You are on page 1of 14

C H A P T E R

2
Overview of Pathology and Treatment
of Primary Brain Tumors
Herbert B. Newton
Division of Neuro-Oncology, Departments of Neurology and Neurosurgery, Dardinger Neuro-Oncology Center,
Wexner Medical Center at The Ohio State University and James Cancer Hospital and Solove Research Institute,
Columbus, OH, USA

The epidemiology of primary brain tumors (PBTs) Gliomas (e.g., GBM, AA, oligodendrogliomas, medullo-
was reviewed in detail in Chapter 1. In this chapter, we blastoma) are the largest subgroup within the neuroepi-
provide an overview of the classification, pathology, and thelial class of neoplasms and are also the most common
treatment of the common PBT. PBTs will be diagnosed type of PBT. Tumors of neuroepithelial origin, and glio-
in approximately 30,000–35,000 patients in the United mas in particular, can grow diffusely within the brain or be
States this year and are associated with significant mor- more circumscribed. Diffusely growing tumors are most
bidity and mortality.1–6 Of the estimated 14 patients per common and include the astrocytomas, oligodendroglio-
100,000 population that will develop a PBT this year, 6–8 mas, and mixed oligoastrocytomas. Any of these subtypes
per 100,000 will have a high-grade neoplasm, usually can undergo malignant transformation and degenerate
some form of glioma such as glioblastoma multiforme into the most aggressive form of glioma, the GBM.
(GBM) or anaplastic astrocytoma (AA). Diffuse Astrocytomas. The current WHO classification
divides astrocytomas into diffuse and localized variet-
ies (see Table 3).7–9 The diffuse astrocytomas are intrinsi-
PATHOLOGY OF SELECTED PBTs cally invasive and often travel along white matter tracts
deep into normal brain. There are three groups of dif-
The application of appropriate therapeutic strat- fuse astrocytic neoplasms: astrocytoma (WHO grade II;
egies is dependent on knowing the type of tumor peak age of 30–39 years), AA (WHO grade III; peak age
affecting a given patient. In addition to assisting with of 40–49 years), and GBM (WHO grade IV; peak age of
treatment decisions, the tumor classification and 50–69 years). Diffuse astrocytic tumors can be divided
grade provide important information regarding prog- into fibrillary, protoplasmic, and gemistocytic forms, with
nosis. This chapter follows the World Health Orga- the fibrillary form being most common. The presence of
nization (WHO) classification that separates nervous gemistocytic and protoplasmic cellular variations are
system tumors into different nosological entities and most often seen in WHO grade II tumors. WHO grade II
assigns a grade of I–IV to each lesion (see Table 1), astrocytomas are considered low-grade tumors and usu-
with grade I being biologically indolent and grade ally occur in the cerebral white matter. These tumors are
IV being biologically most malignant and having characterized by a relatively uniform population of pro-
the worst prognosis.7,8 Within the WHO classifica- liferating neoplastic astrocytes in a fibrillary matrix, with
tion, tumors of neuroepithelial and meningeal origin minimal cellular and nuclear pleomorphism or atypia
contain the two largest and most clinically relevant (see Figure 1). Tumor margins are poorly delineated and
groups of neoplasms. suggest significant infiltration into surrounding brain.
Tumors of neuroepithelial origin comprise a large and Mitotic figures are absent and there is no evidence for
diverse group of neoplasms, with a mixture of slowly vascular hyperplasia. Microcystic change is commonly
growing and malignant tumor types (see Table 2).7–9 noted in all variants of grade II astrocytoma. The Ki-67

Handbook of Neuro-Oncology Neuroimaging, Second Edition 9


http://dx.doi.org/10.1016/B978-0-12-800945-1.00002-1 © 2016 Elsevier Ltd. All rights reserved.
10 2.  TREATMENT OF PRIMARY BRAIN TUMORS

TABLE 1  WHO Classification: Tumors of the Central Nervous


System
Tumors of neuroepithelial tissue

Tumors of cranial nerves and spinal nerves

Tumors of the meninges

Lymphomas and hemopoietic neoplasms

Germ-cell tumors

Tumors of the sellar region

Cysts and tumor-like lesions

Metastatic tumors

FIGURE 1  WHO grade II fibrillary astrocytoma. Note the neo-


TABLE 2  WHO Classification: Tumors of Neuroepithelial Tissue plastic astrocytes in a fibrillary matrix, with mildly increased cellular-
Astrocytic tumors ity and pleomorphism. No mitoses or hypervascularity are present.
Hematoxylin and eosin staining (H&E); original magnification 200×.
Oligodendroglial tumors

