients surgery, and CSF cytologic studies 10–14 days after surgery. These
evaluable for tumor response, complete tumor resolution was obtained studies determine subsequent risk-based treatments.
in 36%, a partial response was obtained in 39%, and no response • Standard-risk patients must meet all of the following criteria: age
occurred in 25%. Dunkel and colleagues621 reported a series of 23 older than 3 years, cerebellar location, little or no residual tumor
patients with recurrent medulloblastomas. The chemotherapy consisted (<1.5 mL), and no evidence of metastatic tumor spread. All other
of carboplatin, thiotepa, and etoposide followed by peripheral blood patients are considered to be in the high-risk category.
stem cell rescue. Three patients died of treatment-related toxicities. • Treatment in standard-risk patients consists of lower-dose
Overall, 7 of the 23 patients (30%) remained free from tumor recurrence craniospinal radiation therapy with doses of 23.8 Gy in 1.8-Gy daily
at a median follow-up of 54 months after high-dose chemotherapy. fractions, tumor irradiation with doses of approximately 55.8 Gy, and
A review of high-dose chemotherapy for recurrent medulloblastoma less intensive chemotherapy.
reported a 22% disease-free survival rate from eight different inter- • MB or PNET in high-risk patients is treated with craniospinal
national studies.622 However, some of these included patients who irradiation with doses of approximately 36 Gy in 1.8-Gy daily
were never treated with radiation therapy, and the event-free survival fractions, tumor irradiation with doses of approximately 55.8 Gy, and
from larger international studies that reported results from the time intensive chemotherapy.
of relapse was only 4.2%. Temozolomide was shown to be effective
Cancer of the Central Nervous System • CHAPTER 63 961

in children younger than 6 months.628,629 An example of a treatment regions of high signal intensity representing tumor infiltration with
regimen for ATRT was used in a prospective study by Chi and colleagues rostral extension into the midbrain and brachium pontis and lateral
and consisted of surgery, radiation, and a five-phase chemotherapeutic extension into the cerebellar peduncles. Brainstem enlargement may
regimen of vincristine, dactinomycin, cyclophosphamide, cisplatin, be unilateral and often is asymmetric on diagnosis. The fourth ventricle
doxorubicin, and temozolomide with intrathecal chemotherapy of usually is distorted; however, obstructive hydrocephalus is distinctly
methotrexate, cytarabine, and hydrocortisone. Outcome with this uncommon at initial presentation.
treatment strategy was 53% 2-year PFS and 70% 2-year OS.630 Because of the distinct characteristic of DIPGs on MRI and the
high risk of harm from biopsy of the brainstem, a multiinstitutional
Childhood Gliomas collaboration in the early 1990s sponsored by the Children’s Cancer
Group determined that MRI was sufficiently accurate for DIPG.631
Astrocytic tumors or gliomas are a diverse group of tumors with glial More recently, the impetus for biopsy and autopsy of these tumors
cell origins, although the exact glial cell types that develop into these has stemmed from the need for tissue samples for research purposes.
tumors are still largely unknown. The new WHO classification system Biopsy is still not widely adopted because of the ethical difficulty in
separates these tumors into two main groups: diffuse, and other weighing the low benefit to the patient against the altruism of contribut-
astrocytic, which includes the low-grade gliomas. Diffuse glial tumors ing to research that can benefit others.
include diffuse astrocytoma, anaplastic astrocytoma, glioblastoma, From tissue that has been obtained during biopsies or postmortem,
oligodendroglioma, and diffuse midline gliomas. In the category of several mutations have been discovered that are highly specific to this
other astrocytic tumors are pilocytic astrocytoma, subependymal giant tumor type. More than 70% of DIPG patients have a mutation in
cell astrocytoma, and pleomorphic xanthoastrocytoma. the K27 position of the histone gene H3F3A that encodes H3.3,
which leads to a key lysine residue replacement with methionine that
Diffuse Midline Gliomas results in biochemical inhibition of PRC2. The inhibition of this
protein causes a global loss of trimethylation of lysine 27 on all histone
The majority of the diffuse gliomas are most commonly seen in adults, H3 molecules.632 In about 20% of high-grade pontine gliomas, gain-
except for the diffuse midline glioma. Tumors of this type occur in of-function mutations in ACVR1 occur in conjunction with H3.1
the brainstem, thalamus, and spinal cord. Those that grow in the mutations. ACVR1 mutation leads to a hyperactivation of the bone
pons were previously called diffuse intrinsic pontine glioma (DIPG). morphogenetic protein–ACVR1 developmental pathway.633
DIPGs constitute more than 70% of all brainstem neoplasms. The DIPGs are among the least responsive and most treatment-resistant
clinical presentation of diffuse pontomedullary brainstem gliomas childhood solid tumors. Conventional radiation therapy consists of
classically involves one or more of the following abnormalities: (1) 54 to 60 Gy, delivered with use of an involved-field irradiation protocol
cranial nerve palsies, typically VI and VII; (2) ataxia; and (3) long and administered in single daily fractions of approximately 1.8 to
tract signs, including hyperreflexia and extensor plantar responses. 2 Gy. This approach results in a median survival time of 9 to 13
The prediagnostic symptomatic interval often is less than 3 months. months from diagnosis.634 Despite encouraging single-institution reports
The MRI characteristics of DIPGs include diffuse infiltrative suggesting prolonged PFS for children with gliomas who are treated
enlargement of the pons and rostral medulla (Fig. 63.29).631 T1-weighted with hyperfractionated radiation therapy in which total radiation doses
MRI sequences usually show mass effect and reduced signal intensity reached 78 Gy, larger cooperative trials failed to demonstrate a thera-
compared with normal brain. T2-weighted MRI sequences often reveal peutic advantage for this approach, and substantially increased risk
of radiation-induced brain injury or radionecrosis was observed.635–637
Radiation implants (i.e., brachytherapy) are not appropriate for these
tumors, and the role of SRS has not been evaluated.
Standard chemotherapy has no current role in DIPGs because no
chemotherapy or biologic agent has demonstrated a significant ability
to reduce tumor volume as an objective response, let alone increase
PFS or OS. Single-agent phase I and phase II clinical trials continue
to have value in the search for active agents. A phase III trial using
CCNU, vincristine, and prednisone after radiation therapy failed to
show a survival advantage compared with use of radiation therapy
alone.638 The use of high-dose chemotherapy followed by autologous
stem cell reinfusion results in relatively brief responses, with few
instances of significant tumor reduction lasting 12 months or longer,
and no examples of curative activity.639 Accordingly, it is not possible
to support the routine use of chemotherapy in persons with brainstem
gliomas outside the setting of well-structured clinical trials.

Low-Grade Astrocytomas of Childhood


Low-grade gliomas-astrocytomas occur throughout the brain and spinal
cord. The predominant histologic subtype for cerebellar astrocytomas
is pilocytic, whereas optic pathway or hypothalamic low-grade gliomas
are either of pilocytic or fibrillary histology. Pilocytic astrocytomas
are the most common brain tumors in the pediatric population, making
up nearly a fifth of all pediatric brain tumors.570 Optic pathway or
hypothalamic gliomas may be classified by location as follows: (1)
those anterior to but not involving the chiasm; (2) chiasmal tumors
with extension posteriorly along the optic radiations; and (3) chiasmal-
Figure 63.29 • Diffuse pontine glioma. A sagittal fluid-attenuated inversion hypothalamic tumors for which the initial site of tumor growth cannot
recovery (FLAIR) magnetic resonance image shows diffuse infiltration of the be determined. The majority of optic pathway gliomas occur in children
pons by tumor. with NF1 who are younger than 6 years at diagnosis.
962 Part III: Specific Malignancies

Molecular factors influencing tumorigenesis of low-grade gliomas 90% of patients with optic pathway–hypothalamic gliomas survive
are becoming more apparent. Many childhood low-grade astrocytomas longer than 10 years,644,646 the late effects of radiation therapy, including
demonstrate a small nonrandom duplication of chromosome 7p neurocognitive problems, endocrinopathy, optic nerve injury, and
involving the oncogene BRAF that results in an upregulation of the radiation-induced second neoplasms, are important considerations.
RAS-RAF-MEK pathway. These issues have stimulated the investigation of alternative treatment
Optic nerve gliomas anterior to the chiasm manifest with progressive approaches, including chemotherapy.
visual loss, proptosis, or both. Their appearance on CT and MRI Chemotherapy has a defined role in the treatment of optic
studies usually is sufficiently diagnostic that routine biopsy is not pathway–hypothalamic gliomas. For younger children, particularly
necessary. Meningiomas of the optic nerve sheath often can be dis- those younger than 5 years, the use of chemotherapy delays or obvi-
tinguished from optic nerve gliomas on the basis of neuroimaging ates the need for radiation therapy, thereby reducing or eliminating
characteristics.640 Optic nerve gliomas should be treated conservatively. the severe neurologic morbidity associated with radiation therapy in
Documented evidence of progressive tumor growth together with young children. Currently used chemotherapy regimens for optic
painful or disfiguring proptosis and complete or nearly complete loss pathway tumors generally are well tolerated, are administered in the
of vision may justify surgical resection of the nerve. Surgical resection outpatient setting, and are not associated with a high incidence of
is curative when tumor-free margins are achieved, and no further serious late effects. In a large multiinstitutional trial of carboplatin
therapy is required. As a rule, optic nerve gliomas anterior to the and vincristine, 60% of the patients with progressive low-grade glioma
chiasm do not invade the chiasm itself. For patients with progressive had a significant reduction in tumor volume, and another 30% of
tumor growth and functional vision, radiation therapy is rarely patients had tumor stabilization.647 A different regimen developed at
indicated, and current chemotherapy is similar to that for chiasmal the University of California at San Francisco included procarbazine,
gliomas, as discussed later. 6-thioguanine, dibromodulcitol, CCNU, and vincristine. This treatment
Chiasmal and chiasmal-hypothalamic gliomas constitute 60% to protocol resulted in prolonged periods of disease stabilization with
85% of all optic pathway–hypothalamic tumors. Patients with these a median time to tumor progression of 132 weeks in children with
tumors, especially very large chiasmal-hypothalamic gliomas, often low-grade gliomas.648 The Children’s Oncology Group completed a
come to medical attention before the age of 5 years with signs and large randomized clinical trial (A9952) comparing these two strategies.
symptoms of visual loss and hydrocephalus. Older children are more Five-year PFS rates were 39% for carboplatin and vincristine and 52%
likely to have symptoms including endocrinopathies and behavioral for thioguanine, procarbazine, CCNU, and vincristine; however, the
changes. Children younger than 2 years may have a diencephalic difference between these two treatment arms did not reach statistical
syndrome characterized by frequent vomiting, anorexia, and failure significance.649 Other chemotherapeutic agents have shown some efficacy
to thrive.641 In children with NF1 and chiasmal tumors involving the in the treatment of optic pathway tumors, including the use of weekly
optic nerve, a diagnosis on the basis of radiographic criteria alone is vinblastine for recurrent or refractory low-grade astrocytomas and
sufficient. For children who do not have NF1, especially those with bevacizumab with irinotecan.650,651 BRAF V600e mutation and fusion
large globoid tumors extending through the hypothalamus, surgical variants have also paved the way for molecular targeted therapy.652
biopsy is standard.642 Although diagnostic confusion is not common, Inhibitors of key proteins in the MAPK pathway are being studied,
these tumors may have a clinical and neuroimaging appearance similar have yielded variable but some positive results, and have the potential
to that of solid craniopharyngiomas or germ cell tumors, for which to soon be considered as first-line therapy for some gliomas with
treatment is very different. BRAF mutations.653–655
Most chiasmal and chiasmal-hypothalamic gliomas are not “cured” The incidence of optic pathway tumors in patients with NF1 is
with currently available surgical approaches, radiation therapy, or markedly higher than in the general population. Up to 15% of children
chemotherapy. The slow and often erratic growth of these tumors has in whom the diagnosis of NF1 is confirmed will be found to have
led some experts to conclude that most of these “benign” tumors will optic pathway tumors when they undergo screening neuroimaging.656
eventually be fatal.642 The development of more effective chemotherapy Because optic pathway tumors nearly always arise in children younger
has proved that this conclusion is incorrect, and a majority of patients than 10 years, all young children with NF1 should undergo yearly
have long-term survival periods in excess of 10 years. In most cases, ophthalmologic evaluation and annual assessment of growth to monitor
rather than automatically initiating treatment at the time of tumor for signs of precocious puberty. Most optic nerve gliomas in patients
diagnosis, the decision to initiate treatment is based on clinical or with NF1 are anterior to the optic chiasm. In slightly more than half
radiographic evidence of tumor growth and evidence of progressive of all children with radiographically identifiable optic pathway tumors,
visual or other neurologic dysfunction during serial observations. however, signs or symptoms directly related to their tumors ultimately
Recent studies have suggested that serial assessment of the retinal developed. When studied systematically, these tumors appear to behave
nerve fiber layer thickness by optic coherence tomography is likely to in a more indolent fashion than their counterparts in children who
add to the accuracy of visual function assessments, especially in younger do not have NF1. However, low-grade gliomas in children with NF1
children.643 Patients with severe visual loss or clear historic evidence are notoriously erratic in their natural history. At times, these tumors
of rapid clinical worsening represent exceptions to this approach. appear to undergo rapid growth and then spontaneously arrest or
Whereas some patients show significant changes in visual acuity or even regress. Furthermore, progression of these tumors on MRI images
neuroimaging scans within weeks or months of initial diagnosis, many does not always correlate with vision loss. For these reasons, most
others remain clinically stable for months or years without interval clinicians treat only patients who have a decline in visual acuity.
treatment. As with their non-NF1 counterparts, management of children
For patients who retain a degree of useful vision, chiasmal and with NF1 and optic pathway glioma is dependent on the location
chiasmal-hypothalamic gliomas cannot be completely resected without of the tumor. Anterior optic pathway gliomas do not invade the
complete visual loss. Surgical debulking of large chiasmal-hypothalamic chiasm in patients with NF1 and are managed according to the clinical
gliomas, however, is increasingly recognized to provide rapid relief of symptoms. Chiasmatic optic pathway gliomas should be watched closely
symptoms caused by mass effect and hydrocephalus, delay the need for neuroimaging or clinical evidence of tumor progression. Progressive
for radiation therapy in young children, result in years of clinical chiasmal tumors are treated with chemotherapy strategies associated
stability without tumor growth, and improve the effectiveness of with a relatively low incidence of second malignancies (e.g., carboplatin
subsequent radiation therapy. and vincristine). A fundamental question remains about whether the
Local involved-field radiation therapy has been shown to be effective use of chemotherapy results in a preservation or improvement of visual
in arresting tumor growth and causing tumor shrinkage.644,645 Complete function. In a recent multiinstitutional study of visual outcomes in
tumor regression is rare after irradiation, however. Because more than patients with NF1 optic pathway glioma, Fisher reported that the
Cancer of the Central Nervous System • CHAPTER 63 963

visual acuity improved in 32% of subjects and remained stable in


40%.657 Only 28% of the 88 subjects studied had a decline in visual
acuity. Chemotherapy is preferred over other treatment modalities
because of potential functional injury to the pathway from surgery
and the unacceptably high risk of secondary tumors from radiation
in NF1 patients with a known cancer predisposition gene mutation.
Pilocytic or low-grade fibrillary tumors can also arise in other brain
regions including the cerebellum and cervicomedullary junction and
can appear as dorsally exophytic tumors and cystic nodular tumors
in the brainstem. Malignant gliomas of the cerebellum in childhood
are extremely rare. The survival rate is determined by the extent of
resection, not by histologic features. Use of radiation therapy is limited
to those cases of recurrent astrocytoma that cannot be resected because
of extensive invasion into the cerebellar peduncles or brainstem.
Cerebellar astrocytomas have, arguably, the best prognosis of any brain
tumor, with 10-year survival rates approaching 100%.658 If postoperative
MRI shows resectable tumor, reexploration may be indicated in order
to achieve complete resection. Despite excellent outcome statistics, it
is increasingly recognized that the long-term clinical consequences of
the tumor, its location, and surgical resection have been underestimated.
A significant number of children experience long-term neurologic
sequelae including cognitive deficits in up to 20% and emotional
disturbances in approximately 30%.659
When low-grade astrocytomas appear in the cervicomedullary region,
they can occupy the inferior two-thirds of the medulla and the upper
portion of the cervical spinal cord. These gliomas tend to extend
from their cervicomedullary center in conformance with anatomic
boundaries.660 In contrast with midbrain tumors, intratumoral cysts Figure 63.30 • Dorsally exophytic brainstem glioma. A sagittal T1-weighted
are uncommon. The prediagnostic symptomatic interval may extend postgadolinium magnetic resonance image shows an intensely enhancing lesion
for several years. Surgical resection of these tumors is indicated with filling the floor of the fourth ventricle, attached only at the floor at the level
of the pontomedullary junction. Enhancement is characteristic of juvenile
clinical or radiographic evidence of tumor growth. Although near-total pilocytic astrocytomas. (From Halperin EG, Constine LS, Tarbell NJ, Kun
resection is possible, the poorly defined interface between tumor and LE. Pediatric Radiation Oncology. 3rd ed. Philadelphia: Lippincott Williams
normal brainstem often precludes complete surgical removal of these & Wilkins; 1999.)
tumors. Long-term follow-up studies have indicated that many patients
will not have evidence of growth for more than 5 years. When tumor
growth is observed, its rate often is extremely slow, and malignant
transformation has not been documented. As with the dorsally exophytic Ependymoma
tumors, use of radiation therapy or chemotherapy is limited to those
few cases in which progressive symptomatic tumor growth is observed Childhood intracranial ependymomas represent approximately 5% to
and cannot be controlled by surgical approaches alone. 10% of all childhood brain tumors (see Table 63.2) and behave primarily
Pilocytic or low-grade fibrillary tumors can also appear as dorsally as localized, relatively noninvasive neoplasms that originate from the
exophytic tumors arising from the floor of the fourth ventricle. Although ventricular ependymal linings, possibly from radial glia.76 Nearly 70%
eventually the tumor often completely fills the ventricle, patients may of childhood ependymomas are located in the posterior fossa arising
have few if any neurologic signs for years before the development of from the floor of the fourth ventricle (see Fig. 63.27, C). The remaining
signs and symptoms of obstructive hydrocephalus. MRI demonstrates 30% are located in supratentorial periventricular regions. The classic
a well-demarcated lesion that is hyperintense on T2-weighted images histologic feature of ependymomas is the perivascular pseudorosette. Two
and hypointense on T1-weighted images but enhancing with gadolinium general histologic classifications have been described. Well-differentiated
(Fig. 63.30). Because these tumors are low-grade fibrillary or pilocytic ependymomas are moderately to highly vascular, with low mitotic
astrocytomas, they are amenable to surgical resection. If substantial indices and little cellular pleomorphism or evidence of necrosis. Ana-
surgical tumor removal is achieved, often no adjuvant treatment is plastic ependymomas exhibit higher mitotic rates, substantial cellular
necessary and the patient can be managed with close observation atypia, and prominent necrosis. The rare “ependymoblastoma” may
using serial MRI scans. Use of local radiation therapy or chemotherapy best be classified as an embryonal tumor with histologic evidence of
is limited to the uncommon cases of malignant dorsally exophytic ependymal differentiation and treated in a fashion identical to that
gliomas or the occurrence of significant tumor growth after surgery.661 for the medulloblastomas. The role of standard histologic classification
Pilocytic astrocytomas can also appear as cystic nodular brainstem in prognosis has been controversial, with difficulties in consensus
tumors that may be located in any region of the brainstem but most regarding the histologic features separating well-differentiated from
often are noted in the midbrain.662 These tumors have a radiographic anaplastic groups. However, agreement is emerging that histology has
appearance identical to that of their cerebellar counterparts, and their prognostic significance.664 Ependymomas originating from different
histologic features typically are those of a juvenile pilocytic astrocytoma. locations within the brain are indistinguishable histologically, but it
Surgery is appropriate treatment for those symptomatic patients with is well recognized that they are different clinically.665
unequivocal evidence of tumor growth on neuroimaging studies. When Recent molecular investigations have resulted in two classification
possible, resection of the mural nodule usually is curative. In cases in systems for ependymomas. The first is dependent of the presence of
which the major portion of the tumor is in the ventral midbrain, a RELA-fusion protein commonly seen in ependymoma. The 2016
surgical options are limited to a diagnostic biopsy, and treatment WHO classifications recognizes only this group as a distinct subtype
consists of radiation therapy or chemotherapy. Long-term survival for of ependymoma. This C11orf95-RELA fusion results from chro-
patients with these tumors often is in excess of 5 to 10 years, and a mothripsis, a shattering and reassembling of chromosomes that generates
conservative management approach often is advisable. this oncogenic product.666 The C11orf95-RELA fusion causes a
964 Part III: Specific Malignancies