Ependymal tumors

Mixed gliomas

Choroid plexus tumors

Neuronal and mixed neuronal–glial tumors

Pineal parenchymal tumors

Neuroepithelial tumors of uncertain origin

Embryonal tumors

TABLE 3  WHO Classification: Astrocytic Tumors


DIFFUSE ASTROCYTOMAS
FIGURE 2  WHO grade III fibrillary astrocytoma (AA). The tumor
Astrocytoma (WHO grade II)
is more densely cellular than grade II, with significant cellular and
Fibrillary nuclear pleomorphism and atypia. Mitotic figures are evident. H origi-
nal magnification 200×.
Protoplasmic

Gemistocytic
grade IV) (see Table 3).7–10 Anaplastic astrocytomas are
Anaplastic astrocytoma (WHO grade III) similar to grade II tumors, except for the presence of
Glioblastoma multiforme (WHO grade IV) more prominent cellular and nuclear pleomorphism and
atypia and mitotic activity (see Figure 2). In addition,
Giant cell glioblastoma
grade III and IV tumors usually do not stain as intensely
Gliosarcoma or as homogeneously with glial fibrillary acidic protein
LOCALIZED ASTROCYTOMAS (WHO GRADE I) (GFAP). According to WHO criteria, the critical feature
that upgrades a grade II tumor to an AA is the presence of
Pilocytic astrocytoma
mitotic activity, with anaplastic tumors having Ki-67 indi-
Pleomorphic xanthoastrocytoma ces in the range of 5–10% in most cases. Other features of
Subependymal giant cell astrocytoma anaplasia can be present, such as multinucleated tumor
cells, abnormal mitotic figures, and regions of vascular
proliferation. Necrosis is absent in grade III astrocytomas.
labeling index of WHO grade II astrocytomas is typically Glioblastoma multiforme is classified as a WHO grade
less than 4%, with a mean of approximately 2.0–2.5%. IV tumor and has histological features similar to those
Higher-grade diffuse astrocytomas include AA (WHO of AA, but with more pronounced anaplasia (see Figure
grade III) and GBM (WHO grade IV), as well as the GBM 3(A)).7–10 The presence of microvascular proliferation
variants giant cell glioblastoma and gliosarcoma (WHO and/or necrosis in an otherwise malignant astrocytoma

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


PATHOLOGY OF SELECTED PBTs 11

(A) (B)

(C)

FIGURE 3  WHO grade IV fibrillary astrocytoma (GBM). (A) A highly cellular tumor with marked cellular and nuclear pleomorphism, numer-
ous mitoses, giant cells (H original magnification 400×); (B) dense vascular proliferation (H original magnification 200×); and (C) regions of necro-
sis with pseudopalisading tumor nuclei (H original magnification 100×).

upgrades the tumor to a GBM. Vascular proliferation is (i.e., piloid), GFAP-positive, bipolar processes. These cells
defined as blood vessels with “piling up” of endothelial are found in a biphasic background, which consists of
cells, including the formation of glomeruloid vessels (see dense fibrillary regions alternating with loose, microcys-
Figure 3(B)). The glomeruloid vessels can form undulat- tic areas. Labeling index studies with Ki-67 report values
ing garlands that surround necrotic zones in some cases. of 0.5–1.5% in most tumors. The PXA is a supratentorial
Necrosis can be noted in large amorphous areas, which tumor with a predilection for the superficial temporal
appear ischemic in nature, or can appear as more ser- lobes that usually occurs in younger patients (mean age
piginous regions with surrounding palisading tumor 15–18 years) with a long-standing history of seizure activ-
cells (i.e., perinecrotic pseudopalisading; see Figure ity.7–10 On histological examination, PXA demonstrates
3(C)). Necrosis with nuclear pseudopalisading is essen- significant pleomorphism, with numerous atypical giant
tially pathognomonic for GBM. Other features of GBM cells and astrocytes with prominent nucleoli.11 Also pres-
that are typically prominent include marked cellular and ent are large foamy (xanthomatous) cells with lipidized
nuclear pleomorphism and atypia, mitotic figures and cytoplasm that express GFAP. Subependymal giant cell
multinucleated giant cells, and pronounced infiltrative astrocytoma is an indolent, slowly growing tumor that
capacity into the surrounding brain. Labeling indices typically arises in the walls of the lateral ventricles and is
with Ki-67 are usually in the range of 15–20%, but can be almost invariably associated with tuberous sclerosis.7–10
much higher in some tumors. Oligodendrogliomas and Oligoastrocytomas. Oligoden-
Localized Astrocytomas. In the WHO classification, the drogliomas are a form of diffuse glioma that can be of
localized astrocytomas include the pilocytic astrocytoma pure or mixed histology and are classified as WHO grade
(WHO grade I), pleomorphic xanthoastrocytoma (PXA; II or III.7–10 They typically occur in young to middle-aged
WHO grade II), and the subependymal giant cell astro- adults (peak age 35–45 years) with a history of seizures,
cytoma (WHO grade I).7–10 Pilocytic astrocytomas are within the white matter of the frontal and temporal lobes.
slow-growing, relatively circumscribed tumors that usu- Pure low-grade oligodendroglial tumors (WHO grade II)
ally occur in children (peak age 10–12 years) and young are characterized histologically by a moderately cellu-
adults. These tumors have a predilection for the cerebel- lar, monotonous pattern of cells with round nuclei and
lum, optic nerves and optic pathways, and hypothala- perinuclear halos (the classic “fried-egg” appearance;
mus. The distinctive histological feature is the presence see Figure 4).12 The perinuclear halos are an artifact of
of cells with slender, elongated nuclei and thin, hair-like the formalin fixation process of the tumor tissue. Foci of