constitutively active NF-κB pathway by an increase in a RELA-encoded critical structures; however, radiation-induced second malignancy rates
transcription factor p65.667 This fusion protein is seen almost exclusively 5 to 7 years after proton radiation are not yet available.
in supratentorial ependymomas. Although not a recognized subgroup The role of chemotherapy in the treatment of ependymoma is
of its own in the WHO classification system, some ependymomas controversial; in fact, no convincing role for conventional chemotherapy
also harbor a fusion of the YAP1 gene with other genes such as has been established for the treatment of these tumors.681 In single-agent
MAMLD1 and FAM118B.666,667 Recent large-scale genomic and studies, cisplatin has had an objective response rate approaching 30%.682
epigenomic studies provided support for the dividing the posterior A Children’s Oncology Group study using lomustine (CCNU),
fossa ependymomas into two subgoups: PF-EPN-A, occurring pre- procarbazine, and vincristine failed to show a significant PFS or OS
dominantly in infants and young children, and PF-EPN-B, typically advantage versus radiation therapy alone. By comparison, a single-agent
seen in adolescents and adults.668,669 Combining all of these classification study of 19 children with newly diagnosed ependymoma reported a
systems, a new system has been proposed that separates all ependymomas 74% 5-year PFS rate in children with postoperative residual tumor
into nine distinct subgroups. Among the three supratentorial subgroups treated with radiation therapy and carboplatin, vincristine, ifosfamide,
are the YAP1 and RELA-fusion anaplastic ependymomas; the third and etoposide.683 This rate was higher than published results for
is a supratentorial subependymoma with a balanced genome. The radiotherapy alone for this group. On the basis of this finding, current
three posterior fossa subgroups include PF-EPN-A and PF-EPN-B clinical trials use chemotherapy for patients whose postoperative imaging
and PF-SE (subependymoma). Three spine ependymomal subgroups studies are positive for significant residual tumor. For patients with
are based on the presence of 6q deletion, myopapillary feature with recurrent ependymoma, options for further therapy other than repeat
chromosomal instability, and NF2 mutation.666 resection are limited and generally considered to be palliative in intent.
The primary challenge in ependymoma treatment is local control, High-dose chemotherapy with autologous stem cell rescue may yield
because metastatic disease at initial diagnosis or first relapse is uncom- longer PFS than conventional chemotherapy in a minority of
mon.670,671 Surgical resection of these tumors often is difficult, and patients.684,685 Continued intensive study of ependymoma genomics
complete removal is achieved in fewer than 50% of the patients. may identify new targets for therapeutic intervention with chemotherapy
Ependymomas often are located close to brainstem structures, which and/or biologic agents.681 There is new interest in the role of reirradia-
increases the risk of morbidity when complete resection is attempted. tion for recurrent ependymoma. A study of 38 patients with childhood
Several studies confirmed the critical role of a radical surgical resection ependymoma reported a 53% 4-year event-free survival with less than
in patients with newly diagnosed ependymomas.672,673 Five-year PFS anticipated toxicity, suggesting that further exploration of this strategy
rates range from 50% to 70% after complete surgical resection and is warranted.686
from zero to 30% after incomplete resection.674,675 Better survival
is noted in children who have undergone complete resection. The Intracranial Germ Cell Tumors
frequency of gross total resections has been increased by sophisticated
technologies including ultrasonic tissue dissociators, argon lasers, and Germ cell tumors constitute 12.5% to 16% of all childhood tumors
robotic localizing devices; by experience of the surgeon with childhood in Japan but only 3% to 11% in the United States and Western
tumors; and by the intent and preoperative plan to perform a radical European countries.687 A majority are located in the pineal region and
surgical resection.676 The availability of intraoperative neuroimaging approximately a third are located in the suprasellar region. The clinical
may provide immediate confirmation of the degree of resection presentation of tumors involving the pineal region and the differential
and allow the surgeon to reoperate, immediately, if necessary.677 diagnosis for these tumors were discussed earlier in the section on
For patients with residual tumor after initial surgery, reresection tumors of the spinal axis. The clinical presentation of suprasellar germ
immediately after the postoperative MRI study reveals residual tumor cell tumors includes panhypopituitarism, diabetes insipidus, and visual
remains an option. Alternatively, deferral of second surgery until the disturbances with usually long prediagnostic symptomatic intervals,
child recovers and receives chemotherapy or radiotherapy may be often exceeding 1 year.
considered. The neuroimaging characteristics of germ cell tumors, suprasellar
Involved-field radiotherapy represents standard therapy for children or pineal, do not provide sufficient differentiation between germ cell
older than 3 years who have intracranial ependymomas. Conventional tumor histologic types to render biopsy unnecessary.687,688 The diversity
radiation therapy doses range from 54 to 56 Gy. Controversial aspects of tumor types in the suprasellar or pineal region underscores the
of radiation therapy for ependymomas include treatment volume and general importance of adequate biopsy samples for accurate diagnosis.
the necessity for craniospinal irradiation. On the basis of published Expression of tumor biomarkers in blood or CSF plays an important
reports that indicated a significant risk of CSF seeding, craniospinal role in diagnosis and therapeutic decisions and often indicates the
irradiation initially was recommended.678 In subsequent studies, neuraxis presence of a nongerminomatous germ cell tumor (NGGCT). Alpha-
relapse in ependymoma occurred in less than 5% of cases.670 Routine fetoprotein (AFP) is produced by fetal yolk sac cells and later by
staging for relapse includes postoperative MRI of the brain to character- hepatocytes. Detection of elevated levels of AFP in a patient with a
ize the extent of resection. MRI of the spine and lumbar puncture to CNS tumor indicates the presence of teratoid tumors687 (Table 63.11).
evaluate CSF cytologic features are critically important to therapeutic β-Human chorionic gonadotropin (β-hCG) indicates germ cell tumors
planning. In the absence of disseminated disease at diagnosis, the with syncytiotrophoblast activity such as choriocarcinomas. Although
pattern of relapse is local in the vast majority of cases and is not pure germinomas may express relatively low levels of β-hCG (e.g.,
influenced by the delivery of craniospinal irradiation. Therefore <50 mIU/dL), choriocarcinomas produce the highest levels of this
prophylactic craniospinal irradiation is no longer recommended. Current hormone (see Table 63.11). β-hCG expression is not a marker of
radiation therapy for ependymoma emphasizes strategies that reduce metastasis or tumor size. Placental alkaline phosphatase is another
or minimize the exposure of surrounding normal brain to radiation. marker that has been found to be elevated in patients with germ cell
Merchant and colleagues679 reported clinical outcomes for 153 patients tumors.689 Its diagnostic use lies more in immunohistochemistry.
with childhood ependymoma who were treated with conformal Placental alkaline phosphatase immunostaining is positive and diagnosti-
radiotherapy after surgery. With careful staging methods, the 7-year cally definitive for germinomas.687
PFS and OS rates were 77% and 85%, respectively. Of note, the Current practice regarding diagnostic biopsy for these tumors is
incidence of radiation-induced second malignancy was 2.3% at 7 evolving. Previously it was understood that small samples obtained
years. Similar survival results have been reported with proton radiation from stereotactic biopsy may not identify mixed tumor types, and
in patients with childhood ependymoma.680 Dosimetric comparisons thus an open surgical approach with maximum feasible resection was
between intensity-modulated radiation therapy and proton radiation preferred. However, elevations of serum or CSF AFP greater than
therapy show significant reduction of radiation exposure to surrounding twice the highest institutional normal value or elevation of serum or
Cancer of the Central Nervous System • CHAPTER 63 965

Table 63.11 Cerebrospinal Fluid and Serum Tumor


Markers for Germ Cell Tumors
Tumor AFP β-hCG
Germinoma − ±
Nongerminomatous GCT
Embryonal carcinoma ± ±
Yolk sac tumor ++ −
Choriocarcinoma − ++
Teratoma, mature − −
Teratoma, immature-malignant ± ±
Mixed germ cell tumor ± ±

AFP, α-Fetoprotein; β-hCG, β-human chorionic gonadotropin; GCT, germ cell


tumor.
Modified from Kretschmar CS. Germ cell tumors of the brain in children: a review
of current literature and new advances in therapy. Cancer Invest. 1997;15:187.

CSF β-hCG greater than 50 mIU/dL clearly indicates the presence


of NGGCT and may obviate the need for either diagnostic biopsy
or extensive surgical resection pending response to chemotherapy and
radiation therapy. By contrast, complete resection is curative for
well-differentiated teratomas, whereas chemotherapy and radiation Figure 63.31 • Craniopharyngioma. A midline sagittal T1-weighted
therapies are ineffective with this tumor. The extent of surgical resection postgadolinium magnetic resonance image shows an enhancing cystic
is less important for germinomas, which are exquisitely sensitive to suprasellar mass.
chemotherapy and radiation therapy, or for the malignant NGGCTs,
which frequently spread throughout the CSF and are less responsive
to irradiation and chemotherapy.
Craniospinal irradiation is clearly indicated in cases of documented found in the suprasellar region. These tumors may arise from embryonic
leptomeningeal metastasis from germinoma; however, its use in patients epithelial cell rests in the region of the Rathke cleft. However, an
with normal findings on CSF examination and spinal MRI is con- embryonic origin is not certain for all such tumors, particularly those
troversial. In a series from the University of Pennsylvania in which that arise later in adulthood. Two histologic subtypes are described:
39 patients with biopsy-proved germinomas all received craniospinal the adamantinomatous craniopharyngioma, which is more common
irradiation, regardless of extent of disease, no relapses occurred during during childhood, and the papillary craniopharyngioma, which is
a median follow-up time of 7.1 years, and the 10-year survival rate more common during adult years. No reported prognostic significance
was 97%.690 Of note, however, other institutions using more limited is associated with the histologic subtypes. Radiographically, their
radiation fields with or without chemotherapy for biopsy-proven appearance typically includes a cystic or multicystic component and
germinomas also have reported 5-year survival rates exceeding 90%.691,692 a solid component (Fig. 63.31). Calcifications are present in a majority
NGGCTs (choriocarcinomas, embryonal carcinomas, yolk sac tumors, of cases. Craniopharyngioma cyst fluid, similar to that of a Rathke
and malignant teratomas) have a high incidence of leptomeningeal cleft cyst, is viscous, with a high cholesterol content. Rupture of the
metastasis,693 and craniospinal radiation therapy is an important cyst contents during surgical removal is well known to produce an
component of the overall treatment plan with these tumors. Well- intense chemical meningitis.
differentiated teratomas generally are unresponsive to radiation, and The clinical presentation of craniopharyngiomas is similar to that
use of radiation therapy is limited to unresectable recurrent or progres- of other suprasellar tumors. The primary age at onset is in the first
sive teratomas in many centers. For these cases, SRS may prove to be decade of life; however, presentation before the age of 2 years is
of greater therapeutic benefit. uncommon. Approximately 25% of craniopharyngiomas are detected
Chemotherapy has an important role in the treatment of germinomas in the third decade or later. The primary signs and symptoms include
and NGGCTs. Germinomas appear to be as sensitive to chemotherapy visual dysfunction; headache; optic pallor; endocrinopathies, including
as they are to radiation.694,695 Chemotherapy has been used effectively growth failure and diabetes insipidus; and behavioral or learning
for germinomas in three settings: (1) chemotherapy without radiation dysfunction. On the basis of these findings, preoperative assessment
therapy; (2) chemotherapy followed by reduced-dose radiation therapy of patients with suspected craniopharyngioma should include a thorough
for tumors with incomplete tumor response; and (3) chemotherapy visual examination and endocrine evaluation. A study of 171 patients
after radiation therapy for tumors with incomplete tumor response. with craniopharyngioma revealed that patients diagnosed before 10
Malignant NGGCTs carry a considerably worse prognosis than that years of age had higher rates of visual and endocrine dysfunction and
for pure germinomas.692 Accordingly, in patients with NGGCTs, social dysfunction.698
intensified chemotherapy and multimodality therapeutic strategies The fundamentals of craniopharyngioma treatment emphasize local
have been used in an attempt to improve survival.696 Studies of preradia- control. Chemotherapy is ineffective for initial or recurrent tumor,
tion and postradiation chemotherapy for patients with NGGCT, but and metastatic disease is reported. Local treatment involves surgery,
no metastases, reported a 3-year PFS rate of 89%.697 radiation, or both. However, little progress has been made to determine
the best strategy—gross total resection with radiation therapy reserved
Craniopharyngioma for residual or recurrent tumor versus surgical biopsy, cyst drainage, and
radiation. Complete surgical resection is often curative and obviates the
Craniopharyngiomas constitute 6% to 10% of all childhood brain need for radiation therapy in a majority of cases.699 When postoperative
tumors and represent one of the three major tumor groups frequently MRI and intraoperative visual assessment show no evidence of tumor,
966 Part III: Specific Malignancies

the rate of recurrence is less than 20%; a majority of recurrences occur in this age group have been significantly less favorable than in older
within the first 2 years after surgery.700 Gross total resection often results children, both overall and for specific tumor types.711 These children
in transection or sacrifice of the pituitary stalk, panhypopituitarism, and also are at increased risk for substantial radiation-related neurotoxicity,
a substantial risk of behavioral and neuropsychologic dysfunction or including mental retardation, growth failure, and leukoencephalopa-
stroke that often severely affects the patient’s quality of life.701–703 These thy.712,713 Therefore primary postoperative chemotherapy approaches
risks may be lower with smaller tumors. In addition, more surgeons are used, especially in children 3 years of age or younger, to postpone
are using the transnasal transsphenoidal route for resection and cyst or replace radiation therapy. Between 1976 and 1988, 17 children
decompression to lower surgical risk of damage to the pituitary, but younger than 3 years of age with PNET/MB or ependymoma underwent
long-term outcome data are limited for this approach.704 Another treatment with a multiagent chemotherapy regimen consisting of
alternative surgical strategy involves planned incomplete resection mechlorethamine, vincristine, procarbazine, and prednisone.714
followed by radiation therapy. Radiation therapy represents standard Radiotherapy was reserved for treatment of recurrent disease. Eight
treatment for patients with residual craniopharyngioma.705–707 Fewer of 12 children with PNET/MB and two of five children with epen-
than 50% of patients with known postoperative residual tumor who dymoma survived, and the children who did not require radiation
do not receive radiation therapy will survive for 10 years. Patients therapy showed normal height and intellectual ability. Results from
who receive local field radiation, typically at doses of 50 to 56 Gy, the Children’s Cancer Group study of 299 infants with malignant
have disease-free survival rates of approximately 80% at 10 years, and brain tumors randomly assigned to receive one of two regimens of
some investigators have suggested that quality of life may be better induction chemotherapy (vincristine, cisplatin, cyclophosphamide,
for these patients than for children managed with aggressive surgical and etoposide or vincristine carboplatinum, ifosfamide, and etoposide)
resection alone.706 Proton radiation may have distinct advantages for showed no significant differences between the two induction regimens.715
craniopharyngioma patients, especially reduction of the dose to a The 5-year PFS rates for infant medulloblastoma, ATRTs, and
large portion of the brain and potential reduction of dose exposure ependymoma were 32%, 17%, and 14%, respectively. These disap-
to the mesial temporal lobe and other brain regions responsible for pointing results underscore the problems associated with treating
memory and cognition.708,709 Unfortunately, no prospective, randomized different tumor types with a single strategy.
clinical trial has been undertaken to compare surgical intent to achieve A subset of infant PNET/MB can be cured by chemotherapy
gross total resection versus biopsy, drainage, and radiation outcomes alone,715,716 and multiinstitutional clinical trials are actively searching
for recurrence and survival or adverse effects of treatment, including for prognostic factors and improved survival outcomes for these infants.
endocrine and cognitive dysfunction, stroke, second malignancies, In infant medulloblastoma, the histologic desmoplastic nodular feature
and overall quality of life. has a better survival outcome than do classic histologic features or
Chemotherapy has no established role in the treatment of newly extensive nodularity.717 Patients with anaplastic large cell histology
diagnosed craniopharyngioma. For patients whose tumor recurrence have the worst survival outcomes.718 Ongoing cooperative group
after external beam irradiation consists primarily of a cystic component, studies are testing chemotherapy dose intensification, addition of
intracystic instillation of colloidal beta-emitting radionuclides, such intrathecal chemotherapy, and earlier introduction of more limited
as phosphorus-32 or yttrium-90, may be therapeutically beneficial.710 radiation therapy, with restriction of treatment volumes and use of
conformal techniques and proton radiation to minimize exposure of
Brain Tumors in Infants normal tissue.

Approximately 20% of childhood brain tumors occur in infants and The complete reference list is available online at
children younger than 3 years. Unfortunately, the survival outcomes ExpertConsult.com.

KEY REFERENCES
1. Ostrom QT, et al. CBTRUS statistical report: 135. Gerstner ER, Frosch MP, Batchelor TT. Diffusion 285. Stupp R, et al. Radiotherapy plus concomitant and
primary brain and other central nervous system magnetic resonance imaging detects pathologically adjuvant temozolomide for glioblastoma. NEJM.
tumors diagnosed in the united states in 2009–2013. confirmed, nonenhancing tumor progression in 2005;352:987–996.
Neuro Oncol. 2016;18:v1–v75. a patient with recurrent glioblastoma receiving 286. Stupp R, et al. Effects of radiotherapy with concomi-
2. Louis DN, et al. The 2007 WHO classification bevacizumab. J Clin Oncol. 2009;28:e91–e93. tant and adjuvant temozolomide versus radiotherapy
of tumours of the central nervous system. Acta 148. Shaw E, et al. Prospective randomized trial of low- alone on survival in glioblastoma in a randomised
Neuropathol. 2007;114:97–109. versus high-dose radiation therapy in adults with phase III study: 5-year analysis of the EORTC-NCIC
85. Rossetti AO, Stupp R. Epilepsy in brain tumor supratentorial low-grade glioma: initial report of a trial. Lancet Oncol. 2009;10:459–466.
patients. Curr Opin Neurol. 2010;23:603–609. North Central Cancer Treatment Group/Radiation 288. Deleted in review.
87. Roth P, Wick W, Weller M. Steroids in neurooncol- Therapy Oncology Group/Eastern Cooperative 306. Cairncross G, et al. Phase III trial of chemo-
ogy: actions, indications, side-effects. Curr Opin Oncology Group study. J Clin Oncol. 2002;20: radiotherapy for anaplastic oligodendroglioma:
Neurol. 2010;23:597–602. 2267–2276. long-term results of RTOG 9402. J Clin Oncol.
96. Gerstner ER, et al. VEGF inhibitors in the treatment 161. Levin VA, et al. Randomized double-blind placebo- 2013;31:337–343.
of cerebral edema in patients with brain cancer. Nat controlled trial of bevacizumab therapy for radiation 315. Wick W, et al. NOA-08 randomized phase III
Rev Clin Oncol. 2009;6:229–236. necrosis of the central nervous system. Int J Radiat trial of 1-week-on/1-week-off temozolomide versus
108. Deleted in review. Oncol Biol Phys. 2011;79:1487–1495. involved-field radiotherapy in elderly (older than
111. Deleted in review. 172. Douw L, et al. Cognitive and radiological effects age 65) patients with newly diagnosed anaplastic
131. Brandes AA, et al. MGMT promoter methylation of radiotherapy in patients with low-grade glioma: astrocytoma or glioblastoma (Methusalem). J Clin
status can predict the incidence and outcome of long-term follow-up. Lancet Neurol. 2009;8:810– Oncol. 2010;28:LBA2001–LBA2001.
pseudoprogression after concomitant radiochemo- 818. 351. Deckert M, et al. Modern concepts in the biology,
therapy in newly diagnosed glioblastoma patients. 270. Mirimanoff RO, et al. Radiotherapy and temozolo- diagnosis, differential diagnosis and treatment
J Clin Oncol. 2008;26:2192–2197. mide for newly diagnosed glioblastoma: recursive of primary central nervous system lymphoma.
133. Wen PY, et al. Updated response assessment criteria partitioning analysis of the EORTC 26981/22981- Leukemia. 2011;25:1797–1807.
for high-grade gliomas: Response Assessment in NCIC CE3 phase III randomized trial. J Clin Oncol. 369. Porter AB, et al. Primary central nervous system
Neuro-Oncology Working Group. J Clin Oncol. 2006;24:2563–2569. lymphoma can be histologically diagnosed after
2010;28:1963–1972. 278. Stupp R, et al. Maintenance therapy with tumor- previous corticosteroid use: a pilot study to determine
134. Norden AD, et al. Bevacizumab for recurrent treating fields plus temozolomide vs temozolomide whether corticosteroids prevent the diagnosis of
malignant gliomas Efficacy, toxicity, and patterns alone for glioblastoma: a randomized clinical trial. primary central nervous system lymphoma. Ann
of recurrence. Neurology. 2008;70:779–787. JAMA. 2015;314:2535–2543. Neurol. 2008;63:662–667.
Cancer of the Central Nervous System • CHAPTER 63 967

399. Ferreri AJM, et al. Chemoimmunotherapy with medulloblastoma in children: conclusions from the glioma following chemotherapy: a multicenter ret-
methotrexate, cytarabine, thiotepa, and rituximab Children’s Cancer Group 921 randomized phase III rospective analysis. Neuro Oncol. 2012;14:790–797.
(MATRix regimen) in patients with primary CNS study. J Clin Oncol. 1999;17:832–845. 666. Pajtler KW, et al. Molecular classification of
lymphoma: results of the first randomisation of 607. Packer RJ, et al. Phase III study of craniospinal ependymal tumors across all CNS compartments,
the International Extranodal Lymphoma Study radiation therapy followed by adjuvant chemotherapy histopathological grades, and age groups. Cancer
Group-32 (IELSG32) phase 2 trial. Lancet Haematol. for newly diagnosed average-risk medulloblastoma. Cell. 2015;27:728–743.
2016;3:e217–e227. J Clin Oncol. 2006;24:4202–4208. 667. Parker M, et al. C11orf95-RELA fusions drive
414. Cahill KS, Claus EB. Treatment and survival of 625. Roberts CWM, Orkin SH. The SWI/SNF oncogenic NF-κB signalling in ependymoma.
patients with nonmalignant intracranial meningioma: complex—chromatin and cancer. Nat Rev Cancer. Nature. 2014;506:451–455.
results from the Surveillance, Epidemiology, and End 2004;4:133–142. 675. Robertson PL, et al. Survival and prognostic factors
Results Program of the National Cancer Institute. 630. Chi SN, et al. Intensive multimodality treatment for following radiation therapy and chemotherapy for
Clinical article. J Neurosurg. 2011;115:259–267. children with newly diagnosed CNS atypical teratoid ependymomas in children: a report of the Children’s
453. Molitch ME. Diagnosis and treatment of pituitary rhabdoid tumor. J Clin Oncol. 2009;27:385–389. Cancer Group. J Neurosurg. 1998;88:695–703.
adenomas: a review. JAMA. 2017;317:516–524. 634. Jennings MT, Freeman ML, Murray MJ. Strategies 694. Bouffet E, et al. Combined treatment modality for
570. Ostrom QT, et al. Alex’s Lemonade Stand Foundation in the treatment of diffuse pontine gliomas: the intracranial germinomas: results of a multicentre
Infant and Childhood Primary Brain and Central therapeutic role of hyperfractionated radiotherapy SFOP experience. Société Française d’Oncologie
Nervous System Tumors Diagnosed in the United and chemotherapy. J Neurooncol. 1996;28:207–222. Pédiatrique. Br J Cancer. 1999;79:1199–1204.
States in 2007-2011. Neuro Oncol. 2015;16(suppl 10): 649. Ater JL, et al. Randomized study of two chemo- 700. Hoffman HJ, et al. Aggressive surgical management
x1–x36. therapy regimens for treatment of low-grade glioma of craniopharyngiomas in children. J Neurosurg.
580. Listernick R, Ferner RE, Liu GT, Gutmann DH. in young children: a report from the Children’s 1992;76:47–52.
Optic pathway gliomas in neurofibromatosis-1: Oncology Group. J Clin Oncol. 2012;30:2641–2647. 706. Merchant TE, et al. Craniopharyngioma: the St. Jude
controversies and recommendations. Ann Neurol. 652. Lin A, et al. BRAF alterations in primary glial and Children’s Research Hospital experience 1984-2001.
2007;61:189–198. glioneuronal neoplasms of the central nervous system Int J Radiat Oncol Biol Phys. 2002;53:533–542.
592. Northcott PA, et al. Medulloblastoma comprises with identification of 2 novel KIAA1549:BRAF 715. Geyer JR, et al. Multiagent chemotherapy and
four distinct molecular variants. J Clin Oncol. fusion variants. J Neuropathol Exp Neurol. 2012; deferred radiotherapy in infants with malignant
2011;29:1408–1414. 71:66–72. brain tumors: a report from the Children’s Cancer
596. Zeltzer PM, et al. Metastasis stage, adjuvant treat- 657. Fisher MJ, et al. Visual outcomes in children with Group. J Clin Oncol. 2005;23:7621–7631.
ment, and residual tumor are prognostic factors for neurofibromatosis type 1-associated optic pathway
Cancer of the Central Nervous System • CHAPTER 63 967.e1
967.e1

REFERENCES
1. Ostrom QT, et al. CBTRUS statistical report: 21. Wrensch M, Minn Y, Chew T, Bondy M, Berger 45. Khalil BD, El-Sibai M. Rho GTPases in primary
primary brain and other central nervous system MS. Epidemiology of primary brain tumors: current brain tumor malignancy and invasion. J Neurooncol.
tumors diagnosed in the United States in 2009–2013. concepts and review of the literature. Neuro Oncol. 2012;108:333–339.
Neuro Oncol. 2016;18:v1–v75. 2002;4:278–299. 46. Hobbs SK, et al. Regulation of transport pathways in
2. Louis DN, et al. The 2007 WHO classification 22. Inskip PD, et al. Cellular-telephone use and brain tumor vessels: role of tumor type and microenviron-
of tumours of the central nervous system. Acta tumors. N Engl J Med. 2001;344:79–86. ment. Proc Natl Acad Sci USA. 1998;95:4607–4612.
Neuropathol. 2007;114:97–109. 23. Christensen HC, et al. Cellular telephones and 47. Fidler IJ, Yano S, Zhang RD, Fujimaki T, Bucana
3. Deorah S, Lynch CF, Sibenaller ZA, Ryken TC. risk for brain tumors: a population-based, incident CD. The seed and soil hypothesis: vascularisation
Trends in brain cancer incidence and survival in the case-control study. Neurology. 2005;64:1189–1195. and brain metastases. Lancet Oncol. 2002;3:53–57.
United States: Surveillance, Epidemiology, and End 24. Hepworth SJ, et al. Mobile phone use and risk 48. Bullitt E, et al. Vessel tortuosity and brain
Results Program, 1973 to 2001. Neurosurg Focus. of glioma in adults: case-control study. BMJ. tumor malignancy: a blinded study. Acad Radiol.
2006;20:E1. 2006;332:883–887. 2005;12:1232–1240.
4. Darefsky AS, King JT, Dubrow R. Adult glioblas- 25. Muscat JE, et al. Handheld cellular telephone use and 49. Jain RK, et al. Angiogenesis in brain tumours. Nat
toma multiforme survival in the temozolomide risk of brain cancer. JAMA. 2000;284:3001–3007. Rev Neurosci. 2007;8:610–622.
era: a population-based analysis of Surveillance, 26. Kan P, Simonsen SE, Lyon JL, Kestle JRW. Cel- 50. Chan AS, et al. Expression of vascular endothelial
Epidemiology, and End Results registries. Cancer. lular phone use and brain tumor: a meta-analysis. growth factor and its receptors in the anaplastic
2012;118:2163–2172. J Neurooncol. 2008;86:71–78. progression of astrocytoma, oligodendroglioma, and
5. Shu XO, et al. Diagnostic X-ray and ultrasound 27. Hardell L, Carlberg M, Mild KH. Case-control ependymoma. Am J Surg Pathol. 1998;22:816–826.
exposure and risk of childhood cancer. Br J Cancer. study of the association between the use of cel- 51. Fukumura D, et al. Hypoxia and acidosis indepen-
1994;70:531. lular and cordless telephones and malignant brain dently up-regulate vascular endothelial growth factor
6. Shore RE, Albert RE, Pasternack BS. Follow-up study tumors diagnosed during 2000-2003. Environ Res. transcription in brain tumors in vivo. Cancer Res.
of patients treated by x-ray epilation for tinea capitis: 2006;100:232–241. 2001;61:6020–6024.
resurvey of post-treatment illness and mortality 28. Baan R, et al. Carcinogenicity of radiofrequency 52. Carmeliet P, Jain RK. Angiogenesis in cancer and
experience. Arch Environ Health. 1976;31:21– electromagnetic fields. Lancet Oncol. 2011;12:624– other diseases. Nature. 2000;407:249–257.
28. 626. 53. Dvorak HF. Vascular permeability factor/vascular
7. Shapiro S, Mealey J Jr, Sartorius C. Radiation- 29. Narod SA, Stiller C, Lenoir GM. An estimate of the endothelial growth factor: a critical cytokine in tumor
induced intracranial malignant gliomas. J Neurosurg. heritable fraction of childhood cancer. Br J Cancer. angiogenesis and a potential target for diagnosis and
1989;71:77–82. 1991;63:993–999. therapy. J Clin Oncol. 2002;20:4368–4380.
8. Relling MV, et al. High incidence of secondary brain 30. Kimmelman A, Liang BC. Familial neurogenic 54. Ferrara N. Vascular endothelial growth factor:
tumours after radiotherapy and antimetabolites. tumor syndromes. Hematol Oncol Clin North Am. basic science and clinical progress. Endocr Rev.
Lancet. 1999;354:34–39. 2001;15:1073–1084. 2004;25:581–611.
9. Liwnicz BH, Berger TS, Liwnicz RG, Aron BS. 31. Ekstrand AJ, et al. Genes for epidermal growth 55. Fukumura D, Kashiwagi S, Jain RK. The role of
Radiation-associated gliomas: a report of four cases factor receptor, transforming growth factor alpha, nitric oxide in tumour progression. Nat Rev Cancer.
and analysis of postradiation tumors of the central and epidermal growth factor and their expression in 2006;6:521–534.
nervous system. Neurosurgery. 1985;17:436–445. human gliomas in vivo. Cancer Res. 1991;51:2164– 56. Kaur B, Tan C, Brat DJ, Post DE, Van Meir EG.
10. Ron E, et al. Tumors of the brain and nervous 2172. Genetic and hypoxic regulation of angiogenesis in
system after radiotherapy in childhood. NEJM. 32. Guha A, Dashner K, Black PM, Wagner JA, Stiles gliomas. J Neurooncol. 2004;70:229–243.
1988;319:1033–1039. CD. Expression of PDGF and PDGF receptors in 57. Zundel W, et al. Loss of PTEN facilitates
11. Karlsson P, et al. Intracranial tumors after exposure to human astrocytoma operation specimens supports HIF-1-mediated gene expression. Genes Dev.
ionizing radiation during infancy: a pooled analysis the existence of an autocrine loop. Int J Cancer. 2000;14:391–396.
of two Swedish cohorts of 28,008 infants with skin 1995;60:168–173. 58. Maity A, Pore N, Lee J, Solomon D, O’Rourke
hemangioma. Radiat Res. 1998;150:357–364. 33. Glick RP, Lichtor T, Unterman TG. Insulin-like DM. Epidermal growth factor receptor transcrip-
12. Jenkinson H, Hawkins M. Secondary brain tumours growth factors in central nervous system tumors. tionally up-regulates vascular endothelial growth
in children with ALL. Lancet. 1999;354. J Neurooncol. 1997;35:315–325. factor expression in human glioblastoma cells via
13. Walter AW, et al. Secondary brain tumors in children 34. Rao RD, James CD. Altered molecular pathways a pathway involving phosphatidylinositol 3′-kinase
treated for acute lymphoblastic leukemia at St in gliomas: an overview of clinically relevant issues. and distinct from that induced by hypoxia. Cancer
Jude Children’s Research Hospital. J Clin Oncol. Semin Oncol. 2004;31:595–604. Res. 2000;60:5879–5886.
1998;16:3761–3767. 35. Tang P, Steck PA, Yung WK. The autocrine loop of 59. Pore N, Liu S, Haas-Kogan DA, O’Rourke DM,
14. Löning L, et al. Secondary neoplasms subsequent TGF-alpha/EGFR and brain tumors. J Neurooncol. Maity A. PTEN mutation and epidermal growth
to Berlin-Frankfurt-Münster therapy of acute 1997;35:303–314. factor receptor activation regulate vascular endothelial
lymphoblastic leukemia in childhood: significantly 36. Abounader R, Laterra J. Scatter factor/hepatocyte growth factor (VEGF) mRNA expression in human
lower risk without cranial radiotherapy. Blood. growth factor in brain tumor growth and angio- glioblastoma cells by transactivating the proximal
2000;95:2770–2775. genesis. Neuro Oncol. 2005;7:436–451. VEGF promoter. Cancer Res. 2003;63:236–241.
15. Neglia JP, et al. Second neoplasms after acute 37. Haas-Kogan D, et al. Protein kinase B (PKB/Akt) 60. Evans SM, et al. Comparative measurements
lymphoblastic leukemia in childhood. NEJM. activity is elevated in glioblastoma cells due to of hypoxia in human brain tumors using
1991;325:1330–1336. mutation of the tumor suppressor PTEN/MMAC. needle electrodes and EF5 binding. Cancer Res.
16. Duffner PK, et al. Second malignancies in young Curr Biol. 1998;8:1195–1198. 2004;64:1886–1892.
children with primary brain tumors following treat- 38. Guha A. Ras activation in astrocytomas and 61. Lally BE, et al. The interactions of polarographic
ment with prolonged postoperative chemotherapy neurofibromas. Can J Neurol Sci. 1998;25:267–281. measurements of oxygen tension and histological
and delayed irradiation: a Pediatric Oncology Group 39. Woods SA, et al. Aberrant G protein signaling grade in human glioma. Cancer J. 2006;12:461–466.
study. Ann Neurol. 1998;44:313–316. in nervous system tumors. J Neurosurg. 2002;97: 62. Evans SM, et al. Hypoxia is important in the biology
17. Shapiro S, Mealey J Jr. Late anaplastic gliomas in 627–642. and aggression of human glial brain tumors. Clin
children previously treated for acute lymphoblastic 40. Augustin I, et al. The Wnt secretion protein Evi/ Cancer Res. 2004;10:8177–8184.
leukemia. Pediatr Neurosurg. 1989;15:176–180. Gpr177 promotes glioma tumourigenesis. EMBO 63. Jensen RL. Brain tumor hypoxia: tumorigenesis,
18. Berleur MP, Cordier S. The role of chemical, physical, Mol Med. 2012;4:38–51. angiogenesis, imaging, pseudoprogression, and as
or viral exposures and health factors in neurocar- 41. Zhang K, Zhang J, Han L, Pu P, Kang C. Wnt/ a therapeutic target. J Neurooncol. 2009;92:317–
cinogenesis: implications for epidemiologic studies beta-catenin signaling in glioma. J Neuroimmune 335.
of brain tumors. Cancer Causes Control. 1995;6: Pharmacol. 2012;7:740–749. 64. Rong Y, Durden DL, Van Meir EG, Brat DJ.
240–256. 42. Bell E, Karnosh LJ. Cerebral hemispherectomy; ‘Pseudopalisading’ necrosis in glioblastoma: a familiar
19. Dziurzynski K, et al. Consensus on the role of report of a case 10 years after operation. J Neurosurg. morphologic feature that links vascular pathology,
human cytomegalovirus in glioblastoma. Neuro 1949;6:285–293. hypoxia, and angiogenesis. J Neuropathol Exp Neurol.
Oncol. 2012;14:246–255. 43. Onishi M, Ichikawa T, Kurozumi K, Date I. 2006;65:529–539.
20. Lehrer S, Green S, Ramanathan L, Rosenzweig K, Angiogenesis and invasion in glioma. Brain Tumor 65. Singh SK, et al. Identification of a cancer stem
Labombardi V. No consistent relationship of glioblas- Pathol. 2011;28:13–24. cell in human brain tumors. Cancer Res. 2003;63:
toma incidence and cytomegalovirus seropositivity 44. Demuth T, Berens ME. Molecular mechanisms of 5821–5828.
in whites, blacks, and Hispanics. Anticancer Res. glioma cell migration and invasion. J Neurooncol. 66. Beier D, et al. CD133+ and CD133− glioblastoma-
2012;32:1113–1115. 2004;70:217–228. derived cancer stem cells show differential growth
967.e2 Part III: Specific Malignancies

characteristics and molecular profiles. Cancer Res. 92. Schiff D. Pneumocystis pneumonia in brain contrast MR imaging. AJR Am J Roentgenol. 1994;163:
2007;67:4010–4015. tumor patients: risk factors and clinical features. 181–186.
67. Bao S, et al. Glioma stem cells promote radioresis- J Neurooncol. 1996;27:235–240. 116. Alsop DC, et al. Functional activation during
tance by preferential activation of the DNA damage 93. Glück T, et al. Pneumocystis carinii pneumonia an auditory comprehension task in patients with
response. Nature. 2006;444:756–760. as a complication of immunosuppressive therapy. temporal lobe lesions. Neuroimage. 1996;4:55–59.
68. Piccirillo SGM, et al. Bone morphogenetic proteins Infection. 2000;28:227–230. 117. Atlas SW, et al. Functional magnetic resonance
inhibit the tumorigenic potential of human brain 94. Coursin DB, Wood KE. Corticosteroid supplemen- imaging of regional brain activity in patients with
tumour-initiating cells. Nature. 2006;444:761–765. tation for adrenal insufficiency. JAMA. 2002;287: intracerebral gliomas: findings and implications
69. Calabrese C, et al. A perivascular niche for brain 236–240. for clinical management. Neurosurgery. 1996;38:
tumor stem cells. Cancer Cell. 2007;11:69–82. 95. Neary N, Nieman L. Adrenal insufficiency: etiology, 329–338.
70. Rahaman SO, et al. Inhibition of constitutively diagnosis and treatment. Curr Opin Endocrinol 118. Roux FE, et al. Usefulness of motor functional
active Stat3 suppresses proliferation and induces Diabetes Obes. 2010;17:217–223. MRI correlated to cortical mapping in Rolandic
apoptosis in glioblastoma multiforme cells. Oncogene. 96. Gerstner ER, et al. VEGF inhibitors in the treatment low-grade astrocytomas. Acta Neurochir (Wien).
2002;21:8404. of cerebral edema in patients with brain cancer. Nat 1999;141:71–79.
71. Hadjipanayis CG, et al. EGFRvIII antibody– Rev Clin Oncol. 2009;6:229–236. 119. Maldjian JA, et al. Intraoperative functional MRI
conjugated iron oxide nanoparticles for magnetic 97. Wu AS, et al. A prospective randomized trial of using a real-time neurosurgical navigation system.
resonance imaging–guided convection-enhanced perioperative seizure prophylaxis in patients with J Comput Assist Tomogr. 1997;21:910–912.
delivery and targeted therapy of glioblastoma. Cancer intraparenchymal brain tumors. J Neurosurg. 2013; 120. Berman JI, Berger MS, Chung S, Nagarajan SS,
Res. 2010;70:6303–6312. 118:873–883. Henry RG. Accuracy of diffusion tensor magnetic
72. Lim KJ, Bisht S, Bar EE, Maitra A, Eberhart CG. 98. Foy PM, Chadwick DW, Rajgopalan N, Johnson AL, resonance imaging tractography assessed using
A polymeric nanoparticle formulation of curcumin Shaw MD. Do prophylactic anticonvulsant drugs intraoperative subcortical stimulation mapping
inhibits growth, clonogenicity and stem-like fraction alter the pattern of seizures after craniotomy? J Neurol and magnetic source imaging; 2007.
in malignant brain tumors. Cancer Biol Ther. 2011;11: Neurosurg Psychiatry. 1992;55:753–757. 121. Nelson SJ, Vigneron DB, Dillon WP. Serial evalua-
464–473. 99. Forsyth PA, et al. Prophylactic anticonvulsants tion of patients with brain tumors using volume MRI
73. Nduom EKE, Hadjipanayis CG, Van Meir EG. in patients with brain tumour. Can J Neurol Sci. and 3D 1H MRSI. NMR Biomed. 1999;12:123–138.
Glioblastoma cancer stem-like cells—implications for 2003;30:106–112. 122. Delorme S, Weber MA. Applications of MRS in the
pathogenesis and treatment. Cancer J. 2012;18:100. 100. Chan AT, et al. Venous thromboembolism occurs evaluation of focal malignant brain lesions. Cancer
74. Tabatabai G, Weller M. Glioblastoma stem cells. frequently in patients undergoing brain tumor Imaging. 2006;6:95–99.
Cell Tissue Res. 2011;343:459–465. surgery despite prophylaxis. J Thromb Thrombolysis. 123. Hornig CR, et al. CT contrast enhancement on brain
75. Manoranjan B, et al. Medulloblastoma stem cells: 1999;8:139–142. scans and blood-CSF barrier disturbances in cerebral
where development and cancer cross pathways. 101. Lim DA, et al. Safety and feasibility of switching ischemic infarction. Stroke. 1985;16:268–273.
Pediatr Res. 2012;71:516–522. from phenytoin to levetiracetam monotherapy for 124. McCormack BM, Miller DC, Budzilovich GN,
76. Poppleton H, Gilbertson RJ. Stem cells of ependy- glioma-related seizure control following craniotomy: Voorhees GJ, Ransohoff J. Treatment and survival
moma. Br J Cancer. 2007;96:6–10. a randomized phase II pilot study. J Neurooncol. of low-grade astrocytoma in adults–1977–1988.
77. Witwer BP, et al. Diffusion-tensor imaging of white 2009;93:349–354. Neurosurgery. 1992;31:636–642.
matter tracts in patients with cerebral neoplasm. J 102. Milligan TA, Hurwitz S, Bromfield EB. Efficacy 125. Philippon JH, Clemenceau SH, Fauchon FH, Foncin
Neurosurg. 2002;97:568–575. and tolerability of levetiracetam versus phe- JF. Supratentorial low-grade astrocytomas in adults.
78. Holodny AI, Ollenschlager M. Diffusion imaging nytoin after supratentorial neurosurgery. Neurology. Neurosurgery. 1993;32:554–559.
in brain tumors. Neuroimaging Clin N Am. 2008;71:665–669. 126. Chamberlain MC, Murovic JA, Levin VA. Absence of
2002;12:107–124. 103. Villanueva V, et al. NEOPLASM study: real-life use contrast enhancement on CT brain scans of patients
79. Sinha S, Bastin ME, Whittle IR, Wardlaw JM. of lacosamide in patients with brain tumor-related with supratentorial malignant gliomas. Neurology.
Diffusion tensor MR imaging of high-grade cerebral epilepsy. Epilepsy Behav. 2016;65:25–32. 1988;38:1371–1374.
gliomas. AJNR Am J Neuroradiol. 2002;23:520–527. 104. Saria MG, et al. Retrospective analysis of the 127. Geer CP, et al. Does MR perfusion imaging impact
80. Mori S, et al. Brain white matter anatomy of tumor tolerability and activity of lacosamide in patients management decisions for patients with brain
patients evaluated with diffusion tensor imaging. with brain tumors: clinical article. J Neurosurg. tumors? A prospective study. AJNR Am J Neuroradiol.
Ann Neurol. 2002;51:377–380. 2013;118:1183–1187. 2012;33:556–562.
81. Kernohan JW, Woltman HW. Incisura of the crus 105. Brandes AA, et al. Incidence and risk of thrombo- 128. Celli P, Nofrone I, Palma L, Cantore G, Fortuna
due to contralateral brain tumor. Arch NeurPsych. embolism during treatment of high-grade gliomas: a A. Cerebral oligodendroglioma prognostic factors
1929;21:274–287. prospective study. Eur J Cancer. 1997;33:1592–1596. and life history. Neurosurgery. 1994;35:1018–
82. Cushing H. Concerning a definite regulatory 106. Iorio A, Agnelli G. Low-molecular-weight and 1035.
mechanism of the vaso-motor centre which controls unfractionated heparin for prevention of venous 129. Cornelius JF, et al. Positron emission tomography
blood pressure during cerebral compression. Bull thromboembolism in neurosurgery: a meta-analysis. imaging of meningioma in clinical practice: review
Hopkins Hosp. 1901;290–292. Arch Intern Med. 2000;160:2327–2332. of literature and future directions. Neurosurgery.
83. Cushing H. Some experimental and clinical obser- 107. Agnelli G, et al. Enoxaparin plus compression stock- 2012;70:1033–1042.
vations concerning states of increased intracranial ings compared with compression stockings alone in 130. Taal W, et al. Incidence of early pseudo-progression
tension. Am J Med Sci. 1902;375–400. the prevention of venous thromboembolism after in a cohort of malignant glioma patients treated
84. Rudà R, Trevisan E, Soffietti R. Epilepsy and brain elective neurosurgery. NEJM. 1998;339:80–85. with chemoirradiation with temozolomide. Cancer.
tumors. Curr Opin Oncol. 2010;22:611–620. 108. Deleted in review. 2008;113:405–410.
85. Rossetti AO, Stupp R. Epilepsy in brain tumor 109. Levin JM, et al. Complications of therapy for 131. Brandes AA, et al. MGMT promoter methylation
patients. Curr Opin Neurol. 2010;23:603–609. venous thromboembolic disease in patients with status can predict the incidence and outcome of
86. Huber A. Eye Symptoms in Brain Tumors. CV brain tumors. Neurology. 1993;43:1111–1114. pseudoprogression after concomitant radiochemo-
Mosby Co, 1971. 110. Kelly PJ, et al. Imaging-based stereotaxic serial therapy in newly diagnosed glioblastoma patients.
87. Roth P, Wick W, Weller M. Steroids in neurooncol- biopsies in untreated intracranial glial neoplasms. J Clin Oncol. 2008;26:2192–2197.
ogy: actions, indications, side-effects. Curr Opin J Neurosurg. 1987;66:865–874. 132. Quant EC, Wen PY. Response Assessment in
Neurol. 2010;23:597–602. 111. Deleted in review. Neuro-Oncology. Curr Oncol Rep. 2011;13:50–56.
88. Ryken TC, et al. The role of steroids in the man- 112. Law M, et al. Glioma grading: sensitivity, specificity, 133. Wen PY, et al. Updated response assessment criteria
agement of brain metastases: a systematic review and predictive values of perfusion MR imaging and for high-grade gliomas: Response Assessment in
and evidence-based clinical practice guideline. proton MR spectroscopic imaging compared with Neuro-Oncology Working Group. J Clin Oncol.
J Neurooncol. 2010;96:103–114. conventional MR imaging. AJNR Am J Neuroradiol. 2010;28:1963–1972.
89. Dietrich J, Rao K, Pastorino S, Kesari S. Corticoster- 2003;24:1989–1998. 134. Norden AD, et al. Bevacizumab for recurrent
oids in brain cancer patients: benefits and pitfalls. 113. Preul C, et al. Differentiation of cerebral tumors malignant gliomas: efficacy, toxicity, and patterns
Expert Rev Clin Pharmacol. 2011;4:233–242. using multi-section echo planar MR perfusion of recurrence. Neurology. 2008;70:779–787.
90. Watling CJ, Cairncross JG. Acetazolamide therapy imaging. Eur J Radiol. 2003;48:244–251. 135. Gerstner ER, Frosch MP, Batchelor TT. Diffusion
for symptomatic plateau waves in patients with 114. Uematsu H, et al. Blood volume of gliomas magnetic resonance imaging detects pathologically
brain tumors. Report of three cases. J Neurosurg. determined by double-echo dynamic perfusion- confirmed, nonenhancing tumor progression in
2002;97:224–226. weighted MR imaging: a preliminary study. AJNR a patient with recurrent glioblastoma receiving
91. Dropcho EJ, Soong SJ. Steroid-induced weakness Am J Neuroradiol. 2001;22:1915–1919. bevacizumab. J Clin Oncol. 2009;28:e91–e93.
in patients with primary brain tumors. Neurology. 115. Maeda M, et al. Vascularity of meningiomas and 136. Brandes AA, et al. Recurrence pattern after temozolo-
1991;41:1235–1239. neuromas: assessment with dynamic susceptibility- mide concomitant with and adjuvant to radiotherapy
Cancer of the Central Nervous System • CHAPTER 63 967.e3
967.e3