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


12 2.  TREATMENT OF PRIMARY BRAIN TUMORS

calcification are frequent and can be quite dense in some in a more aggressive fashion, with a higher proliferative
cases. Delicately branching blood vessels are prominent rate (Ki-67 labeling index >5%) and capacity for invasion
(i.e., “chicken-wire” vasculature), but do not display of surrounding brain. Mixed oligoastrocytomas can be
endothelial proliferation. Oligodendrogliomas have a classified as WHO grade II or III tumors.7–10 Distinct pop-
pronounced invasive capacity and are known to invade ulations of neoplastic oligodendroglial cells and astro-
the gray and white matter diffusely, with a strong ten- cytes can be identified within the mass that have features
dency to form secondary structures of Scherer, in partic- similar to pure versions of the tumor. The percentage of
ular perineuronal satellitosis. Mitoses are absent or rare each cell population can be quite variable, with an even
and necrosis is not present. Labeling studies with Ki-67 mixture of cell types or with one cell type predominating.
usually demonstrate indices less than 5%, with a mean of Advances in molecular neuropathology have begun
approximately 2%. The diagnosis of an anaplastic oligo- to clarify the biological underpinnings of variability in
dendroglioma (WHO grade III) requires the presence of response to treatment of oligodendrogliomas.12–14 The
additional histologic features, including a higher degree majority of tumors demonstrate genetic losses on chro-
of cellularity and mitotic activity, vascular endothe- mosome 1p (40–92%) and/or 19q (50–80%). There is a
lial hyperplasia, nuclear pleomorphism, and regions of strong predilection for deletions of 1p and 19q to occur
necrosis (see Figure 5(A) and (B)). These tumors behave together, but in some tumors they can be singular events.
Patients with oligodendrogliomas that contain deletions
of 1p and 19q are consistently more responsive to irra-
diation and chemotherapy and have an overall median
survival of 8–10 years. In contrast, patients with tumors
that do not have deletion of 1p and 19q are more resis-
tant to all forms of therapy and have an overall median
survival of only 3–4 years.
Medulloblastoma and Other Embryonal Tumors. Embry-
onal tumors are a group of aggressive, malignant neo-
plasms that usually affect children. They are classified
by the WHO as grade IV in all cases (see Table 4).7–10
All embryonal tumors share the common features of
high cellularity, frequent mitoses, regions of necrosis,
and a propensity for metastases along cerebrospinal
fluid (CSF) pathways. Medulloblastoma is the most
common of the embryonal tumors and is considered a
FIGURE 4  WHO grade II oligodendroglioma. This image demon-
strates the classic features of typical oligodendroglioma, with moder-
primitive neuroectodermal tumor (PNET) of the cerebel-
ate cellularity and numerous round cells with the “fried-egg” pattern lum. It usually arises in the midline in children, within
of perinuclear halos and delicate “chicken-wire” vasculature. H origi- the cerebellar vermis, whereas in adults it is more likely
nal magnification 400×. to have an off-center location within the cerebellar

(A) (B)

FIGURE 5  WHO grade III oligodendroglioma. A more densely cellular tumor with prominent cellular and nuclear pleomorphism, mitotic
activity (A), and increased vascularity (B). H original magnification 400×.