in newly diagnosed patients with glioblastoma: irradiation in normal monkey brain. Neurosurgery. of human embryonic stem cells. Proc Natl Acad Sci
correlation with MGMT promoter methylation 1997;41:209–219. USA. 2009;106:19150–19155.
status. J Clin Oncol. 2009;27:1275–1279. 157. Chuba PJ, et al. Hyperbaric oxygen therapy for 177. Acharya MM, Roa DE, Bosch O, Lan ML, Limoli
137. Batchelor TT, Sorensen AG, Louis DN. Case radiation-induced brain injury in children. Cancer. CL. Stem cell transplantation strategies for the
17-2012: a 54-year-old man with visual-field loss and 1997;80:2005–2012. restoration of cognitive dysfunction caused by cranial
a mass in the brain. NEJM. 2012;366:2112–2120. 158. Glantz MJ, et al. Treatment of radiation-induced radiotherapy. J Vis Exp. 2011;e3107–e3107.
138. Lacroix M, et al. A multivariate analysis of 416 nervous system injury with heparin and warfarin. 178. Griffin T. White matter necrosis, microangiopathy
patients with glioblastoma multiforme: prognosis, Neurology. 1994;44:2020–2027. and intellectual abilities in survivors of childhood
extent of resection, and survival. J Neurosurg. 2001;95: 159. Chan AS, Cheung MC, Law SC, Chan JH. Phase II leukemia. Association with central nervous system
190–198. study of alpha-tocopherol in improving the cognitive irradiation and methotrexate toxicity. In: Ang K, van
139. Wisoff JH, et al. Current neurosurgical management function of patients with temporal lobe radionecrosis. der Kogel A, van der Schueren E, eds. Radiation
and the impact of the extent of resection in the Cancer. 2004;100:398–404. Damage to the Central Nervous System. Raven Press;
treatment of malignant gliomas of childhood: a 160. Chen J, et al. Radiation induced temporal lobe 1980:155–174.
report of the Children’s Cancer Group trial no. necrosis in patients with nasopharyngeal carcinoma: 179. Mitchell WG, et al. Stroke as a late sequela of
CCG-945. J Neurosurg. 1998;89:52–59. a review of new avenues in its management. Radiat cranial irradiation for childhood brain tumors.
140. Sanai N, Polley MY, McDermott MW, Parsa AT, Oncol. 2011;6:128. J Child Neurol. 1991;6:128–133.
Berger MS. An extent of resection threshold for 161. Levin VA, et al. Randomized double-blind placebo- 180. Livesey EA, et al. Endocrine disorders following
newly diagnosed glioblastomas: clinical article. J controlled trial of bevacizumab therapy for radiation treatment of childhood brain tumours. Br J Cancer.
Neurosurg. 2011;115:3–8. necrosis of the central nervous system. Int J Radiat 1990;61:622.
141. Yuh GE, et al. Reducing toxicity from craniospinal Oncol Biol Phys. 2011;79:1487–1495. 181. Sklar CA, Constine LS. Chronic neuroendocrinologi-
irradiation: using proton beams to treat medulloblas- 162. Jankovic M, et al. Association of 1800 cGy cranial cal sequelae of radiation therapy. Int J Radiat Oncol
toma in young children. Cancer J. 2004;10:386–390. irradiation with intellectual function in children Biol Phys. 1995;31:1113–1121.
142. Yoon M, et al. Craniospinal irradiation techniques: with acute lymphoblastic leukaemia. Lancet. 182. Oberfield SE, et al. Endocrine late effects of child-
a dosimetric comparison of proton beams with 1994;344:224–227. hood cancers. J Pediatr. 1997;131:S37–S41.
standard and advanced photon radiotherapy. Int J 163. Halberg FE, et al. Prophylactic cranial irradiation 183. Rappaport R, Brauner R. Growth and endocrine
Radiat Oncol Biol Phys. 2011;81:637–646. dose effects on late cognitive function in children disorders secondary to cranial irradiation. Pediatr
143. Schultheiss TE, Kun LE, Ang KK, Stephens LC. treated for acute lymphoblastic leukemia. Int J Radiat Res. 1989;25:561–567.
Radiation response of the central nervous system. Oncol Biol Phys. 1992;22:13–16. 184. Clayton PE, Shalet SM. Dose dependency of time
Int J Radiat Oncol Biol Phys. 1995;31:1093–1112. 164. Meadows A, et al. Declines in IQ scores and cognitive of onset of radiation-induced growth hormone
144. Freeman JE, Johnston PGB, Voke JM. Somnolence dysfunctions in children with acute lymphocytic deficiency. J Pediatr. 1991;118:226–228.
after prophylactic cranial irradiation in children leukaemia treated with cranial irradiation. Lancet. 185. Leiper AD, Stanhope R, Kitching P, Chessells JM.
with acute lymphoblastic leukaemia. Br Med J. 1981;318:1015–1018. Precocious and premature puberty associated with
1973;4:523–525. 165. Mulhern RK, Fairclough D, Ochs J. A prospective treatment of acute lymphoblastic leukaemia. Arch
145. Cohen ME, Duffner PK. Long-term consequences of comparison of neuropsychologic performance of chil- Dis Child. 1987;62:1107–1112.
CNS treatment for childhood cancer, Part I: patho- dren surviving leukemia who received 18-Gy, 24-Gy, 186. Oberfield SE, Allen JC, Pollack J, New MI, Levine
logic consequences and potential for oncogenesis. or no cranial irradiation. J Clin Oncol. 1991;9: LS. Long-term endocrine sequelae after treatment of
Pediatr Neurol. 1991;7:157–163. 1348–1356. medulloblastoma: prospective study of growth and
146. Duffner PK, Cohen ME. Long-term consequences of 166. Ochs J, et al. Comparison of neuropsychologic thyroid function. J Pediatr. 1986;108:219–223.
CNS treatment for childhood cancer, Part II: clinical functioning and clinical indicators of neurotoxic- 187. Constine LS, et al. Hypothalamic-pituitary dys-
consequences. Pediatr Neurol. 1991;7:237–242. ity in long-term survivors of childhood leukemia function after radiation for brain tumors. NEJM.
147. Siu A, et al. Radiation necrosis following treatment given cranial radiation or parenteral methotrexate: 1993;328:87–94.
of high grade glioma—a review of the literature a prospective study. J Clin Oncol. 1991;9:145–151. 188. Ogilvy-Stuart AL, Shalet SM, Gattamaneni HR.
and current understanding. Acta Neurochir (Wien). 167. Waber DP, et al. Cognitive sequelae of treatment Thyroid function after treatment of brain tumors
2012;154:191–201. in childhood acute lymphoblastic leukemia: cranial in children. J Pediatr. 1991;119:733–737.
148. Shaw E, et al. Prospective randomized trial of low- radiation requires an accomplice. J Clin Oncol. 189. Parsons JT, Bova FJ, Fitzgerald CR, Mendenhall
versus high-dose radiation therapy in adults with 1995;13:2490–2496. WM, Million RR. Radiation optic neuropathy after
supratentorial low-grade glioma: initial report of a 168. Palmer SL, et al. Patterns of intellectual development megavoltage external-beam irradiation: analysis of
North Central Cancer Treatment Group/Radiation among survivors of pediatric medulloblastoma: a time-dose factors. Int J Radiat Oncol Biol Phys.
Therapy Oncology Group/Eastern Cooperative longitudinal analysis. J Clin Oncol. 2001;19:2302– 1994;30:755–763.
Oncology Group study. J Clin Oncol. 2002;20:2267– 2308. 190. Kline LB, Kim JY, Ceballos R. Radiation optic
2276. 169. Brown PD, et al. The effects of radiation on cognitive neuropathy. Ophthalmology. 1985;92:1118–1126.
149. Ruben JD, et al. Cerebral radiation necrosis: function in patients with low-grade glioma. Int J 191. Young WC, Thornton AF, Gebarski SS, Cornblath
incidence, outcomes, and risk factors with emphasis Radiat Oncol Biol Phys. 2001;51:135. WT. Radiation-induced optic neuropathy: correlation
on radiation parameters and chemotherapy. Int J 170. Armstrong CL, et al. Late cognitive and radiographic of MR imaging and radiation dosimetry. Radiology.
Radiat Oncol Biol Phys. 2006;65:499–508. changes related to radiotherapy Initial prospective 1992;185:904–907.
150. Constine LS, Konski A, Ekholm S, McDonald findings. Neurology. 2002;59:40–48. 192. Aristizabal S, Caldwell WL, Avila J. The relationship
S, Rubin P. Adverse effects of brain irradiation 171. Klein M, et al. Effect of radiotherapy and other of time-dose fractionation factors to complications
correlated with MR and CT imaging. Int J Radiat treatment-related factors on mid-term to long-term in the treatment of pituitary tumors by irradiation.
Oncol Biol Phys. 1988;15:319–330. cognitive sequelae in low-grade gliomas: a compara- Int J Radiat Oncol Biol Phys. 1977;2:667–673.
151. Buchpiguel CA, Alavi JB, Alavi A, Kenyon LC. PET tive study. Lancet. 2002;360:1361–1368. 193. Harris JR, Levene MB. Visual Complications
versus SPECT in distinguishing radiation necrosis 172. Douw L, et al. Cognitive and radiological effects following irradiation for pituitary adenomas and cra-
from tumor recurrence in the brain. J Nucl Med. of radiotherapy in patients with low-grade glioma: niopharyngiomas 1. Radiology. 1976;120:167–171.
1995;36:159–164. long-term follow-up. Lancet Neurol. 2009;8:810– 194. Stafford SL, et al. A study on the radiation tolerance
152. Di Chiro G, et al. Cerebral necrosis after radio- 818. of the optic nerves and chiasm after stereotactic
therapy and/or intraarterial chemotherapy for brain 173. Mulhern RK, et al. Short-term efficacy of methyl- radiosurgery. Int J Radiat Oncol Biol Phys. 2003;55:
tumors: PET and neuropathologic studies. AJR Am phenidate: a randomized, double-blind, placebo- 1177–1181.
JRoentgenol. 1988;150:189–197. controlled trial among survivors of childhood cancer. 195. Ove R, Kelman S, Amin PP, Chin LS. Preserva-
153. Phillips P, Packer R, Bleyer W, Pochedly C. J Clin Oncol. 2004;22:4795–4803. tion of visual fields after peri-sellar gamma-knife
Positon emission tomography studies of transport 174. Butler JM, et al. A phase III, double-blind, placebo- radiosurgery. Int J Cancer. 2000;90:343–350.
and metabolism in brain tumors. In: Pediatric controlled prospective randomized clinical trial of 196. Danesh-Meyer HV. Radiation-induced optic
Neuro-Oncology. Harwood Academic Publishers; d-threo-methylphenidate HCl in brain tumor neuropathy. J Clin Neurosci. 2008;15:95–100.
1992:91–110. patients receiving radiation therapy. Int J Radiat 197. Hawkins MM, Draper GJ, Kingston JE. Incidence
154. Deleted in review. Oncol Biol Phys. 2007;69:1496–1501. of second primary tumours among childhood cancer
155. Pružincová L, et al. MR imaging of late radiation 175. Shaw EG, et al. Phase II study of donepezil in survivors. Br J Cancer. 1987;56:339.
therapy-and chemotherapy-induced injury: a pictorial irradiated brain tumor patients: effect on cognitive 198. Jones A. Transient radiation myelopathy. Br J Radiol.
essay. Eur Radiol. 2009;19:2716. function, mood, and quality of life. J Clin Oncol. 1964;37:727–744.
156. Tada E, Matsumoto K, Kinoshita K, Furuta T, 2006;24:1415–1420. 199. Schultheiss TE, Stephens LC, Peters LJ. Survival in
Ohmoto T. The protective effect of dexamethasone 176. Acharya MM, et al. Rescue of radiation-induced radiation myelopathy. Int J Radiat Oncol Biol Phys.
against radiation damage induced by interstitial cognitive impairment through cranial transplantation 1986;12:1765–1769.
967.e4 Part III: Specific Malignancies

200. Wang PY, Shen WC, Jan JS. MR imaging in 222. Shaw EG, Scheithauer BW, O’Fallon JR. Supraten- enhanced tumorigenicity. Proc Natl Acad Sci USA.
radiation myelopathy. AJNR Am J Neuroradiol. torial gliomas: a comparative study by grade and 1994;91:7727–7731.
1992;13:1049–1055. histologic type. J Neurooncol. 1997;31:273–278. 245. Schmidt EE, et al. Mutational profile of the PTEN
201. Marcus RB, Million RR. The incidence of myelitis 223. Nobusawa S, Watanabe T, Kleihues P, Ohgaki gene in primary human astrocytic tumors and culti-
after irradiation of the cervical spinal cord. Int J H. IDH1 mutations as molecular signature and vated xenografts. J Neuropathol Exp Neurol. 1999;58:
Radiat Oncol Biol Phys. 1990;19:3–8. predictive factor of secondary glioblastomas. Clin 1170–1183.
202. Dietrich J, Monje M, Wefel J, Meyers C. Clinical Cancer Res. 2009;15:6002–6007. 246. Ermoian RP, et al. Dysregulation of PTEN and
patterns and biological correlates of cognitive dys- 224. Parsons DW, et al. An integrated genomic protein kinase B is associated with glioma histol-
function associated with cancer therapy. Oncologist. analysis of human glioblastoma multiforme. Science. ogy and patient survival. Clin Cancer Res. 2002;8:
2008;13:1285–1295. 2008;321:1807–1812. 1100–1106.
203. Hochberg FH, Pruitt A. Assumptions in the 225. Yan H, et al. IDH1 and IDH2 mutations in gliomas. 247. Smith JS, et al. PTEN mutation, EGFR ampli-
radiotherapy of glioblastoma. Neurology. 1980;30: NEJM. 2009;360:765–773. fication, and outcome in patients with anaplastic
907–911. 226. Fu Y, et al. Glioma-derived mutations in IDH: from astrocytoma and glioblastoma multiforme. J Natl
204. Sahm F, et al. Addressing diffuse glioma as a systemic mechanism to potential therapy. Biochem Biophys Cancer Inst. 2001;93:1246–1256.
brain disease with single-cell analysis. Arch Neurol. Res Commun. 2010;397:127–130. 248. Reifenberger J, et al. Analysis of p53 mutation and
2012;69:523–526. 227. Shibahara I, et al. New insights into glioma clas- epidermal growth factor receptor amplification in
205. Kloosterhof NK, Bralten LB, Dubbink HJ, French sification based on isocitrate dehydrogenase 1 and recurrent gliomas with malignant progression.
PJ, van den Bent MJ. Isocitrate dehydrogenase-1 2 gene status. Brain Tumor Pathol. 2011;28:203. J Neuropathol Exp Neurol. 1996;55:822–831.
mutations: a fundamentally new understanding of 228. Dang L, et al. Cancer-associated IDH1 mutations 249. Tohma Y, et al. PTEN (MMAC1) mutations are
diffuse glioma? Lancet Oncol. 2011;12:83–91. produce 2-hydroxyglutarate. Nature. 2009;462: frequent in primary glioblastomas (de novo) but
206. Ishihara H, et al. Endothelial cell barrier impairment 739–744. not in secondary glioblastomas. J Neuropathol Exp
induced by glioblastomas and transforming growth 229. Kil IS, Kim SY, Lee SJ, Park JW. Small interfering Neurol. 1998;57:684–689.
factor β2 involves matrix metalloproteinases and RNA-mediated silencing of mitochondrial NADP+- 250. Watanabe K, et al. Overexpression of the EGF recep-
tight junction proteins. J Neuropathol Exp Neurol. dependent isocitrate dehydrogenase enhances the tor and p53 mutations are mutually exclusive in the
2008;67:435–448. sensitivity of HeLa cells toward tumor necrosis evolution of primary and secondary glioblastomas.
207. Benarroch EE. Blood–brain barrier: recent develop- factor-α and anticancer drugs. Free Radic Biol Med. Brain Pathol. 1996;6:217–223.
ments and clinical correlations. Neurology. 2012;78: 2007;43:1197–1207. 251. Deleted in review.
1268–1276. 230. Ronnebaum SM, et al. A pyruvate cycling 252. Jackson RJ, et al. Limitations of stereotactic biopsy
208. Mass MK, et al. Neurotoxicity of chemotherapeutic- pathway involving cytosolic NADP-dependent in the initial management of gliomas. Neuro Oncol.
agents and immunoconjugates delivered after isocitrate dehydrogenase regulates glucose-stimulated 2001;3:193–200.
blood-brain-barrier modification—neuropathological insulin secretion. J Biol Chem. 2006;281:30593– 253. Kondziolka D, Dade Lunsford L. The role of
studies. Ann Neurol. 1995;38:342–342. 30602. stereotactic biopsy in the management of gliomas.
209. Shapiro WR, et al. A randomized comparison of 231. Bralten LB, et al. IDH1 R132H decreases prolifera- J Neurooncol. 1999;42:205–213.
intra-arterial versus intravenous with or without tion of glioma cell lines in vitro and in vivo. Ann 254. Laws ER Jr. Radical resection for the treatment of
intravenous 5-fluorouracil, for newly diagnosed Neurol. 2011;69:455–463. glioma. Clin Neurosurg. 1994;42:480–487.
patients with malignant glioma. J Neurosurg. 1992;76: 232. Capper D, et al. Characterization of R132H 255. Duffau H. A new philosophy in surgery for diffuse
772–781. mutation-specific IDH1 antibody binding in brain low-grade glioma (DLGG): oncological and func-
210. Prados MD, et al. A randomized, double-blind, tumors. Brain Pathol. 2010;20:245–254. tional outcomes. Neurochirurgie. 2013;59:2–8.
placebo-controlled, phase 2 study of RMP-7 in 233. Jeuken JW, Von Deimling A, Wesseling P. Molecular 256. Coffey RJ, Lunsford DL, Taylor FH. Survival
combination with carboplatin administered intra- pathogenesis of oligodendroglial tumors. J Neuroon- after stereotactic biopsy of malignant gliomas.
venously for the treatment of recurrent malignant col. 2004;70:161–181. Neurosurgery. 1988;22:465–473.
glioma. Neuro Oncol. 2003;5:96–103. 234. Reifenberger J, et al. Molecular genetic analysis of 257. Schulder M, Sernas TJ, Carmel PW. Cranial surgery
211. Cao Y, et al. Use of magnetic resonance imaging oligodendroglial tumors shows preferential allelic and navigation with a compact intraoperative MRI
to assess blood-brain/blood-glioma barrier opening deletions on 19q and 1p. Am J Pathol. 1994;145: system. In: Intraoperative Imaging in Neurosurgery.
during conformal radiotherapy. J Clin Oncol. 1175. Springer; 2003:79–86.
2005;23:4127–4136. 235. Maintz D, et al. Molecular genetic evidence for 258. Matz PG, Cobbs C, Berger MS. Intraoperative
212. Ludwig CL, Smith MT, Godfrey AD, Armbrustm- subtypes of oligoastrocytomas. J Neuropathol Exp cortical mapping as a guide to the surgical resection
acher VW. A clinicopathological study of 323 patients Neurol. 1997;56:1098–1104. of gliomas. J Neurooncol. 1999;42:233–245.
with oligodendrogliomas. Ann Neurol. 1986;19: 236. Phillips HS, et al. Molecular subclasses of high- 259. Duffau H, et al. Usefulness of intraoperative electrical
15–21. grade glioma predict prognosis, delineate a pattern subcortical mapping during surgery for low-grade
213. Adson AW, Svien H. Symposium on a new and of disease progression, and resemble stages in gliomas located within eloquent brain regions:
simplified concept of gliomas. Proc Staff Meet Mayo neurogenesis. Cancer Cell. 2006;9:157–173. functional results in a consecutive series of 103
Clin. 1949;24:53–83. 237. Brennan CW, et al. The somatic genomic landscape patients. J Neurosurg. 2003;98:764–778.
214. Ringertz N. Grading of gliomas. APMIS. 1950;27: of glioblastoma. Cell. 2013;155:462–477. 260. Chang EF, et al. Functional mapping–guided resec-
51–64. 238. Van Meir EG, et al. Exciting new advances in tion of low-grade gliomas in eloquent areas of the
215. Daumas-Duport C, Scheithauer B, O’Fallon J, neuro-oncology: the avenue to a cure for malignant brain: improvement of long-term survival: clinical
Kelly P. Grading of astrocytomas. A simple and glioma. CA Cancer J Clin. 2010;60:166–193. article. J Neurosurg. 2011;114:566–573.
reproducible method. Cancer. 1988;62:2152–2165. 239. Verhaak RG, et al. Integrated genomic analysis 261. Chang SM, et al. Perioperative complications
216. Louis DN, et al. The 2016 World Health Orga- identifies clinically relevant subtypes of glioblastoma and neurological outcomes of first and second
nization classification of tumors of the central characterized by abnormalities in PDGFRA, IDH1, craniotomies among patients enrolled in the Glioma
nervous system: a summary. Acta Neuropathol. EGFR, and NF1. Cancer Cell. 2010;17:98–110. Outcome Project. J Neurosurg. 2003;98:1175–
2016;131:803–820. 240. Ichimura K, Schmidt EE, Goike HM, Collins VP. 1181.
217. Burns D, Kumar V. The nervous system. In: Kumar Human glioblastomas with no alterations of the 262. Leighton C, et al. Supratentorial low-grade glioma in
V, Cotrin R, Collins T, eds. Robbins Basic Pathology. CDKN2A (p16INK4A, MTS1) and CDK4 genes adults: an analysis of prognostic factors and timing
WB Saunders; 2003:832–836. have frequent mutations of the retinoblastoma gene. of radiation. J Clin Oncol. 1997;15:1294–1301.
218. Maher E, McKee A. Neoplasms of the central nervous Oncogene. 1996;13:1065–1072. 263. Karim AB, et al. A randomized trial on dose-response
system. In: Skarin A, ed. Dana-Farber Cancer Institute 241. Ichimura K, Ohgaki H, Kleihues P, Collins VP. in radiation therapy of low-grade cerebral glioma:
Atlas of Diagnostic Oncology. Mosby; 2003:395–434. Molecular pathogenesis of astrocytic tumours. European Organization for Research and Treatment
219. Nelson JS, et al. Necrosis as a prognostic criterion in J Neurooncol. 2004;70:137–160. of Cancer (EORTC) Study 22844. Int J Radiat
malignant supratentorial, astrocytic gliomas. Cancer. 242. Ichimura K, et al. Deregulation of the p14ARF/ Oncol Biol Phys. 1996;36:549–556.
1983;52:550–554. MDM2/p53 pathway is a prerequisite for human 264. van den Bent MJ, et al. Long-term efficacy of early
220. McKeever PE, et al. A comparison of the predictive astrocytic gliomas with G1-S transition control gene versus delayed radiotherapy for low-grade astrocy-
power for survival in gliomas provided by MIB-1, abnormalities. Cancer Res. 2000;60:417–424. toma and oligodendroglioma in adults: the EORTC
bromodeoxyuridine and proliferating cell nuclear 243. Ekstrand AJ, et al. Functional characterization of 22845 randomised trial. Lancet. 2005;366:985–990.
antigen with histopathologic and clinical parameters. an EGF receptor with a truncated extracellular 265. Pignatti F, et al. Prognostic factors for survival in
J Neuropathol Exp Neurol. 1997;56:798–805. domain expressed in glioblastomas with EGFR adult patients with cerebral low-grade glioma. J Clin
221. Shaw EG, Scheithauer BW, O’Fallon JR, Tazelaar gene amplification. Oncogene. 1994;9:2313–2320. Oncol. 2002;20:2076–2084.
HD, Davis DH. Oligodendrogliomas: the Mayo 244. Nishikawa R, et al. A mutant epidermal growth 266. Walker MD, et al. Evaluation of BCNU and/or
clinic experience. J Neurosurg. 1992;76:428–434. factor receptor common in human glioma confers radiotherapy in the treatment of anaplastic gliomas:
Cancer of the Central Nervous System • CHAPTER 63 967.e5
967.e5

a cooperative clinical trial. J Neurosurg. 1978;49: 283. Green SB, et al. Comparisons of carmustine, procar- 303. Cairncross G, et al. Chemotherapy for anaplastic
333–343. bazine, and high-dose methylprednisolone as addi- oligodendroglioma. National Cancer Institute
267. Kristiansen K, et al. Combined modality therapy tions to surgery and radiotherapy for the treatment of Canada Clinical Trials Group. J Clin Oncol.
of operated astrocytomas grade III and IV. of malignant glioma. Cancer Treat Rep. 1983;67: 1994;12:2013–2021.
Confirmation of the value of postoperative irra- 121–132. 304. van den Bent MJ, et al. Adjuvant procarbazine,
diation and lack of potentiation of bleomycin on 284. Dropcho EJ. Novel chemotherapeutic approaches lomustine, and vincristine chemotherapy in newly
survival time: a prospective multicenter trial of the to brain tumors. Hematol Oncol Clin North Am. diagnosed anaplastic oligodendroglioma: long-term
Scandinavian Glioblastoma Study Group. Cancer. 2001;15:1027–1052. follow-up of EORTC brain tumor group study
1981;47:649–652. 285. Stupp R, et al. Radiotherapy plus concomitant and 26951. J Clin Oncol. 2012;31:344–350.
268. Walker MD, Strike TA, Sheline GE. An analysis of adjuvant temozolomide for glioblastoma. NEJM. 305. Cairncross G, et al. Phase III trial of chemotherapy
dose-effect relationship in the radiotherapy of malig- 2005;352:987–996. plus radiotherapy compared with radiotherapy alone
nant gliomas. Int J Radiat Oncol Biol Phys. 1979;5: 286. Stupp R, et al. Effects of radiotherapy with concomi- for pure and mixed anaplastic oligodendroglioma:
1725–1731. tant and adjuvant temozolomide versus radiotherapy Intergroup Radiation Therapy Oncology Group Trial
269. Curran WJ, et al. Recursive partitioning analysis alone on survival in glioblastoma in a randomised 9402. J Clin Oncol. 2006;24:2707–2714.
of prognostic factors in three Radiation Therapy phase III study: 5-year analysis of the EORTC-NCIC 306. Cairncross G, et al. Phase III trial of chemoradio-
Oncology Group malignant glioma trials. J Natl trial. Lancet Oncol. 2009;10:459–466. therapy for anaplastic oligodendroglioma: long-term
Cancer Inst. 1993;85:704–710. 287. Brandes AA, et al. O(6)-methylguanine DNA- results of RTOG 9402. J Clin Oncol. 2013;31:
270. Mirimanoff RO, et al. Radiotherapy and temozolo- methyltransferase methylation status can change 337–343.
mide for newly diagnosed glioblastoma: recursive between first surgery for newly diagnosed glioblas- 307. Chinot O. Chemotherapy for the treatment of oligo-
partitioning analysis of the EORTC 26981/22981- toma and second surgery for recurrence: clinical dendroglial tumors. Semin Oncol. Elsevier, 2001;28:
NCIC CE3 phase III randomized trial. J Clin Oncol. implications. Neuro Oncol. 2010;12:283–288. 13–18.
2006;24:2563–2569. 288. Deleted in review. 308. van den Bent MJ, et al. Temozolomide chemotherapy
271. Li J, et al. Validation and simplification of the Radia- 289. Brem H, et al. Placebo-controlled trial of safety in recurrent oligodendroglioma. Neurology. 2001;57:
tion Therapy Oncology Group recursive partitioning and efficacy of intraoperative controlled delivery 340–342.
analysis classification for glioblastoma. Int J Radiat by biodegradable polymers of chemotherapy for 309. Brandes AA, et al. Correlations between
Oncol Biol Phys. 2011;81:623–630. recurrent gliomas. The Polymer-brain Tumor O6-methylguanine DNA methyltransferase pro-
272. Chang CH, et al. Comparison of postoperative Treatment Group. Lancet. 1995;345:1008–1012. moter methylation status, 1p and 19q deletions,
radiotherapy and combined postoperative radio- 290. Preusser M, et al. Current concepts and management and response to temozolomide in anaplastic and
therapy and chemotherapy in the multidisciplinary of glioblastoma. Ann Neurol. 2011;70:9–21. recurrent oligodendroglioma: a prospective GICNO
management of malignant gliomas. A joint Radiation 291. van den Bent MJ, et al. Randomized phase II trial study. J Clin Oncol. 2006;24:4746–4753.
Therapy Oncology Group and Eastern Cooperative of erlotinib versus temozolomide or carmustine in 310. Nayak L, Iwamoto FM. Primary brain tumors in
Oncology Group study. Cancer. 1983;52:997– recurrent glioblastoma: EORTC brain tumor group the elderly. Curr Neurol Neurosci Rep. 2010;10:252–
1007. study 26034. J Clin Oncol. 2009;27:1268–1274. 258.
273. Deutsch M, et al. Results of a randomized trial 292. Friedman HS, et al. Bevacizumab alone and in com- 311. Brandes AA, et al. A prospective study on glioblas-
comparing BCNU plus radiotherapy, streptozotocin bination with irinotecan in recurrent glioblastoma. toma in the elderly. Cancer. 2003;97:657–662.
plus radiotherapy, BCNU plus hyperfractionated J Clin Oncol. 2009;27:4733–4740. 312. Vuorinen V, Hinkka S, Färkkilä M, Jääskeläinen J.
radiotherapy, and BCNU following misonidazole 293. Kreisl TN, et al. Phase II trial of single-agent beva- Debulking or biopsy of malignant glioma in elderly
plus radiotherapy in the postoperative treatment cizumab followed by bevacizumab plus irinotecan people–a randomised study. Acta Neurochir (Wien).
of malignant glioma. Int J Radiat Oncol Biol Phys. at tumor progression in recurrent glioblastoma. 2003;145:5–10.
1989;16:1389–1396. J Clin Oncol. 2008;27:740–745. 313. Keime-Guibert F, et al. Radiotherapy for glioblastoma
274. Scott CB, et al. Validation and predictive power 294. Gilbert MR, et al. A randomized trial of bevaci- in the elderly. NEJM. 2007;356:1527–1535.
of Radiation Therapy Oncology Group (RTOG) zumab for newly diagnosed glioblastoma. NEJM. 314. Malmstrom A, et al. Glioblastoma (GBM) in elderly
recursive partitioning analysis classes for malignant 2014;370:699–708. patients: a randomized phase III trial comparing
glioma patients: a report using RTOG 90-06. Int J 295. Wick W, Weller M, van den Bent M, Stupp R. survival in patients treated with 6-week radiotherapy
Radiat Oncol Biol Phys. 1998;40:51–55. Bevacizumab and recurrent malignant gliomas: a (RT) versus hypofractionated RT over 2 weeks versus
275. Prados MD, et al. Interstitial brachytherapy for European perspective. J Clin Oncol. 2010;28:e188– temozolomide single-agent chemotherapy (TMZ).
newly diagnosed patients with malignant gliomas: e189. J Clin Oncol. 2010;28:LBA2002–LBA2002.
the UCSF experience. Int J Radiat Oncol Biol Phys. 296. Reijneveld JC, Sitskoorn MM, Klein M, Nuyen J, 315. Wick W, et al. NOA-08 randomized phase III
1992;24:593–597. Taphoorn MJB. Cognitive status and quality of life trial of 1-week-on/1-week-off temozolomide versus
276. Selker RG, et al. The Brain Tumor Cooperative in patients with suspected versus proven low-grade involved-field radiotherapy in elderly (older than
Group NIH Trial 87-01: a randomized comparison gliomas. Neurology. 2001;56:618–623. age 65) patients with newly diagnosed anaplastic
of surgery, external radiotherapy, and carmustine 297. Buckner JC, et al. Radiation plus procarbazine, astrocytoma or glioblastoma (Methusalem). J Clin
versus surgery, interstitial radiotherapy boost, external CCNU, and vincristine in low-grade glioma. NEJM. Oncol. 2010;28:LBA2001–LBA2001.
radiation therapy, and carmustine. Neurosurgery. 2016;374:1344–1355. 316. Iwamoto FM, Cooper AR, Reiner AS, Nayak L,
2002;51:343–357. 298. Baumert BG, et al. Temozolomide chemotherapy Abrey LE. Glioblastoma in the elderly: the Memorial
277. Souhami L, et al. Randomized prospective com- versus radiotherapy in high-risk low-grade glioma Sloan-Kettering Cancer Center Experience (1997-
parison of stereotactic radiosurgery (SRS) followed (EORTC 22033-26033): a randomised, open- 2007). Cancer. 2009;115:3758–3766.
by conventional radiotherapy (RT) with BCNU to label, phase 3 intergroup study. Lancet Oncol. 317. Roa W, et al. Abbreviated course of radiation therapy
RT with BCNU alone for selected patients with 2016;17:1521–1532. in older patients with glioblastoma multiforme: a
supratentorial glioblastoma multiforme (GBM): 299. van den Bent MJ, et al. Second-line chemotherapy prospective randomized clinical trial. J Clin Oncol.
report of RTOG 93-05 protocol. Int J Radiat Oncol with temozolomide in recurrent oligodendroglioma 2004;22:1583–1588.
Biol Phys. 2002;54:94–95. after PCV (procarbazine, lomustine and vincristine) 318. Laperriere N, et al. Optimal management of
278. Stupp R, et al. Maintenance therapy with tumor- chemotherapy: EORTC Brain Tumor Group phase elderly patients with glioblastoma. Cancer Treat
treating fields plus temozolomide vs temozolomide II study 26972. Ann Oncol. 2003;14:599–602. Rev. 2013;39:350–357.
alone for glioblastoma: a randomized clinical trial. 300. van den Bent MJ, et al. Phase II study of first-line 319. Triebel KL, Martin RC, Nabors LB, Marson DC.
JAMA. 2015;314:2535–2543. chemotherapy with temozolomide in recurrent Medical decision-making capacity in patients with
279. Kirson ED, et al. Alternating electric fields arrest cell oligodendroglial tumors: the European Organization malignant glioma. Neurology. 2009;73:2086–2092.
proliferation in animal tumor models and human for Research and Treatment of Cancer Brain Tumor 320. Lobb EA, Halkett GKB, Nowak AK. Patient
brain tumors. Proc Natl Acad Sci USA. 2007;104: Group Study 26971. J Clin Oncol. 2003;21:2525– and caregiver perceptions of communication of
10152–10157. 2528. prognosis in high grade glioma. J Neurooncol.
280. Kirson ED, et al. Disruption of cancer cell rep- 301. Kouwenhoven MCM, et al. 1p/19q loss within 2011;104:315–322.
lication by alternating electric fields. Cancer Res. oligodendroglioma is predictive for response to first 321. Kiebert GM, et al. Quality of life after radiation
2004;64:3288–3295. line temozolomide but not to salvage treatment. Eur therapy of cerebral low-grade gliomas of the adult:
281. Sampson JH. Alternating electric fields for the treat- J Cancer. 2006;42:2499–2503. results of a randomised phase III trial on dose response
ment of glioblastoma. JAMA. 2015;314:2511–2513. 302. Hoang-Xuan K, et al. Temozolomide as initial (EORTC trial 22844). Eur J Cancer. 1998;34:
282. Walker MD, et al. Randomized comparisons of treatment for adults with low-grade oligodendro- 1902–1909.
radiotherapy and nitrosoureas for the treatment of gliomas or oligoastrocytomas and correlation with 322. Taphoorn MJ, et al. Health-related quality of life in
malignant glioma after surgery. NEJM. 1980;303: chromosome 1p deletions. J Clin Oncol. 2004;22: patients with glioblastoma: a randomised controlled
1323–1329. 3133–3138. trial. Lancet Oncol. 2005;6:937–944.
967.e6 Part III: Specific Malignancies

323. Heimans JJ, Taphoorn MJ. Impact of brain tumour 343. Weller M, et al. Vaccine-based immunotherapeutic 366. Schroers R, et al. Detection of free immunoglobulin
treatment on quality of life. J Neurol. 2002;249: approaches to gliomas and beyond. Nat Rev Neurol. light chains in cerebrospinal fluids of patients with
955–960. 2017;13:363–374. central nervous system lymphomas. Eur J Haematol.
324. Pace A, et al. End of life issues in brain tumor 344. Brown CE, et al. Regression of glioblastoma after 2010;85:236–242.
patients. J Neurooncol. 2009;91:39–43. chimeric antigen receptor T-cell therapy. NEJM. 367. Baraniskin A, et al. Identification of microRNAs in
325. Yamanaka R, Saya H. Molecularly targeted therapies 2016;375:2561–2569. the cerebrospinal fluid as marker for primary diffuse
for glioma. Ann Neurol. 2009;66:717–729. 345. Mathios D, et al. Anti-PD-1 antitumor immunity is large B-cell lymphoma of the central nervous system.
326. Sampson JH, et al. Immunologic escape after enhanced by local and abrogated by systemic chemo- Blood. 2011;117:3140–3146.
prolonged progression-free survival with epidermal therapy in GBM. Sci Transl Med. 2016;8:370ra180. 368. Baraniskin A, Deckert M, Schulte-Altedorneburg G,
growth factor receptor variant III peptide vaccination 346. Topalian SL, et al. Safety, activity, and immune Schlegel U, Schroers R. Current strategies in the diag-
in patients with newly diagnosed glioblastoma. correlates of anti-PD-1 antibody in cancer. N Engl nosis of diffuse large B-cell lymphoma of the central
J Clin Oncol. 2010;28:4722–4729. J Med. 2012;366:2443–2454. nervous system. Br J Haematol. 2012;156:421–
327. Mellinghoff IK, et al. Molecular determinants of the 347. Shrieve DC, et al. Hyperfractionated radiation 432.
response of glioblastomas to EGFR kinase inhibitors. therapy for gliomas of the brainstem in children 369. Porter AB, et al. Primary central nervous system
NEJM. 2005;353:2012–2024. and in adults. Int J Radiat Oncol Biol Phys. 1992;24: lymphoma can be histologically diagnosed after
328. Takebe N, Harris PJ, Warren RQ, Ivy SP. Targeting 599–610. previous corticosteroid use: a pilot study to determine
cancer stem cells by inhibiting Wnt, Notch, and 348. Guillamo JS, et al. Brainstem gliomas in adults: whether corticosteroids prevent the diagnosis of
Hedgehog pathways. Nat Rev Clin Oncol. 2011;8: prognostic factors and classification. Brain. 2001;124: primary central nervous system lymphoma. Ann
97–106. 2528–2539. Neurol. 2008;63:662–667.
329. Hadjipanayis CG, Van Meir EG. Brain cancer 349. Landolfi JC, Thaler HT, DeAngelis LM. Adult 370. Mathew BS, Carson KA, Grossman SA. Initial
propagating cells: biology, genetics and targeted brainstem gliomas. Neurology. 1998;51:1136–1139. response to glucocorticoids. Cancer. 2006;106:383–
therapies. Trends Mol Med. 2009;15:519–530. 350. Montesinos-Rongen M, et al. Gene expression 387.
330. Chamberlain MC, Cloughsey T, Reardon DA, Wen profiling suggests primary central nervous system 371. Braaten KM, et al. BCL-6 expression predicts improved
PY. A novel treatment for glioblastoma: integrin lymphomas to be derived from a late germinal center survival in patients with primary central nervous
inhibition. Expert Rev Neurother. 2012;12:421– B cell. Leukemia. 2008;22:400–405. system lymphoma. Clin Cancer Res. 2003;9:1063–
435. 351. Deckert M, et al. Modern concepts in the biology, 1069.
331. Nabors LB, et al. A safety run-in and random- diagnosis, differential diagnosis and treatment 372. Chang CC, et al. Expression of p53, c-Myc, or
ized phase 2 study of cilengitide combined with of primary central nervous system lymphoma. Bcl-6 suggests a poor prognosis in primary central
chemoradiation for newly diagnosed glioblastoma Leukemia. 2011;25:1797–1807. nervous system diffuse large B-cell lymphoma among
(NABTT 0306). Cancer. 2012;118:5601–5607. 352. Gerstner ER, Batchelor TT. Primary central nervous immunocompetent individuals. Arch Pathol Lab Med.
332. Vredenburgh JJ, et al. Phase II trial of bevacizumab system lymphoma. In: Primary Central Nervous 2003;127:208–212.
and irinotecan in recurrent malignant glioma. Clin System Tumors. Springer; 2011:333–353. 373. Abrey LE, et al. Primary central nervous system lym-
Cancer Res. 2007;13:1253–1259. 353. Zacharia TT, Law M, Naidich TP, Leeds NE. Central phoma: the Memorial Sloan-Kettering Cancer Center
333. Ram Z, et al. Therapy of malignant brain tumors nervous system lymphoma characterization by prognostic model. J Clin Oncol. 2006;24:5711–5715.
by intratumoral implantation of retroviral vector- diffusion-weighted imaging and MR spectroscopy. 374. Damek DM. Primary central nervous system
producing cells. Nat Med. 1997;3:1354–1361. J Neuroimaging. 2008;18:411–417. lymphoma. Curr Treat Options Neurol. 2003;5:213–
334. Palu G, et al. Gene therapy of glioblastoma 354. Lai R, Rosenblum MK, DeAngelis LM. Primary 222.
multiforme via combined expression of suicide CNS lymphoma: a whole-brain disease? Neurology. 375. Ling SM, Iii MR, Larson DA, Wara WM.
and cytokine genes: a pilot study in humans. Gene 2002;59:1557–1562. Radiotherapy of primary central nervous system
Ther. 1999;6:330–337. 355. Maher EA, Fine HA. Primary CNS lymphoma. lymphoma in patients with and without human
335. Shand N, et al. A phase 1-2 clinical trial of gene Semin Oncol. 1999;26:346–356. immunodeficiency virus. Ten years of treatment
therapy for recurrent glioblastoma multiforme 356. Ciacci JD, Tellez C, VonRoenn J, Levy RM. experience at the University of California San
by tumor transduction with the herpes simplex Lymphoma of the central nervous system in AIDS. Francisco. Cancer. 1994;73:2570–2582.
thymidine kinase gene followed by ganciclovir. Semin Neurol. \copyright 1999 by Thieme Medical 376. Nelson DF, et al. Non-Hodgkin’s lymphoma of
Hum Gene Ther. 1999;10:2325–2335. Publishers, Inc., 1999;19:213–221. the brain: can high dose, large volume radiation
336. Rainov NG. A phase III clinical evaluation of 357. Kadan-Lottick NS, Skluzacek MC, Gurney JG. therapy improve survival? Report on a prospective
herpes simplex virus type 1 thymidine kinase and Decreasing incidence rates of primary central nervous trial by the Radiation Therapy Oncology Group
ganciclovir gene therapy as an adjuvant to surgical system lymphoma. Cancer. 2002;95:193–202. (RTOG): RTOG 8315. Int J Radiat Oncol Biol Phys.
resection and radiation in adults with previously 358. Chamberlain MC, Kormanik PA. AIDS-related 1992;23:9–17.
untreated glioblastoma multiforme. Hum Gene Ther. central nervous system lymphomas. J Neurooncol. 377. DeAngelis LM, Seiferheld W, Schold SC, Fisher
2000;11:2389–2401. 1999;43:269–276. B, Schultz CJ. Combination chemotherapy and
337. Sotelo J, Briceno E, López-González MA. Adding 359. Ferreri AJ, Assanelli A, Crocchiolo R, Ciceri F. radiotherapy for primary central nervous system
chloroquine to conventional treatment for glio- Central nervous system dissemination in immu- lymphoma: Radiation Therapy Oncology Group
blastoma multiforme: a randomized, double-blind, nocompetent patients with aggressive lymphomas: Study 93-10. J Clin Oncol. 2002;20:4643–4648.
placebo-controlled trial. Ann Intern Med. 2006;144: incidence, risk factors and therapeutic options. 378. Glass J, Gruber ML, Cher L, Hochberg FH. Preir-
337–343. Hematol Oncol. 2009;27:61–70. radiation methotrexate chemotherapy of primary
338. Liu Y, et al. TGF-β2 inhibition augments the effect of 360. Haldorsen IS, Espeland A, Larsen JL, Mella O. central nervous system lymphoma: long-term
tumor vaccine and improves the survival of animals Diagnostic delay in primary central nervous system outcome. J Neurosurg. 1994;81:188–195.
with pre-established brain tumors. J Neurooncol. lymphoma. Acta Oncol (Madr). 2005;44:728–734. 379. Keime-Guibert F, Napolitano M, Delattre JY.
2007;81:149–162. 361. Mohile NA, DeAngelis LM, Abrey LE. The utility of Neurological complications of radiotherapy and
339. Mintz A, Gibo DM, Madhankumar AB, Debinski body FDG PET in staging primary central nervous chemotherapy. J Neurol. 1998;245:695–708.
W. Molecular targeting with recombinant cytotoxins system lymphoma. Neuro Oncol. 2008;10:223– 380. Sarazin M, et al. Primary central nervous system
of interleukin-13 receptor α 2-expressing glioma. 228. lymphoma: treatment with chemotherapy and
J Neurooncol. 2003;64:117–123. 362. Abrey LE, et al. Report of an international workshop radiotherapy. Eur J Cancer. 1995;31:2003–2007.
340. Dorsey JF, et al. Tumor necrosis factor–related to standardize baseline evaluation and response 381. Correa DD, et al. Cognitive functions in survivors
apoptosis-inducing ligand (TRAIL) and paclitaxel criteria for primary CNS lymphoma. J Clin Oncol. of primary central nervous system lymphoma.
have cooperative in vivo effects against glioblas- 2005;23:5034–5043. Neurology. 2004;62:548–555.
toma multiforme cells. Mol Cancer Ther. 2009;8: 363. Quijano S, et al. Identification of leptomeningeal 382. Cher L, Glass J, Harsh GR, Hochberg FH. Therapy
3285–3295. disease in aggressive B-cell non-Hodgkin’s lymphoma: of primary CNS lymphoma with methotrexate-based
341. Tian X, et al. Modulation of CCAAT/enhancer improved sensitivity of flow cytometry. J Clin Oncol. chemotherapy and deferred radiotherapy: preliminary
binding protein homologous protein (CHOP) – 2009;27:1462–1469. results. Neurology. 1996;46:1757–1759.
dependent DR5 expression by nelfinavir sensitizes 364. Gleissner B, Siehl J, Korfel A, Reinhardt R, Thiel E. 383. Gerstner ER, Carson KA, Grossman SA, Batchelor
glioblastoma multiforme cells to tumor necrosis CSF evaluation in primary CNS lymphoma patients by TT. Long-term outcome in PCNSL patients treated
factor-related apoptosis-inducing ligand (TRAIL). PCR of the CDR III IgH genes. Neurology. 2002;58: with high-dose methotrexate and deferred radiation.
J Biol Chem. 2011;286:29408–29416. 390–396. Neurology. 2008;70:401–402.
342. O’Rourke DM, et al. Pilot study of T cells redirected 365. Kros JM, et al. Analysis of immunoglobulin H 384. Glantz MJ, et al. High-dose intravenous methotrexate
to EGFRvIII with a chimeric antigen receptor in gene rearrangement by polymerase chain reaction for patients with nonleukemic leptomeningeal cancer:
patients with EGFRvIII+ glioblastoma. JCO. 2016;34: in primary central nervous system lymphoma. is intrathecal chemotherapy necessary? J Clin Oncol.
2067–2067. J Neurosurg. 2002;97:1390–1396. 1998;16:1561–1567.
Cancer of the Central Nervous System • CHAPTER 63 967.e7
967.e7