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


PATHOLOGY OF SELECTED PBTs 13
hemispheres. The typical medulloblastoma is densely TABLE 5  WHO Classification: Tumors of the Meninges
cellular and composed of undifferentiated cells with
TUMORS OF MENINGOTHELIAL CELLS (MENINGIOMAS)
hyperchromatic, oval to carrot-shaped nuclei with scant
cytoplasm (see Figure 6).9,15 The nuclei have a tendency Meningothelial
to mold against one another. Mitoses and single cell Fibrous (fibroblastic)
necrosis are frequently present. Evidence of anaplasia is
Transitional (mixed)
variable and may include increased nuclear size, abun-
dant mitoses, and the presence of large cells or similar Psammomatous
aggressive cellular morphology. Some tumors may dis- Angiomatous
play immunohistochemical and morphological evidence
Microcystic
for differentiation along neuronal, glial, or mesenchymal
lines. Medulloblastomas are highly proliferative tumors, Secretory
with Ki-67 labeling indices ranging from 15% to 50%. Lymphoplasmacyte rich
Meningioma and Other Tumors of the Meninges. Tumors
Metaplastic
of the meninges comprise a large and diverse group of
neoplasms that mostly have meningothelial or mesenchy- Clear cell
mal, nonmeningothelial origins (see Table 5).8,9 The most Chordoid
common primary tumor of this group is the meningioma
Atypical

TABLE 4  WHO Classification: Embryonal Tumors Papillary

Rhabdoid
Medulloepithelioma
Anaplastic
Ependymoblastoma
MESENCHYMAL, NONMENINGOTHELIAL TUMORS
Medulloblastoma
Lipoma
  Desmoplastic medulloblastoma
Angiolipoma
  Large-cell medulloblastoma
Hibernoma
 Medullomyoblastoma
Liposarcoma (intracranial)
  Melanotic medulloblastoma
Solitary fibrous tumor
Supratentorial PNET
Fibrosarcoma
 Neuroblastoma
Malignant fibrous histiocytoma
 Ganglioneuroblastoma
Leiomyoma
Atypical teratoid/rhabdoid tumor
Leiomyosarcoma

Rhabdomyoma

Rhabdomyosarcoma

Chondroma

Chondrosarcoma

Osteoma

Osteosarcoma

Osteochondroma

Hemangioma

Epithelioid hemangioendothelioma

Hemangiopericytoma

Angiosarcoma
FIGURE 6  WHO grade IV medulloblastoma. Note the dense cel-
lularity and presence of undifferentiated cells with hyperchromatic, Kaposi sarcoma
oval to carrot-shaped nuclei with scant cytoplasm. The nuclei have a
tendency to mold against one another. H original magnification 200×. Continued

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


14 2.  TREATMENT OF PRIMARY BRAIN TUMORS

TABLE 5  WHO Classification: Tumors of the Meninges—cont’d


PRIMARY MELANOCYTIC LESIONS

Diffuse melanocytosis

Melanocytoma

Malignant melanoma

Meningeal melanomatosis

FIGURE 8  WHO grade III anaplastic meningioma. This higher


power view demonstrates increased nuclear pleomorphism and fre-
quent mitoses. H original magnification 400×.

Meningothelial meningiomas are composed of lobules of


typical meningioma cells, with minimal whorl formation.
The tumor cells are uniform in shape, with oval nuclei
that may show central clearing. Fibrous variants have
spindle-shaped cells resembling fibroblasts that form
parallel and interlacing bundles within a matrix of col-
FIGURE 7  WHO grade II meningioma. The tumor demonstrates
a moderately dense, uniform pattern of cells with oval-shaped nuclei lagen and reticulin.
and the presence of many cellular whorl patterns. H original magnifi- Meningioma subtypes that are more likely to display
cation 200×. aggressive clinical behavior and to recur are classified
by the WHO as grade II (atypical, clear cell, chordoid)
(18–20% of intracranial tumors), which has meningothe- and grade III (rhabdoid, papillary, anaplastic).8,9,16,17 On
lial cell origins and is composed of neoplastic arachnoi- histological examination, all of the grade II tumors are
dal cap cells of the arachnoidal villi and granulations. likely to demonstrate increased cellularity, more fre-
Meningiomas can occur anywhere within the intracra- quent mitoses, diffuse or sheetlike growth, nuclear pleo-
nial cavity, but favor the sagittal area along the superior morphism and atypia, and evidence for micronecrosis.
longitudinal sinus, over the lateral cerebral convexities, Grade III tumors such as anaplastic meningioma show
at the tuberculum sellae and parasellar region, the sphe- features consistent with frank malignancy, including a
noidal ridge, and along the olfactory grooves. Numerous high mitotic rate, advanced cytological atypia, nuclear
histologic variants of meningioma are described and pleomorphism, and necrosis (see Figure 8). Invasion of
recognized by the WHO (see Table 5). However, the his- underlying brain is frequently noted in grade III menin-
topathological description of most of these variants has giomas, but can also occur in lower grade variants.
no bearing on the clinical behavior of the tumor. Menin- Proliferation studies using Ki-67 demonstrate labeling
gioma subtypes that have a more indolent nature and low indices ranging from 8% to 15%.
risk for aggressive growth or recurrence are classified as Primary Central Nervous System Lymphoma. Primary
WHO grade I and include the meningothelial, fibrous/ central nervous system lymphomas (PCNSLs) are
fibroblastic, transitional (mixed), secretory, psammoma- malignant tumors classified as WHO grade IV that
tous, angiomatous, microcystic, lymphoplasmocyte-rich, affect adults in the sixth or seventh decade of life.8,9
and metaplastic variants.16,17 Of this group, the menin- They are often multifocal and usually arise in the deep
gothelial, fibrous, and transitional variants are most fre- supratentorial white matter, with a predilection for the
quently diagnosed. The histological features common to periventricular region and basal ganglia. PCNSLs are
most low-grade meningiomas are the presence of whorls composed of a clonal expansion of neoplastic lympho-
(tightly wound, rounded collections of cells), psammoma cytes, typically of the diffuse, large-cell, or immuno-
bodies (concentrically laminated mineral deposits that blastic variety. In 95% of the tumors, the cells have a B
often begin in the center of whorls), intranuclear pseudoin- cell lineage, often with monoclonal IgMκ production.
clusions (areas in which pink cytoplasm protrudes into a On histological examination, PCNSLs display a peri-
nucleus to produce a hollowed-out appearance), and occa- vascular cellular orientation, with expansion of vessel
sional pleomorphic nuclei and mitoses (see Figure 7).16,17 walls and reticulin deposition (see Figure 9).18 Regions