385. Dahlborg SA, et al. Non-AIDS primary CNS 405. Hall WA, Luciano MG, Doppman JL, Patronas 427. Andrews DW, et al. Fractionated stereotactic
lymphoma: first example of a durable response in NJ, Oldfield EH. Pituitary magnetic resonance radiotherapy for the treatment of optic nerve
a primary brain tumor using enhanced chemotherapy imaging in normal human volunteers: occult sheath meningiomas: preliminary observations
delivery without cognitive loss and without radio- adenomas in the general population. Ann Intern of 33 optic nerves in 30 patients with historical
therapy. Cancer J Sci Am. 1995;2:166–174. Med. 1994;120:817–820. comparison to observation with or without prior
386. Nasir S, DeAngelis LM. Update on the management 406. Thakker RV. Multiple endocrine neoplasia. Horm surgery. Neurosurgery. 2002;51:890–902, 904.
of primary CNS lymphoma. Oncology (Williston Res. 2001;56(suppl 1):67–72. 428. Metellus P, et al. Evaluation of fractionated radio-
Park, NY). 2000;14:228–234. 407. Longstreth WT, Dennis LK, McGuire VM, therapy and gamma knife radiosurgery in cavernous
387. Graber JJ, Omuro A. Primary central nervous system Drangsholt MT, Koepsell TD. Epidemiology of sinus meningiomas: treatment strategy. Neurosurgery.
lymphoma: is there still a role for radiotherapy? Curr intracranial meningioma. Cancer. 1993;72:639–648. 2005;57:873–886, 886.
Opin Neurol. 2011;24:633–640. 408. Helseth A, Mørk SJ, Glattre E. Neoplasms of the 429. A Trial of Pembrolizumab for Refractory Atypical
388. Thiel E, et al. High-dose methotrexate with or central nervous system in Norway. V. Meningioma and Anaplastic Meningioma. ClinicalTrials.gov.
without whole brain radiotherapy for primary and cancer of other sites. An analysis of the occur- Available at: https://clinicaltrials.gov/ct2/show/
CNS lymphoma (G-PCNSL-SG-1): a phase 3, rence of multiple primary neoplasms in meningioma NCT03016091. Accessed: June 13, 2017.
randomised, non-inferiority trial. Lancet Oncol. patients in Norway from 1955 through 1986. 430. Arita K, et al. Natural course of incidentally found
2010;11:1036–1047. APMIS. 1989;97:738–744. nonfunctioning pituitary adenoma, with special
389. Omuro A, et al. Primary CNS lymphoma in patients 409. Evans DG, et al. A genetic study of type 2 reference to pituitary apoplexy during follow-up
younger than 60: can whole-brain radiotherapy be neurofibromatosis in the United Kingdom. II. examination. J Neurosurg. 2006;104:884–891.
deferred? J Neurooncol. 2011;104:323–330. Guidelines for genetic counselling. J Med Genet. 431. Dubuisson AS, Beckers A, Stevenaert A. Classical
390. Freilich RJ, Delattre JY, Monjour A, DeAngelis 1992;29:847–852. pituitary tumour apoplexy: clinical features, manage-
LM. Chemotherapy without radiation therapy as 410. Ruttledge MH, et al. Evidence for the complete ment and outcomes in a series of 24 patients. Clin
initial treatment for primary CNS lymphoma in inactivation of the NF2 gene in the majority of Neurol Neurosurg. 2007;109:63–70.
older patients. Neurology. 1996;46:435–439. sporadic meningiomas. Nat Genet. 1994;6:180–184. 432. Semple PL, Webb MK, de Villiers JC, Laws ER.
391. Kim L, Hochberg FH, Shaeffer P. White-matter 411. Clark VE, et al. Genomic analysis of non-NF2 Pituitary apoplexy. Neurosurgery. 2005;56:65–72,
abnormalities in unirradiated patients cured of meningiomas reveals mutations in TRAF7, KLF4, 73.
primary central nervous system lymphoma. Neu- AKT1, and SMO. Science. 2013;339:1077–1080. 433. Nielsen EH, et al. Frequent occurrence of pituitary
roradiology. 2000;42:406–412. 412. Clark VE, et al. Recurrent somatic mutations in apoplexy in patients with non-functioning pituitary
392. Fliessbach K, et al. Cognitive performance and POLR2A define a distinct subset of meningiomas. adenoma. Clin Endocrinol (Oxf). 2006;64:319–322.
magnetic resonance imaging findings after high- Nat Genet. 2016;48:1253–1259. 434. Oruçkaptan HH, Senmevsim Ö, Özcan OE, Özgen
dose systemic and intraventricular chemotherapy 413. McCarthy BJ, et al. Factors associated with sur- T. Pituitary adenomas: results of 684 surgically
for primary central nervous system lymphoma. Arch vival in patients with meningioma. J Neurosurg. treated patients and review of the literature. Surg
Neurol. 2003;60:563–568. 1998;88:831–839. Neurol. 2000;53:211–219.
393. Zhu JJ, et al. High-dose methotrexate for elderly 414. Cahill KS, Claus EB. Treatment and survival of 435. Fernandez A, Karavitaki N, Wass JAH. Prevalence
patients with primary CNS lymphoma. Neuro Oncol. patients with nonmalignant intracranial meningioma: of pituitary adenomas: a community-based, cross-
2009;11:211–215. results from the Surveillance, Epidemiology, and End sectional study in Banbury (Oxfordshire, UK). Clin
394. Omuro AM, et al. Temozolomide and methotrexate Results Program of the National Cancer Institute. Endocrinol (Oxf ). 2010;72:377–382.
for primary central nervous system lymphoma in Clinical article. J Neurosurg. 2011;115:259–267. 436. Daly AF, et al. High prevalence of pituitary
the elderly. J Neurooncol. 2007;85:207–211. 415. Mirimanoff RO, Dosoretz DE, Linggood RM, adenomas: a cross-sectional study in the province
395. Fliessbach K, et al. Neuropsychological outcome Ojemann RG, Martuza RL. Meningioma: analysis of Liege, Belgium. J Clin Endocrinol Metab. 2006;91:
after chemotherapy for primary CNS lymphoma of recurrence and progression following neurosurgical 4769–4775.
A prospective study. Neurology. 2005;64:1184– resection. J Neurosurg. 1985;62:18–24. 437. Howlett TA, et al. Megavoltage pituitary irradiation
1188. 416. Miralbell R, et al. The role of radiotherapy in the in the management of Cushing’s disease and Nelson’s
396. Harder H, et al. Cognitive status and quality of treatment of subtotally resected benign meningiomas. syndrome: long-term follow-up. Clin Endocrinol
life after treatment for primary CNS lymphoma. J Neurooncol. 1992;13:157–164. (Oxf ). 1989;31:309–323.
Neurology. 2004;62:544–547. 417. Goldsmith BJ, Wara WM, Wilson CB, Larson DA. 438. Lloyd RV. Molecular pathology of pituitary adeno-
397. Lai R, Abrey LE, Rosenblum MK, DeAngelis Postoperative irradiation for subtotally resected mas. J Neurooncol. 2001;54:111–119.
LM. Treatment-induced leukoencephalopathy in meningiomas. A retrospective analysis of 140 patients 439. Hardy J. Transphenoidal microsurgery of the
primary CNS lymphoma: a clinical and autopsy treated from 1967 to 1990. J Neurosurg. 1994;80: normal and pathological pituitary. Clin Neurosurg.
study. Neurology. 2004;62:451–456. 195–201. 1969;16:185–217.
398. Slobod KS, et al. Epstein-Barr virus-targeted therapy 418. Kondziolka D, Levy EI, Niranjan A, Flickinger 440. Ciric I, Ragin A, Baumgartner C, Pierce D.
for AIDS-related primary lymphoma of the central JC, Lunsford LD. Long-term outcomes after Complications of transsphenoidal surgery: results
nervous system. Lancet. 2000;356:1493–1494. meningioma radiosurgery: physician and patient of a national survey, review of the literature, and
399. Ferreri AJM, et al. Chemoimmunotherapy with perspectives. J Neurosurg. 1999;91:44–50. personal experience. Neurosurgery. 1997;40:225–236,
methotrexate, cytarabine, thiotepa, and rituximab 419. Stafford SL, et al. Meningioma radiosurgery: 237.
(MATRix regimen) in patients with primary CNS tumor control, outcomes, and complications 441. Jho HD. Endoscopic transsphenoidal surgery. J
lymphoma: results of the first randomisation of among 190 consecutive patients. Neurosurgery. Neurooncol. 2001;54:187–195.
the International Extranodal Lymphoma Study 2001;49:1029–1037, 1038. 442. Kabil MS, Eby JB, Shahinian HK. Fully endoscopic
Group-32 (IELSG32) phase 2 trial. Lancet Haematol. 420. Kollová A, et al. Gamma Knife surgery for benign endonasal vs. transseptal transsphenoidal pituitary
2016;3:e217–e227. meningioma. J Neurosurg. 2007;107:325–336. surgery. Minim Invasive Neurosurg. 2005;48:348–
400. Mathew BS, Grossman SA. Pneumocystis carinii 421. Bledsoe JM, Link MJ, Stafford SL, Park PJ, Pollock 354.
pneumonia prophylaxis in HIV negative patients BE. Radiosurgery for large-volume (> 10 cm3) benign 443. Fatemi N, Dusick JR, de Paiva Neto MA, Kelly DF.
with primary CNS lymphoma. Cancer Treat Rev. meningiomas. J Neurosurg. 2010;112:951–956. The endonasal microscopic approach for pituitary
2003;29:105–119. 422. O’Sullivan MG, van Loveren HR, Tew JM. The adenomas and other parasellar tumors: a 10-year
401. Harjunpää A, et al. Complement activation in cir- surgical resectability of meningiomas of the cavernous experience. Neurosurgery. 2008;63:244–256, discus-
culation and central nervous system after rituximab sinus. Neurosurgery. 1997;40:238–244, 247. sion 256.
(anti-CD20) treatment of B-cell lymphoma. Leuk 423. Spiegelmann R, Nissim O, Menhel J, Alezra D, 444. Dehdashti AR, Ganna A, Karabatsou K, Gentili F.
Lymphoma. 2001;42:731–738. Pfeffer MR. Linear accelerator radiosurgery for Pure endoscopic endonasal approach for pituitary
402. Pels H, et al. Intraventricular and intravenous treat- meningiomas in and around the cavernous sinus. adenomas: early surgical results in 200 patients
ment of a patient with refractory primary CNS Neurosurgery. 2002;51:1373–1379, 1380. and comparison with previous microsurgical series.
lymphoma using rituximab. J Neurooncol. 2002;59: 424. Morita A, Coffey RJ, Foote RL, Schiff D, Gorman Neurosurgery. 2008;62:1006–1017.
213–216. D. Risk of injury to cranial nerves after gamma knife 445. Paluzzi A, et al. Endoscopic endonasal approach
403. Soussain C, et al. Results of intensive chemotherapy radiosurgery for skull base meningiomas: experience for pituitary adenomas: a series of 555 patients.
followed by hematopoietic stem-cell rescue in 22 in 88 patients. J Neurosurg. 1999;90:42–49. Pituitary. 2014;17:307–319.
patients with refractory or recurrent primary CNS 425. Lee JYK, et al. Stereotactic radiosurgery provid- 446. Laws ER, Vance ML, Thapar K. Pituitary surgery
lymphoma or intraocular lymphoma. J Clin Oncol. ing long-term tumor control of cavernous sinus for the management of acromegaly. Horm Res.
2001;19:742–749. meningiomas. J Neurosurg. 2002;97:65–72. 2000;53(suppl 3):71–75.
404. Rohringer M, Sutherland GR, Louw DF, Sima 426. Debus J, et al. High efficacy of fractionated stereotac- 447. Lüdecke DK, Flitsch J, Knappe UJ, Saeger W. Cush-
AA. Incidence and clinicopathological features of tic radiotherapy of large base-of-skull meningiomas: ing’s disease: a surgical view. J Neurooncol. 2001;54:
meningioma. J Neurosurg. 1989;71:665–672. long-term results. J Clin Oncol. 2001;19:3547–3553. 151–166.
967.e8 Part III: Specific Malignancies