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


RADIATION THERAPY OF PBTs 15
of benign tumors such as meningioma, pilocytic astro-
cytoma, and schwannomas can be curative. For malig-
nant tumors (i.e., GBM, AA), although the lack of a
prospective, controlled randomized clinical trial still
fosters debate in the literature, most neurosurgeons rec-
ommend a near-total or gross-total resection, whenever
possible, of all enhancing tumor volume and regionally
infiltrated brain as defined on T2-weighted or FLAIR
MRI sequences. Gross-total tumor resection is not cura-
tive for these tumor types, but has been associated with
longer overall and progression-free survival (PFS) in
several studies, as well as improved neurological qual-
ity of life.24,25 For tumors that are diffusely infiltrative
or multifocal, a stereotactic biopsy is more likely to pre-
FIGURE 9  WHO grade IV primary CNS lymphoma. Note the pres- serve neurological function than an attempt at resection
ence of neoplastic lymphocytes in an angiocentric growth pattern, with and, in most cases, will be able to provide a histological
nuclear pleomorphism and mitoses. H original magnification 100×. diagnosis to guide further treatment. The accuracy of
stereotactic biopsy is further improved when the region
of necrosis are common, especially if steroids have of interest is defined by contrast enhancement on MRI
been administered prior to the biopsy. The lympho- or abnormal signal on MRI spectroscopy or positron
matous cells are noncohesive and usually have large, emission tomography.
irregular nuclei, prominent nucleoli, and scant cyto-
plasm. From the perivascular region, tumor cells are
noted to invade the surrounding brain parenchyma, RADIATION THERAPY OF PBTs
either in compact cellular aggregates or as singly infil-
trating tumor cells. PCNSLs are highly proliferative External beam fractionated radiation therapy is an
tumors, with Ki-67 labeling indices ranging from 20% appropriate form of treatment for virtually all patients
to 50% in most studies. The diagnosis can be confirmed with high-grade gliomas (i.e., GBM, AA, anaplastic oli-
by immunohistochemical positivity for leukocyte com- godendroglioma (AO), medulloblastoma), as well as for
mon antigen (CD45) and specific B cell markers (CD19, selected low-grade PBTs that are surgically inaccessible
CD20, and CD79a). or have progressed after initial resection.1,26–29 Numer-
ous randomized controlled trials have demonstrated a
survival benefit for high-grade glioma patients receiving
SURGICAL THERAPY OF PBTs surgical resection and irradiation in comparison to resec-
tion alone (approximately 34–38 weeks vs 14–18 weeks,
Surgical intervention is the most common form of respectively). The mechanism of cell death appears to
treatment for PBTs and is an important aspect of ini- be the production of DNA strand damage by ionizing
tial therapy in most patients. Indications for surgery radiation and the generation of highly reactive oxy-
include reduction of tumor burden, alleviation of mass gen radicals that induce further DNA damage and dis-
effect, control of seizures and reversal of neurologi- rupt cellular processes. Sublethal or mortal damage to
cal deficit, confirmation of the histological diagnosis, endothelial cells in tumor vessels may also be of impor-
diversion of CSF by shunting procedures in selected tance. The standard approach is administered in the
cases, and the introduction of local antineoplastic early postoperative phase and initially uses conformal
agents.1,19,20 Advances in neurosurgical technology radiation ports that encompass the T2-weighted target
offer new approaches to tumor removal, such as frame- with a margin of 1–3 cm, using a dose of approximately
based and frameless stereotactic biopsy, preoperative 4500–4700 cGy in 180- to 200-cGy daily fractions. After
functional magnetic resonance imaging (MRI) and this portion has been completed, a “cone down” is per-
intraoperative cortical mapping, neuronavigation and formed, targeting the T1-weighted contrast-enhancing
tumor resection in the awake patient, and the use of volume of the tumor with a 1- to 3-cm margin, bringing
intraoperative MRI.21–23 These techniques allow the the total dose to approximately 6000 cGy. Irradiation is
surgeon to more carefully delineate tumor margins performed over the course of 6–7 weeks, with the patient
and to preserve surrounding regions of eloquent brain receiving treatment 5 days per week. Radiation therapy
(e.g., Broca’s area, primary motor cortex) and delicate schedules can sometimes be modified with hypofrac-
vascular structures, while performing a more aggres- tionation and/or an abbreviated treatment course for
sive and thorough tumor resection. Complete removal elderly patients or for those with a low performance

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


16 2.  TREATMENT OF PRIMARY BRAIN TUMORS

status, while maintaining a similar level of toxicity and effective chemotherapeutic agents for these tumors.37,38
overall survival.30,31 More aggressive approaches to irra- Other agents with mild activity included procarbazine
diation using hyperfractionation schemes have not been (administered alone or in combination with CCNU and
shown to improve tumor control and, in some reports, vincristine), cisplatin, etoposide, carboplatin, and cyclo-
have been associated with worse outcomes.28 Other phosphamide. For the treatment of PCNSL, methotrex-
techniques to increase localized radiation doses to the ate has been shown to be the most active agent, either
tumor resection cavity, such as brachytherapy with per- alone or in combination with other drugs (e.g., cytara-
manent or temporary radioactive seeds, have also had bine, rituximab).43
disappointing results in controlled trials.32 In addition Since 2005, the focus has been on the second-genera-
to the cranial dosage, spinal-axis radiation therapy is tion alkylating agent temozolomide (TZM), which has
necessary for tumors that often seed the meninges, such an activity profile superior to those of the nitrosoureas
as medulloblastoma, pineoblastoma, and anaplastic and other agents. Temozolomide is an imidazotetrazine
ependymoma. derivative of the alkylating agent dacarbazine, with activ-
Stereotactic radiosurgery, using a linear accelerator- ity against systemic and CNS malignancies.37,38,44–47 The
based system (e.g., Cyberknife®) or a Co60-based system drug undergoes chemical conversion at physiological
(e.g., Gamma Knife®) to deliver a single (or a few) high- pH to the active species 5-(3-methyl-1-triazeno)imidaz-
dose radiation fraction(s) to a defined volume using ole-4-carboxamide. Temozolomide exhibits schedule-
stereotactic localization, is another method to boost dependent antineoplastic activity by interfering with
radiation doses in the tumor bed of a newly diagnosed DNA replication through the process of methylation.
or recurrent glioma, while sparing normal surrounding The methylation of DNA is dependent on the forma-
tissues.33–35 Because of the diffuse, infiltrative nature of tion of a reactive methyldiazonium cation, which inter-
the growth pattern of these tumors, the application of acts with DNA at the following sites: N7-guanine (70%),
focal treatment modalities such as radiosurgery remains N3-adenine (9.2%), and O6-guanine (5%). Because TZM
controversial. Retrospective and single-armed, uncon- is stable at acid pH, it can be taken orally in capsules.
trolled prospective trials suggest an improvement in Oral bioavailability is approximately 100%, with rapid
local tumor control rates and survival when using either absorption of the drug. In addition, TZM has excellent
radiosurgical system. However, these results were not penetration of the blood–brain barrier and brain tumor
been confirmed in the randomized, controlled trial of tissue.
radiosurgery for GBM reported by the Radiation Ther- Initial studies of TZM were in patients with recurrent
apy Oncology Group (RTOG 93-05).35,36 In this study, AA and GBM and suggested significant activity.37,38,44–47
203 GBM patients were randomized to receive radio- Subsequent larger studies demonstrated unequivocal
surgery followed by conventional irradiation (60 Gy) efficacy in the recurrent setting. The first study evalu-
and intravenous carmustine chemotherapy (80 mg/m2/ ated the use of TZM (150–200 mg/m2/day × 5 days every
day × 3 days every 8 weeks) or irradiation plus chemo- 28 days) in a series of 162 patients with recurrent malig-
therapy alone. The median survival for the radiosurgi- nant gliomas, including 97 patients with AA.48 In the AA
cal and conventional treatment groups were 13.5 and cohort, there were 6 patients with complete response
13.6 months, respectively (p = 0.5711). In addition, the (CR), 27 with partial response (PR), and another 31 with
2- and 3-year survival rates and patterns of failure were stable disease (SD) (CR + PR + SD = 66%). The response
similar between groups. There was no difference in rate was similar in patients that had failed prior chemo-
general quality of life or retention of cognitive function therapy or were chemotherapy-naïve. Median overall
between groups. PFS was 5.4 months, with 6- and 12-months PFS rates of
46% and 24%, respectively. The median overall survival
was 13.6 months, with 6- and 12-months survival rates of
CHEMOTHERAPY OF PBTs 75% and 56%, respectively. A similar comparative phase
II trial evaluated the activity of TZM versus procarba-
Chemotherapy is used as an adjunctive treatment zine (125–150 mg/m2/day × 28 days every 8 weeks) in a
for malignant PBTs (i.e., mostly high-grade gliomas— cohort of 225 patients with GBM at first relapse.49 Over-
GBM, AA) and for selected low-grade gliomas that all response rates (PR + SD) were significantly higher for
progress through initial surgical resection and irradia- patients in the TZM cohort (45.6% vs 32.7%; p = 0.049).
tion.1,37–40 The addition of chemotherapy has resulted in Treatment with TZM resulted in a significant improve-
modest improvements in the survival of patients with ment in median PFS (12.4 weeks vs 8.32 weeks; p = 0.0063)
malignant glioma, as demonstrated by two detailed and 6-month PFS (21% vs 8%; p = 0.008) in comparison to
meta-analyses.41,42 Over the past two decades and until procarbazine. In addition, the 6-month overall survival
recently, nitrosourea alkylating drugs such as carmus- rate was significantly higher for patients in the TZM arm
tine and lomustine (CCNU) were considered the most of the study (60% vs 44%; p = 0.019).