448. Sanno N, Teramoto A, Osamura RY. Thyrotropin- 473. Grigsby PW, Thomas PR, Simpson JR, Fineberg BB. 496. Koh ES, et al. Fractionated stereotactic radiotherapy
secreting pituitary adenomas. Clinical and biological Long-term results of radiotherapy in the treatment for acoustic neuroma: single-institution experi-
heterogeneity and current treatment. J Neurooncol. of pituitary adenomas in children and adolescents. ence at The Princess Margaret Hospital. Cancer.
2001;54:179–186. Am J Clin Oncol. 1988;11:607–611. 2007;109:1203–1210.
449. Losa M, et al. Proliferation index of nonfunction- 474. Mondok A, et al. Treatment of pituitary tumors: 497. Shirato H, et al. Comparison between observation
ing pituitary adenomas: correlations with clinical radiation. Endocrine. 2005;28:77–85. policy and fractionated stereotactic radiotherapy
characteristics and long-term follow-up results. 475. Minniti G, Traish D, Ashley S, Gonsalves A, Brada (SRT) as an initial management for vestibular
Neurosurgery. 2000;47:1313–1318., 1319. M. Risk of second brain tumor after conservative schwannoma. Int J Radiat Oncol Biol Phys. 1999;44:
450. Lillehei KO, Kirschman DL, Kleinschmidt- surgery and radiotherapy for pituitary adenoma: 545–550.
DeMasters BK, Ridgway EC. Reassessment of update after an additional 10 years. J Clin Endocrinol 498. Combs SE, et al. Management of acoustic neu-
the role of radiation therapy in the treatment Metab. 2005;90:800–804. romas with fractionated stereotactic radiotherapy
of endocrine-inactive pituitary macroadenomas. 476. Goel A, Muzumdar D, Raman C. Trigeminal (FSRT): long-term results in 106 patients treated
Neurosurgery. 1998;43:432–438, 439. neuroma: analysis of surgical experience with 73 in a single institution. Int J Radiat Oncol Biol Phys.
451. Woollons AC, et al. Non-functioning pituitary cases. Neurosurgery. 2003;52:783–790. 2005;63:75–81.
adenomas: indications for postoperative radiotherapy. 477. McCormick PC, Bello JA, Post KD. Trigeminal 499. Combs SE, Welzel T, Schulz-Ertner D, Huber
Clin Endocrinol (Oxf ). 2000;53:713–717. schwannoma: Surgical series of 14 cases with PE, Debus J. Differences in clinical results after
452. Losa M, Mortini P, Barzaghi R, Franzin A, review of the literature. J Neurosurg. 1988;69:850– LINAC-based single-dose radiosurgery versus
Giovanelli M. Endocrine inactive and gonadotroph 860. fractionated stereotactic radiotherapy for patients
adenomas: diagnosis and management. J Neurooncol. 478. Harner SG, Laws ER. Clinical findings in with vestibular schwannomas. Int J Radiat Oncol
2001;54:167–177. patients with acoustic neurinoma. Mayo Clin Proc. Biol Phys. 2010;76:193–200.
453. Molitch ME. Diagnosis and treatment of pituitary 1983;58:721–728. 500. Fong BM, et al. Hearing preservation after LINAC
adenomas: a review. JAMA. 2017;317:516–524. 479. Bijlsma EK, Brouwer-Mladin R, Bosch DA, radiosurgery and LINAC radiotherapy for vestibular
454. Casanueva FF, et al. Guidelines of the Pituitary Westerveld A, Hulsebos TJ. Molecular characteriza- schwannoma. J Clin Neurosci. 2012;19:1065–1070.
Society for the diagnosis and management of pro- tion of chromosome 22 deletions in schwannomas. 501. Huang CF, Kondziolka D, Flickinger JC, Lunsford
lactinomas. Clin Endocrinol (Oxf). 2006;65:265–273. Genes Chromosomes Cancer. 1992;5:201–205. LD. Stereotactic radiosurgery for trigeminal schwan-
455. Nomikos P, Buchfelder M, Fahlbusch R. Current 480. Plotkin SR, et al. Hearing improvement after nomas. Neurosurgery. 1999;45:11–16, discussion 16.
management of prolactinomas. J Neurooncol. 2001;54: bevacizumab in patients with neurofibromatosis 502. Plotkin SR, et al. Bevacizumab for progressive
139–150. type 2. NEJM. 2009;361:358–367. vestibular schwannoma in neurofibromatosis type
456. Klibanski A. Clinical practice. Prolactinomas. N 481. Jackler RK, Pitts LH. Selection of surgical approach 2: a retrospective review of 31 patients. Otol Neurotol.
Engl J Med. 2010;362:1219–1226. to acoustic neuroma. Otolaryngol Clin North Am. 2012;33:1046–1052.
457. Colao A, et al. New medical approaches in pituitary 1992;25:361–387. 503. Slusarz KM, Merker VL, Muzikansky A, Francis
adenomas. Horm Res. 2000;53(suppl 3):76–87. 482. Briggs RJ, Luxford WM, Atkins JS Jr, Hitselberger SA, Plotkin SR. Long-term toxicity of bevacizumab
458. Manjila S, et al. Pharmacological management of WE. Translabyrinthine removal of large acoustic therapy in neurofibromatosis 2 patients. Cancer
acromegaly: a current perspective. Neurosurg Focus. neuromas. Neurosurgery. 1994;34:785–791. Chemother Pharmacol. 2014;73:1197–1204.
2010;29:E14. 483. Post KD, Eisenberg MB, Catalano PJ. Hearing 504. Clifford SC, Maher ER. Von Hippel-Lindau disease:
459. Melmed S. Medical progress: acromegaly. N Engl J preservation in vestibular schwannoma surgery: clinical and molecular perspectives. Adv Cancer Res.
Med. 2006;355:2558–2573. what factors influence outcome? J Neurosurg. 2001;82:85–105.
460. Giustina A, et al. Current management practices 1995;83:191–196. 505. Kaelin WG. Molecular basis of the VHL hereditary
for acromegaly: an international survey. Pituitary. 484. Shelton C, Brackmann DE, House WF, Hitselberger cancer syndrome. Nat Rev Cancer. 2002;2:673–682.
2011;14:125–133. WE. Middle fossa acoustic tumor surgery: results 506. Kanno H, et al. Somatic mutations of the von
461. Melmed S, et al. Guidelines for acromegaly in 106 cases. Laryngoscope. 1989;99:405–408. Hippel-Lindau tumor suppressor gene in sporadic
management: an update. J Clin Endocrinol Metab. 485. Wiet RJ, Teixido M, Liang JG. Complications in central nervous system hemangioblastomas. Cancer
2009;94:1509–1517. acoustic neuroma surgery. Otolaryngol Clin North Res. 1994;54:4845–4847.
462. Loeffler JS, Shih HA. Radiation therapy in the Am. 1992;25:389–412. 507. Mondkar VP, McKissock W, Russell RW. Cerebellar
management of pituitary adenomas. J Clin Endocrinol 486. Samii M, Matthies C. Management of 1000 ves- haemangioblastomas. Br J Surg. 1967;54:45–49.
Metab. 2011;96:1992–2003. tibular schwannomas (acoustic neuromas): hearing 508. Okawara SH. Solid cerebellar hemangioblastoma.
463. Sheehan JP, et al. Gamma Knife radiosurgery for the function in 1000 tumor resections. Neurosurgery. J Neurosurg. 1973;39:514–518.
management of nonfunctioning pituitary adenomas: 1997;40:248–262. 509. Smalley SR, et al. Radiotherapeutic considerations
a multicenter study. J Neurosurg. 2013;119:446– 487. Kondziolka D, Lunsford LD, McLaughlin MR, in the treatment of hemangioblastomas of the
456. Flickinger JC. Long-term outcomes after radiosurgery central nervous system. Int J Radiat Oncol Biol
464. Sheehan JP, Xu Z, Salvetti DJ, Schmitt PJ, Vance for acoustic neuromas. NEJM. 1998;339:1426–1433. Phys. 1990;18:1165–1171.
ML. Results of Gamma Knife surgery for Cushing’s 488. Foote RL, et al. Stereotactic radiosurgery using the 510. Sung DI, Chang CH, Harisiadis L. Cerebellar
disease. J Neurosurg. 2013;119:1486–1492. gamma knife for acoustic neuromas. Int J Radiat hemangioblastomas. Cancer. 1982;49:553–555.
465. Minniti G, Gilbert DC, Brada M. Modern tech- Oncol Biol Phys. 1995;32:1153–1160. 511. Koh ES, et al. Role of fractionated external beam
niques for pituitary radiotherapy. Rev Endocr Metab 489. Mendenhall WM, Friedman WA, Bova FJ. Linear radiotherapy in hemangioblastoma of the central
Disord. 2009;10:135–144. accelerator-based stereotactic radiosurgery for nervous system. Int J Radiat Oncol Biol Phys. 2007;69:
466. Landolt AM, Lomax N. Gamma Knife radiosurgery acoustic schwannomas. Int J Radiat Oncol Biol 1521–1526.
for prolactinomas. J Neurosurg. 2000;93(suppl Phys. 1994;28:803–810. 512. Patrice SJ, et al. Radiosurgery for hemangioblastoma:
3):14–18. 490. Flickinger JC, Kondziolka D, Niranjan A, Lunsford results of a multiinstitutional experience. Int J Radiat
467. Landolt AM, et al. Octreotide may act as a radio- LD. Results of acoustic neuroma radiosurgery: an Oncol Biol Phys. 1996;35:493–499.
protective agent in acromegaly. J Clin Endocrinol analysis of 5 years’ experience using current methods. 513. Asthagiri AR, et al. Prospective evaluation of
Metab. 2000;85:1287–1289. J Neurosurg. 2001;94:1–6. radiosurgery for hemangioblastomas in von Hippel–
468. McCord MW, et al. Radiotherapy for pituitary 491. Murphy ES, Suh JH. Radiotherapy for vestibular Lindau disease. Neuro Oncol. 2010;12:80–86.
adenoma: long-term outcome and sequelae. Int J schwannomas: a critical review. Int J Radiat Oncol 514. McMaster ML, Goldstein AM, Bromley CM, Ishibe
Radiat Oncol Biol Phys. 1997;39:437–444. Biol Phys. 2011;79:985–997. N, Parry DM. Chordoma: incidence and survival
469. Tsang RW, et al. Radiation therapy for pituitary 492. Friedman WA, Bradshaw P, Myers A, Bova FJ. Linear patterns in the United States, 1973–1995. Cancer
adenoma: treatment outcome and prognostic accelerator radiosurgery for vestibular schwannomas. Causes Control. 2001;12:1–11.
factors. Int J Radiat Oncol Biol Phys. 1994;30:557– J Neurosurg. 2006;105:657–661. 515. Jemal A, Siegel R, Xu J, Ward E. Cancer statistics,
565. 493. Poen JC, et al. Fractionated stereotactic radiosurgery 2010. CA Cancer J Clin. 2010;60:277–300.
470. Zierhut D, et al. External radiotherapy of and preservation of hearing in patients with vestibu- 516. Chugh R, et al. Chordoma: the nonsarcoma primary
pituitary adenomas. Int J Radiat Oncol Biol Phys. lar schwannoma: a preliminary report. Neurosurgery. bone tumor. Oncologist. 2007;12:1344–1350.
1995;33:307–314. 1999;45:1299. 517. Fuller DB, Bloom JG. Radiotherapy for chordoma.
471. Tsang RW, et al. Role of radiation therapy in clinical 494. Varlotto JM, et al. Fractionated stereotactic radio- Int J Radiat Oncol Biol Phys. 1988;15:331–339.
hormonally-active pituitary adenomas. Radiother therapy for the treatment of acoustic neuromas: 518. Rosenberg AE, et al. Chondrosarcoma of the base
Oncol. 1996;41:45–53. preliminary results. Int J Radiat Oncol Biol Phys. of the skull: a clinicopathologic study of 200 cases
472. Breen P, Flickinger JC, Kondziolka D, Martinez AJ. 1996;36:141–145. with emphasis on its distinction from chordoma.
Radiotherapy for nonfunctional pituitary adenoma: 495. Fuss M, et al. Conventionally fractionated stereotactic Am J Surg Pathol. 1999;23:1370.
analysis of long-term tumor control. J Neurosurg. radiotherapy (FSRT) for acoustic neuromas. Int J 519. Crapanzano JP, Ali SZ, Ginsberg MS, Zakowski
1998;89:933–938. Radiat Oncol Biol Phys. 2000;48:1381–1387. MF. Chordoma. Cancer. 2001;93:40–51.
Cancer of the Central Nervous System • CHAPTER 63 967.e9
967.e9

520. Fechner RE, Mills SE. Atlas of tumor pathology, 544. Bergh P, et al. Prognostic factors in chordoma of 569. Gurney JG, Severson RK, Davis S, Robison LL.
tumors of the bones and joints, fascicle 8, 3rd series. the sacrum and mobile spine. Cancer. 2000;88: Incidence of cancer in children in the United States.
Armed Forces Institute of Pathology, Washington Google 2122–2134. Sex-, race-, and 1-year age-specific rates by histologic
Scholar; 1993. 545. Solero CL, et al. Spinal meningiomas: review of 174 type. Cancer. 1995;75:2186–2195.
521. Larson TC, Houser OW, Laws ER. Imaging of cranial operated cases. Neurosurgery. 1989;25:153–160. 570. Ostrom QT, et al. Alex’s Lemonade Stand Foundation
chordomas. Mayo Clin Proc. Elsevier, 1987;62: 546. Levy WJ Jr, Bay J, Dohn D. Spinal cord meningioma. infant and childhood primary brain and central
886–893. J Neurosurg. 1982;57:804–812. nervous system tumors diagnosed in the United
522. Boriani S, et al. Chordoma of the mobile spine: 547. Dodd RL, et al. CyberKnife radiosurgery for States in 2007–2011. Neuro Oncol. 2015;16(suppl
fifty years of experience. Spine. 2006;31:493–503. benign intradural extramedullary spinal tumors. 10):x1–x36.
523. Gay E, et al. Chordomas and chondrosarcomas of Neurosurgery. 2006;58:674–685. 571. Ries LA, Kosary C, Hankey B. SEER cancer statistics
the cranial base: results and follow-up of 60 patients. 548. Epstein FJ, Farmer JP, Freed D. Adult intramedullary review, 1973-1005. The Cancer Statistics Branch,
Neurosurgery. 1995;36:887–897. spinal cord ependymomas: the result of surgery in Cancer Control Research Program, Division of Cancer
524. Keisch ME, Garcia DM, Shibuya RB. Retrospective 38 patients. J Neurosurg. 1993;79:204–209. Prevention and Control, national Cancer Institute,
long-term follow-up analysis in 21 patients with 549. Cooper PR. Outcome after operative treatment Bethesda, MD.
chordomas of various sites treated at a single institu- of intramedullary spinal cord tumors in adults: 572. Smith MA, Freidlin B, Ries LA, Simon R. Trends
tion. J Neurosurg. 1991;75:374–377. intermediate and long-term results in 51 patients. in reported incidence of primary malignant brain
525. Forsyth PA, et al. Intracranial chordomas: a clini- Neurosurgery. 1989;25:855–859. tumors in children in the United States. J Natl Cancer
copathological and prognostic study of 51 cases. J 550. Aghakhani N, et al. Intramedullary spinal ependy- Inst. 1998;90:1269–1277.
Neurosurg. 1993;78:741–747. momas: analysis of a consecutive series of 82 adult 573. Bishop AJ, McDonald MW, Chang AL, Esiashvili N.
526. Austin-Seymour M, et al. Fractionated proton cases with particular attention to patients with no Infant brain tumors: incidence, survival, and the role
radiation therapy of chordoma and low-grade preoperative neurological deficit. Neurosurgery. of radiation based on Surveillance, Epidemiology,
chondrosarcoma of the base of the skull. J Neurosurg. 2008;62:1279–1285. and End Results (SEER) Data. Int J Radiat Oncol
1989;70:13–17. 551. Quigley DG, et al. Outcome predictors in the Biol Phys. 2012;82:341–347.
527. Munzenrider JE, Liebsch NJ. Proton therapy management of spinal cord ependymoma. Eur Spine 574. Lubin F, et al. The role of nutritional habits during
for tumors of the skull base. Strahlenther Onkol. J. 2007;16:399–404. gestation and child life in pediatric brain tumor
1999;175:57–63. 552. Akyurek S, et al. Spinal myxopapillary ependymoma etiology. Int J Cancer. 2000;86:139–143.
528. Hug EB, et al. Proton radiation therapy for outcomes in patients treated with surgery and 575. Gurney JG, Smith MA, Bunin GR. CNS and Miscel-
chordomas and chondrosarcomas of the skull base. radiotherapy at MD Anderson Cancer Center. laneous Intracranial and Intraspinal Neoplasms (SEER
J Neurosurg. 1999;91:432–439. J Neurooncol. 2006;80:177–183. Pediatric Monograph). Reston, VA: National Cancer
529. Walcott BP, et al. Chordoma: current concepts, 553. Gomez DR, et al. High failure rate in spinal Institute; 2000.
management, and future directions. Lancet Oncol. ependymomas with long-term follow-up. Neuro 576. Bunin GR, Kuijten RR, Buckley JD, Rorke LB,
2012;13:e69–e76. Oncol. 2005;7:254–259. Meadows AT. Relation between maternal diet and
530. Spector GJ, Druck NS, Gado M. Neurologic 554. Isaacson SR. Radiation therapy and the management subsequent primitive neuroectodermal brain tumors
manifestations of glomus tumors in the head and of intramedullary spinal cord tumors. J Neurooncol. in young children. N Engl J Med. 1993;329:536–
neck. Arch Neurol. 1976;33:270–274. 2000;47:231–238. 541.
531. Hinerman RW, et al. Definitive radiotherapy in 555. Waldron JN, et al. Spinal cord ependymomas: a 577. Bunin GR, Gallagher PR, Rorke-Adams LB, Robison
the management of chemodectomas arising in the retrospective analysis of 59 cases. Int J Radiat Oncol LL, Cnaan A. Maternal supplement, micronutrient,
temporal bone, carotid body, and glomus vagale. Biol Phys. 1993;27:223–229. and cured meat intake during pregnancy and risk
Head Neck. 2001;23:363–371. 556. Whitaker SJ, et al. Postoperative radiotherapy in of medulloblastoma during childhood: a children’s
532. Powell S, Peters N, Harmer C. Chemodectoma the management of spinal cord ependymoma. J oncology group study. Cancer Epidemiol Biomarkers
of the head and neck: results of treatment in 84 Neurosurg. 1991;74:720–728. Prev. 2006;15:1660–1667.
patients. Int J Radiat Oncol Biol Phys. 1992;22:919– 557. Shirato H, et al. The role of radiotherapy in the 578. Feychting M, Floderus B, Ahlbom A. Parental
924. management of spinal cord glioma. Int J Radiat occupational exposure to magnetic fields and
533. Suárez C, et al. Jugular and vagal paragangliomas: Oncol Biol Phys. 1995;33:323–328. childhood cancer (Sweden). Cancer Causes Control.
systematic study of management with surgery and 558. Minehan KJ, Shaw EG, Scheithauer BW, Davis 2000;11:151–156.
radiotherapy. Head Neck. 2013;35:1195–1204. DL, Onofrio BM. Spinal cord astrocytoma: patho- 579. Deleted in review.
534. Konovalov AN, Pitskhelauri DI. Principles of treatment logical and treatment considerations. J Neurosurg. 580. Listernick R, Ferner RE, Liu GT, Gutmann DH.
of the pineal region tumors. Surg Neurol. 2003;59: 1995;83:590–595. Optic pathway gliomas in neurofibromatosis-1:
252–270. 559. Linstadt DE, et al. Postoperative radiotherapy of controversies and recommendations. Ann Neurol.
535. Abay EO, et al. Pineal tumors in children and primary spinal cord tumors. Int J Radiat Oncol Biol 2007;61:189–198.
adolescents: treatment by CSF shunting and Phys. 1989;16:1397–1403. 581. Penn I, Porat G. Central nervous system lympho-
radiotherapy. J Neurosurg. 1981;55:889–895. 560. Sandler HM, Papadopoulos SM, Thornton AF mas in organ allograft recipients. Transplantation.
536. Regis J, et al. Pineal region tumors and the role of Jr, Ross DA. Spinal cord astrocytomas: results of 1995;59:240–244.
stereotactic biopsy: review of the mortality, morbid- therapy. Neurosurgery. 1992;30:490–493. 582. Granovsky MO, Mueller BU, Nicholson HS,
ity, and diagnostic rates in 370 cases. Neurosurgery. 561. Jyothirmayi R, Madhavan J, Nair MK, Rajan Rosenberg PS, Rabkin CS. Cancer in human immu-
1996;39:907–914. B. Conservative surgery and radiotherapy in the nodeficiency virus-infected children: a case series
537. Schild SE, et al. Histologically confirmed pineal treatment of spinal cord astrocytoma. J Neurooncol. from the Children’s Cancer Group and the National
tumors and other germ cell tumors of the brain. 1997;33:205–211. Cancer Institute. J Clin Oncol. 1998;16:1729–
Cancer. 1996;78:2564–2571. 562. Cohen AR, Wisoff JH, Allen JC, Epstein F. Malig- 1735.
538. Lutterbach J, et al. Malignant pineal parenchymal nant astrocytomas of the spinal cord. J Neurosurg. 583. Spitzer A, et al. Complications of immunosuppres-
tumors in adult patients: patterns of care and 1989;70:50–54. sion. J Pediatr. 1992;121:145–153.
prognostic factors. Neurosurgery. 2002;51:44– 563. McGirt MJ, et al. Extent of surgical resection of 584. Nichols KE, Malkin D, Garber JE, Fraumeni JF, Li
56. malignant astrocytomas of the spinal cord: outcome FP. Germ-line p53 mutations predispose to a wide
539. Tate M, et al. The long-term postsurgical prog- analysis of 35 patients. Neurosurgery. 2008;63:55– spectrum of early-onset cancers. Cancer Epidemiol
nosis of patients with pineoblastoma. Cancer. 61. Biomarkers Prev. 2001;10:83–87.
2012;118:173–179. 564. Balmaceda C. Chemotherapy for intramedullary 585. Garrè ML, et al. Medulloblastoma variants: age-
540. Schild SE, et al. Pineal parenchymal tumors: spinal cord tumors. J Neurooncol. 2000;47:293–307. dependent occurrence and relation to Gorlin
clinical, pathologic, and therapeutic aspects. Cancer. 565. Miller DJ, McCutcheon IE. Hemangioblastomas syndrome—a new clinical perspective. Clin Cancer
1993;72:870–880. and other uncommon intramedullary tumors. Res. 2009;15:2463–2471.
541. Preston-Martin S. Descriptive epidemiology of J Neurooncol. 2000;47:253–270. 586. Evans DG, Farndon PA, Burnell LD, Gattamaneni
primary tumors of the spinal cord and spinal 566. Matsuoka S, Itoh M, Shinonome T, Tanimura A. HR, Birch JM. The incidence of Gorlin syndrome
meninges in Los Angeles County, 1972–1985. Intramedullary spinal cord germinoma: case report. in 173 consecutive cases of medulloblastoma. Br J
Neuroepidemiology. 1990;9:106–111. Surg Neurol. 1991;35:122–126. Cancer. 1991;64:959–961.
542. Lowe GM. Magnetic resonance imaging of 567. Spetzger U, et al. Hemangioblastomas of the spinal 587. Hegde MR, et al. A homozygous mutation in
intramedullary spinal cord tumors. J Neurooncol. cord and the brainstem: diagnostic and therapeutic MSH6 causes Turcot syndrome. Clin Cancer Res.
2000;47:195–210. features. Neurosurg Rev. 1996;19:147–151. 2005;11:4689–4693.
543. Catton C, et al. Chordoma: long-term follow-up 568. Lang FF, et al. Central nervous system gan- 588. Deleted in review.
after radical photon irradiation. Radiother Oncol. gliogliomas: Part 2: clinical outcome. J Neurosurg. 589. Koeller KK, Rushing EJ. From the archives of the
1996;41:67–72. 1993;79:867–873. AFIP: medulloblastoma: a comprehensive review
967.e10 Part III: Specific Malignancies

with radiologic-pathologic correlation. Radiographics. 612. Silber JH, et al. Whole-brain irradiation and decline 633. Fontebasso AM, et al. Recurrent somatic muta-
2003;23:1613–1637. in intelligence: the influence of dose and age on IQ tions in ACVR1 in pediatric midline high-grade
590. Barkovich A, ed. Pediatric Neuroimaging. 4th ed. score. J Clin Oncol. 1992;10:1390–1396. astrocytoma. Nat Genet. 2014;46:462–466.
Lippinocott Williams & Wilkins; 2005. 613. Goldwein JW, et al. Updated results of a pilot study of 634. Jennings MT, Freeman ML, Murray MJ. Strategies
591. Arle JE, et al. Prediction of posterior fossa tumor low dose craniospinal irradiation plus chemotherapy in the treatment of diffuse pontine gliomas: the
type in children by means of magnetic resonance for children under five with cerebellar primitive therapeutic role of hyperfractionated radiotherapy
image properties, spectroscopy, and neural networks. neuroectodermal tumors (medulloblastoma). Int J and chemotherapy. J Neurooncol. 1996;28:207–222.
J Neurosurg. 1997;86:755–761. Radiat Oncol Biol Phys. 1996;34:899–904. 635. Packer RJ, et al. Outcome of children with brain
592. Northcott PA, et al. Medulloblastoma comprises 614. Michalski J, Janss A, Vezina G. Results of COG stem gliomas after treatment with 7800 cGy of
four distinct molecular variants. J Clin Oncol. ACNS0331: a phase III trial of involved-field hyperfractionated radiotherapy. A Childrens Cancer
2011;29:1408–1414. radiotherapy (IFRT) and low dose craniospinal Group Phase I/II Trial. Cancer. 1994;74:1827–
593. Robinson G, et al. Novel mutations target distinct irradiation (LD-CSI) with chemotherapy in average- 1834.
subgroups of medulloblastoma. Nature. 2012;488: risk medulloblastoma: a report from COG. Int J 636. Mandell LR, et al. There is no role for hyperfraction-
43–48. Radiat Oncol Biol Phys. 2016;96:937–938. ated radiotherapy in the management of children
594. Jones DTW, et al. Dissecting the genomic 615. Friedman HS, Oakes WJ, Bigner SH, Wikstrand CJ, with newly diagnosed diffuse intrinsic brainstem
complexity underlying medulloblastoma. Nature. Bigner DD. Medulloblastoma: tumor biological and tumors: results of a Pediatric Oncology Group phase
2012;488:100–105. clinical perspectives. J Neurooncol. 1991;11:1–15. III trial comparing conventional vs. hyperfraction-
595. von Bueren AO, et al. Treatment of children and 616. Packer RJ. Chemotherapy for medulloblastoma/ ated radiotherapy. Int J Radiat Oncol Biol Phys.
adolescents with metastatic medulloblastoma primitive neuroectodermal tumors of the posterior 1999;43:959–964.
and prognostic relevance of clinical and biologic fossa. Ann Neurol. 1990;28:823–828. 637. Kaplan AM, et al. Brainstem gliomas in children.
parameters. J Clin Oncol. 2016;34:4151–4160. 617. Gajjar A, et al. Risk-adapted craniospinal radio- A Children’s Cancer Group review of 119 cases.
596. Zeltzer PM, et al. Metastasis stage, adjuvant treat- therapy followed by high-dose chemotherapy and Pediatr Neurosurg. 1996;24:185–192.
ment, and residual tumor are prognostic factors for stem-cell rescue in children with newly diagnosed 638. Jenkin RD, et al. Brain-stem tumors in childhood: a
medulloblastoma in children: conclusions from the medulloblastoma (St Jude Medulloblastoma-96): prospective randomized trial of irradiation with and
Children’s Cancer Group 921 randomized phase III long-term results from a prospective, multicentre without adjuvant CCNU, VCR, and prednisone.
study. J Clin Oncol. 1999;17:832–845. trial. Lancet Oncol. 2006;7:813–820. A report of the Childrens Cancer Study Group.
597. Jallo GI, Kothbauer KF, Abbott IR. Endoscopic third 618. Chintagumpala M, et al. A pilot study of risk-adapted J Neurosurg. 1987;66:227–233.
ventriculostomy. Neurosurg Focus. 2005;19:E11. radiotherapy and chemotherapy in patients with 639. Bouffet E, et al. Radiotherapy followed by high dose
598. Berger MS, Ghatan S, Haglund MM, Dobbins supratentorial PNET. Neuro Oncol. 2009;11:33–40. busulfan and thiotepa: a prospective assessment of
J, Ojemann GA. Low-grade gliomas associated 619. Kalifa C, et al. High-dose chemotherapy in childhood high dose chemotherapy in children with diffuse
with intractable epilepsy: seizure outcome utiliz- brain tumours. Childs Nerv Syst. 1999;15:498–505. pontine gliomas. Cancer. 2000;88:685–692.
ing electrocorticography during tumor resection. 620. Kalifa C, et al. High-dose busulfan and thiotepa 640. Mafee MF, Goodwin J, Dorodi S. Optic nerve sheath
J Neurosurg. 1993;79:62–69. with autologous bone marrow transplantation in meningiomas. Role of MR imaging. Radiol Clin
599. Hirsch JF, Renier D, Czernichow P, Benveniste L, childhood malignant brain tumors: a phase II study. North Am. 1999;37:37–58, ix.
Pierre-Kahn A. Medulloblastoma in childhood. Bone Marrow Transplant. 1992;9:227–233. 641. Gropman AL, et al. Treatment of diencephalic syn-
Survival and functional results. Acta Neurochir 621. Dunkel IJ, et al. High-dose carboplatin, thiotepa, drome with chemotherapy: growth, tumor response,
(Wien). 1979;48:1–15. and etoposide with autologous stem-cell rescue for and long term control. Cancer. 1998;83:166–172.
600. Avula S, et al. Diffusion abnormalities on patients with recurrent medulloblastoma. Children’s 642. Wisoff JH. Management of optic pathway tumors of
intraoperative magnetic resonance imaging as an early Cancer Group. J Clin Oncol. 1998;16:222–228. childhood. Neurosurg Clin N Am. 1992;3:791–802.
predictor for the risk of posterior fossa syndrome. 622. Gajjar A, Pizer B. Role of high-dose chemotherapy 643. Avery RA, et al. Retinal nerve fiber layer thickness
Neuro Oncol. 2015;17:614–622. for recurrent medulloblastoma and other CNS in children with optic pathway gliomas. Am J
601. Pollack IF. Posterior fossa syndrome. Int Rev primitive neuroectodermal tumors. Pediatr Blood Ophthalmol. 2011;151:542–549.e2.
Neurobiol. 1997;41:411–432. Cancer. 2010;54:649–651. 644. Pierce SM, Barnes PD, Loeffler JS, McGinn C,
602. Robertson PL, et al. Incidence and severity of 623. Cefalo G, et al. Temozolomide is an active agent in Tarbell NJ. Definitive radiation therapy in the
postoperative cerebellar mutism syndrome in children with recurrent medulloblastoma/primitive management of symptomatic patients with optic
children with medulloblastoma: a prospective study neuroectodermal tumor: an Italian multi-institutional glioma. Survival and long-term effects. Cancer.
by the Children’s Oncology Group. J Neurosurg. phase II trial. Neuro Oncol. 2014;16:748–753. 1990;65:45–52.
2006;105:444–451. 624. Zamora C, Huisman TAGM, Izbudak I. Supratento- 645. Erkal HS, Serin M, Cakmak A. Management of
603. Liu GT, et al. Visual impairment associated with rial tumors in pediatric patients. Neuroimaging Clin optic pathway and chiasmatic-hypothalamic gliomas
mutism after posterior fossa surgery in children. N Am. 2017;27:39–67. in children with radiation therapy. Radiother Oncol.
Neurosurgery. 1998;42:253–256, 257. 625. Roberts CWM, Orkin SH. The SWI/SNF 1997;45:11–15.
604. Garton GR, et al. Medulloblastoma—prognostic complex—chromatin and cancer. Nat Rev Cancer. 646. Horwich A, Bloom HJ. Optic gliomas: radiation
factors and outcome of treatment: review of the Mayo 2004;4:133–142. therapy and prognosis. Int J Radiat Oncol Biol Phys.
Clinic experience. Mayo Clin Proc. 1990;65:1077– 626. Johann PD, et al. Atypical teratoid/rhabdoid 1985;11:1067–1079.
1086. tumors are comprised of three epigenetic subgroups 647. Packer RJ, et al. Carboplatin and vincristine chemo-
605. Packer RJ, Cogen P, Vezina G, Rorke LB. Medul- with distinct enhancer landscapes. Cancer Cell. therapy for children with newly diagnosed progressive
loblastoma: clinical and biologic aspects. Neuro 2016;29:379–393. low-grade gliomas. J Neurosurg. 1997;86:747–754.
Oncol. 1999;1:232–250. 627. Torchia J, et al. Integrated (epi)-genomic analyses 648. Prados MD, et al. Treatment of pediatric low-grade
606. Warren K, et al. Response Assessment in medul- identify subgroup-specific therapeutic targets in CNS gliomas with a nitrosourea-based multiagent chemo-
loblastoma and leptomeningeal seeding tumours: rhabdoid tumors. Cancer Cell. 2016;30:891–908. therapy regimen. J Neurooncol. 1997;32:235–241.
recommendations from the Response Assessment 628. Fischer-Valuck BW, et al. Assessment of the treat- 649. Ater JL, et al. Randomized study of two chemo-
in Pediatric Neuro-oncology Committee. Neuro- ment approach and survival outcomes in a modern therapy regimens for treatment of low-grade glioma
Oncology In Press. cohort of patients with atypical teratoid rhabdoid in young children: a report from the Children’s
607. Packer RJ, et al. Phase III study of craniospinal tumors using the National Cancer Database. Cancer. Oncology Group. J Clin Oncol. 2012;30:2641–2647.
radiation therapy followed by adjuvant chemotherapy 2017;123:682–687. 650. Bouffet E, et al. Phase II study of weekly vinblastine
for newly diagnosed average-risk medulloblastoma. 629. Fossey M, et al. Atypical teratoid rhabdoid tumor in recurrent or refractory pediatric low-grade glioma.
J Clin Oncol. 2006;24:4202–4208. in the first year of life: the Canadian ATRT registry J Clin Oncol. 2012;30:1358–1363.
608. Sanders RP, et al. M1 Medulloblastoma: high risk experience and review of the literature. J Neurooncol. 651. Packer RJ, et al. Objective response of multiply
at any age. J Neurooncol. 2008;90:351–355. 2017;132:155–162. recurrent low-grade gliomas to bevacizumab and
609. Silverman CL, Simpson JR. Cerebellar medul- 630. Chi SN, et al. Intensive multimodality treatment for irinotecan. Pediatr Blood Cancer. 2009;52:791–795.
loblastoma: the importance of posterior fossa dose children with newly diagnosed CNS atypical teratoid 652. Lin A, et al. BRAF alterations in primary glial and
to survival and patterns of failure. Int J Radiat Oncol rhabdoid tumor. J Clin Oncol. 2009;27:385–389. glioneuronal neoplasms of the central nervous system
Biol Phys. 1982;8:1869–1876. 631. Albright AL, et al. Magnetic resonance scans should with identification of 2 novel KIAA1549:BRAF
610. Rutkowski S, et al. Medulloblastoma in young replace biopsies for the diagnosis of diffuse brain fusion variants. J Neuropathol Exp Neurol. 2012;71:
children. Pediatr Blood Cancer. 2010;54:635–637. stem gliomas: a report from the Children’s Cancer 66–72.
611. Rutkowski S, et al. Treatment of early childhood Group. Neurosurgery. 1993;33:1026–1029, 1030. 653. Sievert AJ, et al. Paradoxical activation and RAF
medulloblastoma by postoperative chemotherapy 632. Lewis PW, et al. Inhibition of PRC2 activity by a inhibitor resistance of BRAF protein kinase fusions
and deferred radiotherapy. Neuro Oncol. 2009;11: gain-of-function H3 mutation found in pediatric characterizing pediatric astrocytomas. Proc Natl Acad
201–210. glioblastoma. Science. 2013;340:857–861. Sci USA. 2013;110:5957–5962.
Cancer of the Central Nervous System • CHAPTER 63 967.e11
967.e11

654. Karajannis MA, et al. Phase II study of sorafenib 677. Wirtz CR, et al. Clinical evaluation and follow-up 700. Hoffman HJ, et al. Aggressive surgical management
in children with recurrent or progressive low-grade results for intraoperative magnetic resonance imaging of craniopharyngiomas in children. J Neurosurg.
astrocytomas. Neuro Oncol. 2014;16:1408–1416. in neurosurgery. Neurosurgery. 2000;46:1112–1120, 1992;76:47–52.
655. Banerjee A, et al. A phase I trial of the MEK inhibitor 1122. 701. Duff J, et al. Long-term outcomes for surgically resected
selumetinib (AZD6244) in pediatric patients with 678. Kim YH, Fayos JV. Intracranial ependymomas. craniopharyngiomas. Neurosurgery. 2000;46:291–
recurrent or refractory low-grade glioma: a Pediatric Radiology. 1977;124:805–808. 302, 305.
Brain Tumor Consortium (PBTC) study. Neuro 679. Merchant TE, et al. Conformal radiotherapy after 702. Carpentieri SC, et al. Memory deficits among
Oncol. 2017;doi:10.1093/neuonc/now282. surgery for paediatric ependymoma: a prospective children with craniopharyngiomas. Neurosurgery.
656. Listernick R, Charrow J, Greenwald M, Mets M. study. Lancet Oncol. 2009;10:258–266. 2001;49:1053–1057, 1058.
Natural history of optic pathway tumors in children 680. MacDonald SM, et al. Proton radiotherapy for 703. Habrand JL, et al. The role of radiation therapy in
with neurofibromatosis type 1: a longitudinal study. childhood ependymoma: initial clinical outcomes the management of craniopharyngioma: a 25-year
J Pediatr. 1994;125:63–66. and dose comparisons. Int J Radiat Oncol Biol Phys. experience and review of the literature. Int J Radiat
657. Fisher MJ, et al. Visual outcomes in children with 2008;71:979–986. Oncol Biol Phys. 1999;44:255–263.
neurofibromatosis type 1-associated optic pathway 681. Wright KD, Gajjar A. New chemotherapy strategies 704. Lin Y, Hansen D, Sayama CM, Pan IW, Lam S.
glioma following chemotherapy: a multicenter ret- and biological agents in the treatment of childhood Transfrontal and transsphenoidal approaches to
rospective analysis. Neuro Oncol. 2012;14:790–797. ependymoma. Childs Nerv Syst. 2009;25:1275–1282. pediatric craniopharyngioma: a national perspective.
658. Sutton LN, et al. Postoperative surveillance imaging 682. Walker RW, Allen JC. Cisplatin in the treatment of Pediatr Neurosurg. 2017;doi:10.1159/000455919.
in children with cerebellar astrocytomas. J Neurosurg. recurrent childhood primary brain tumors. J Clin 705. Kalapurakal JA, Goldman S, Hsieh YC, Tomita T,
1996;84:721–725. Oncol. 1988;6:62–66. Marymont MH. Clinical outcome in children with
659. Zuzak TJ, et al. Outcome of children with low-grade 683. Needle MN, et al. Adjuvant chemotherapy for the craniopharyngioma treated with primary surgery
cerebellar astrocytoma: long-term complications and treatment of intracranial ependymoma of childhood. and radiotherapy deferred until relapse. Med Pediatr
quality of life. Childs Nerv Syst. 2008;24:1447– Cancer. 1997;80:341–347. Oncol. 2003;40:214–218.
1455. 684. Goldwein JW, et al. Recurrent intracranial epen- 706. Merchant TE, et al. Craniopharyngioma: the St. Jude
660. Robertson PL, et al. Cervicomedullary tumors in dymomas in children. Survival, patterns of failure, Children’s Research Hospital experience 1984-2001.
children: a distinct subset of brainstem gliomas. and prognostic factors. Cancer. 1990;66:557–563. Int J Radiat Oncol Biol Phys. 2002;53:533–542.
Neurology. 1994;44:1798–1803. 685. Mason WP, et al. Survival following intensive 707. Stripp DCH, et al. Surgery with or without radiation
661. Pollack IF, Hoffman HJ, Humphreys RP, Becker L. chemotherapy with bone marrow reconstitution for therapy in the management of craniopharyngiomas
The long-term outcome after surgical treatment of children with recurrent intracranial ependymoma—a in children and young adults. Int J Radiat Oncol
dorsally exophytic brain-stem gliomas. J Neurosurg. report of the Children’s Cancer Group. J Neurooncol. Biol Phys. 2004;58:714–720.
1993;78:859–863. 1998;37:135–143. 708. Beltran C, Roca M, Merchant TE. On the
662. Greenberger JS, Cassady JR, Levene MB. Radia- 686. Merchant TE, Boop FA, Kun LE, Sanford RA. A benefits and risks of proton therapy in pediatric
tion therapy of thalamic, midbrain and brain stem retrospective study of surgery and reirradiation for craniopharyngioma. Int J Radiat Oncol Biol Phys.
gliomas. Radiology. 1977;122:463–468. recurrent ependymoma. Int J Radiat Oncol Biol Phys. 2012;82:e281–e287.
663. Deleted in review. 2008;71:87–97. 709. Winkfield KM, et al. Surveillance of craniopha-
664. Merchant TE, et al. Influence of tumor grade on 687. Kretschmar CS. Germ cell tumors of the brain in ryngioma cyst growth in children treated with
time to progression after irradiation for localized children: a review of current literature and new proton radiotherapy. Int J Radiat Oncol Biol Phys.
ependymoma in children. Int J Radiat Oncol Biol advances in therapy. Cancer Invest. 1997;15:187–198. 2009;73:716–721.
Phys. 2002;53:52–57. 688. Kang JK, et al. Experience with pineal region tumors. 710. Jürgen V, et al. Cystic Craniopharyngioma: long-term
665. Moynihan TJ. Ependymal tumors. Curr Treat Options Childs Nerv Syst. 1998;14:63–68. results after intracavitary irradiation with stereotacti-
Oncol. 2003;4:517–523. 689. Shinoda J, et al. Placental alkaline phosphatase as a cally applied colloidal β-emitting radioactive sources.
666. Pajtler KW, et al. Molecular Classification of tumor marker for primary intracranial germinoma. Neurosurgery. 1997;40:263–270.
Ependymal Tumors across All CNS Compartments, J Neurosurg. 1988;68:710–720. 711. Kun LE. Brain tumors. Challenges and directions.
Histopathological Grades, and Age Groups. Cancer 690. Maity A, et al. Craniospinal radiation in the treat- Pediatr Clin North Am. 1997;44:907–917.
Cell. 2015;27:728–743. ment of biopsy-proven intracranial germinomas: 712. Duffner PK, et al. Postoperative chemotherapy and
667. Parker M, et al. C11orf95-RELA fusions drive twenty-five years’ experience in a single center. Int delayed radiation in children less than three years
oncogenic NF-κB signalling in ependymoma. J Radiat Oncol Biol Phys. 2004;58:1165–1170. of age with malignant brain tumors. N Engl J Med.
Nature. 2014;506:451–455. 691. Wolden SL, et al. Radiation therapy for primary 1993;328:1725–1731.
668. Witt H, et al. Delineation of two clinically and intracranial germ-cell tumors. Int J Radiat Oncol 713. Suc E, et al. Brain tumours under the age of
molecularly distinct subgroups of posterior fossa Biol Phys. 1995;32:943–949. three. The price of survival. A retrospective study
ependymoma. Cancer Cell. 2011;20:143–157. 692. Matsutani M, et al. Primary intracranial germ cell of 20 long-term survivors. Acta Neurochir (Wien).
669. Wani K, et al. A prognostic gene expression signature tumors: a clinical analysis of 153 histologically 1990;106:93–98.
in infratentorial ependymoma. Acta Neuropathol. verified cases. J Neurosurg. 1997;86:446–455. 714. Ater JL, et al. MOPP chemotherapy without
2012;123:727–738. 693. Jennings MT, Gelman R, Hochberg F. Intracranial irradiation as primary postsurgical therapy for brain
670. Goldwein JW, et al. Is craniospinal irradiation germ-cell tumors: natural history and pathogenesis. tumors in infants and young children. J Neurooncol.
required to cure children with malignant (anaplas- J Neurosurg. 1985;63:155–167. 1997;32:243–252.
tic) intracranial ependymomas? Cancer. 1991;67: 694. Bouffet E, et al. Combined treatment modality for 715. Geyer JR, et al. Multiagent chemotherapy and
2766–2771. intracranial germinomas: results of a multicentre deferred radiotherapy in infants with malignant
671. Vanuytsel L, Brada M. The role of prophylactic spinal SFOP experience. Société Française d’Oncologie brain tumors: a report from the Children’s Cancer
irradiation in localized intracranial ependymoma. Pédiatrique. Br J Cancer. 1999;79:1199–1204. Group. J Clin Oncol. 2005;23:7621–7631.
Int J Radiat Oncol Biol Phys. 1991;21:825–830. 695. Buckner JC, et al. Phase II trial of primary chemo- 716. Geyer JR, et al. Survival of infants with primitive
672. Nazar GB, et al. Infratentorial ependymomas therapy followed by reduced-dose radiation for CNS neuroectodermal tumors or malignant ependymomas
in childhood: prognostic factors and treatment. germ cell tumors. J Clin Oncol. 1999;17:933–940. of the CNS treated with eight drugs in 1 day: a
J Neurosurg. 1990;72:408–417. 696. Balmaceda C, et al. Chemotherapy without report from the Childrens Cancer Group. J Clin
673. Evans AE, Anderson JR, Lefkowitz-Boudreaux IB, irradiation—a novel approach for newly diagnosed Oncol. 1994;12:1607–1615.
Finlay JL. Adjuvant chemotherapy of childhood CNS germ cell tumors: results of an international 717. Leary SES, Zhou T, Holmes E, Geyer JR, Miller DC.
posterior fossa ependymoma: cranio-spinal irradia- cooperative trial. The First International Central Histology predicts a favorable outcome in young
tion with or without adjuvant CCNU, vincristine, Nervous System Germ Cell Tumor Study. J Clin children with desmoplastic medulloblastoma: a report
and prednisone: a Childrens Cancer Group study. Oncol. 1996;14:2908–2915. from the children’s oncology group. Cancer. 2011;117:
Med Pediatr Oncol. 1996;27:8–14. 697. Khatua S, et al. Treatment of primary CNS germi- 3262–3267.
674. Pollack IF, et al. Intracranial ependymomas of child- nomatous germ cell tumors with chemotherapy prior 718. Rutkowski S, et al. Survival and prognostic factors of
hood: long-term outcome and prognostic factors. to reduced dose whole ventricular and local boost early childhood medulloblastoma: an international
Neurosurgery. 1995;37:655–666, 667. irradiation. Pediatr Blood Cancer. 2010;55:42–46. meta-analysis. J Clin Oncol. 2010;28:4961–
675. Robertson PL, et al. Survival and prognostic factors 698. Gautier A, et al. Markers of recurrence and long- 4968.
following radiation therapy and chemotherapy for term morbidity in craniopharyngioma: a systematic 719. Shaw EG, Wang M, Coons S, et al. MMP. Final report
ependymomas in children: a report of the Children’s analysis of 171 patients. J Clin Endocrinol Metab. of Radiation Therapy Oncology Group (RTOG)
Cancer Group. J Neurosurg. 1998;88:695–703. 2012;97:1258–1267. protocol 9802: Radiation therapy (RT) versus RT
676. Allen JC, Siffert J, Hukin J. Clinical manifestations of 699. Sanford RA. Craniopharyngioma: results of survey + procarbazine, CCNU, and vincristine (PCV)
childhood ependymoma: a multitude of syndromes. of the American Society of Pediatric Neurosurgery. chemotherapy for adult low-grade glioma (LGG).
Pediatr Neurosurg. 1998;28:49–55. Pediatr Neurosurg. 1994;21(suppl 1):39–43. J Clin Oncol. 2008;26(May 20 suppl):abstr 2006.
967.e12 Part III: Specific Malignancies

http://meeting.ascopubs.org/cgi/content/abstract/ management of patients with malignant astrocytoma. 723. Prados MD, Scott C, Sandler H, et al. A phase 3
26/15_suppl/2006. Int J Radiat Oncol Biol Phys. 1998;41(5):1005– randomized study of radiotherapy plus procarbazine,
720. Bleehen NM, Stenning SP. A Medical Research 1011. CCNU, and vincristine (PCV) with or without
Council trial of two radiotherapy doses in the treat- 722. Nelson DF, Diener-West M, Weinstein AS, et al. A BUdR for the treatment of anaplastic astrocytoma:
ment of grades 3 and 4 astrocytoma. The Medical randomized comparison of misonidazole sensitized a preliminary report of RTOG 9404. Int J Radiat
Research Council Brain Tumour Working Party. Br radiotherapy plus BCNU and radiotherapy plus Oncol Biol Phys. 1999;45(5):1109–1115.
J Cancer. 1991;64(4):769–774. BCNU for treatment of malignant glioma after
721. Laperriere NJ, Leung PM, McKenzie S, et al. surgery: final report of an RTOG study. Int J Radiat
Randomized study of brachytherapy in the initial Oncol Biol Phys. 1986;12(10):1793–1800.

You might also like