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


CHEMOTHERAPY OF PBTs 17
Temozolomide has also been applied to GBM patients on/7 days off; 75–100 mg/m2/day), for a total of 6–12
in the “up-front” setting by Stupp and colleagues in a cycles. The two treatment groups were not statistically
set of phase II and III studies, in combination with stan- different at final analysis in terms of median overall
dard external beam irradiation and monthly adjuvant survival (16.6 vs 14.9 months; HR = 1.03) or median PFS
chemotherapy.37,38,50,51 For the phase III study, a total of (5.5 vs 6.7 months; HR = 0.87). MGMT promoter meth-
573 patients were randomly assigned to receive radia- ylation was associated with improved overall survival
tion alone (6000 cGy; 200 cGy/day × 5 days/week for (21.2 vs 14 months; HR = 1.74), PFS (8.7 vs 5.7 months;
6 weeks) or radiotherapy in combination with daily TZM HR = 1.63), and response rate.
(75 mg/m2/day × 7 days/week for 6 weeks).51 After the Angiogenesis is a tightly controlled process that
completion of irradiation, each patient in the chemo- involves growth and maintenance of blood vessels
therapy arm went on to receive six cycles of adjuvant within tissues and organs.55–58 A delicate equilibrium
single-agent TZM (150–200 mg/m2/day × 5 days, every exists between positive angiogenic factors (e.g., vas-
28 days). The overall median survival was 14.6 months cular endothelial growth factor (VEGF), transform-
for the radiotherapy plus TZM cohort and 12.1 months ing growth factor-β (TGF-β)) and inhibitory factors
for the cohort that received irradiation alone, for an (e.g., thrombospondin-1, angiostatin, endostatin).55–58
overall median survival benefit of 2.5 months. The These factors interact with specific receptors on endo-
unadjusted hazard ratio (HR) for death due to the thelial cells and the extracellular matrix, such as VEGF
GBM for the radiotherapy plus TZM cohort was 0.63 receptor-1 (VEGFR-1), VEGFR-2, VEGFR-3, TIE1, and
(p < 0.001, log-rank test). The 2-year survival rate was TIE2.57 There are several important stimuli for conver-
26.5% for the chemoradiation cohort and 10.4% for the sion to the angiogenic phenotype in GBM. The pres-
cohort receiving radiotherapy alone. Responsiveness to ence of hypoxia induces upregulation of secretion of
chemoradiation and overall survival were found to cor- VEGF and expression of VEGFRs in tumor endothelial
relate strongly with the presence of promoter methyla- cells and surrounding regional vasculature.57 Another
tion of the methylguanine-methyltransferase (MGMT) critical element for the switch to the angiogenic phe-
gene, which is important for tumor cell resistance to notype is overactivity of the major growth factor sig-
radiotherapy and chemotherapy. Since the publication naling pathways and loss of certain tumor suppressor
of this report, chemoradiation using low-dose TZM, genes.59 Overexpression and excessive activity of
followed by adjuvant monthly TZM, has become FDA platelet-derived growth factor (PDGF), epidermal
approved and is now the “standard of care” for newly growth factor (EGF), fibroblast growth factor, Ras,
diagnosed GBM patients in the United States and TGF-α, and TGF-β are critical to the development of
around the world. In a 5-year follow-up of this cohort of the angiogenic phenotype.60–62 In addition to growth
patients, Stupp and colleagues report a persistent and factor activity, internal signal transduction mediators
significant difference in survival between the group of also appear to play a role in the angiogenic phenotype.
patients that received radiotherapy and TZM versus The PI3K/Akt/PTEN signaling pathway is involved
the group that received radiotherapy alone.52 For the in the regulation of angiogenesis through the control
chemoradiation group, overall survival at 4 and 5 years of expression of VEGF, hypoxia-inducible factor-1,
was 12.1% and 9.8%, respectively. In contrast, the radio- and thrombospondin-1.63–65
therapy-alone group had 4- and 5-year overall surviv- Numerous investigators have begun to focus on treat-
als of 3.0% and 1.9%, respectively. The differences in ment approaches that capitalize on the prominence
overall survival were highly significant, with a HR of of VEGF signaling in solid tumor angiogenesis.95 The
0.60 (p < 0.0001). Methylation of the MGMT promoter most promising approaches include monoclonal anti-
region was still a very strong predictor of response to bodies against VEGF (e.g., bevacizumab) and VEGFR
TZM chemotherapy. (e.g., IMC-1C11, DC101).59,66–68 Bevacizumab (Avastin™)
Dose-intensive or dose-dense schedules of TZM has advanced the farthest in terms of preclinical evalu-
(e.g., 7  days on/7  days off, 3  weeks on/1  week off) ation and clinical trials.69,70 Bevacizumab is a human-
have also been under investigation, because labora- ized IgG1 monoclonal antibody that is specific for all
tory studies suggest that the higher cumulative dose isoforms of VEGF, preventing their binding to VEGFR.
may result in improved efficacy through augmented It has demonstrated significant activity in preclinical
depletion of MGMT in tumor cells.53 This premise studies against a wide variety of solid tumors, includ-
was tested by the RTOG, in the RTOG-0525 phase III ing gliomas, as a single agent and in combination with
trial, in a cohort of 833 patients with newly diagnosed conventional chemotherapy.71–73
GBM.54 Patients were treated with standard surgical The first report of the use of bevacizumab for brain
resection and chemoradiation, and then randomized tumor patients was by Stark-Vance in 2005, who treated
to receive either conventional TZM (5 days per month; 21 patients with malignant gliomas with bevacizumab
150–200 mg/m2/day) or dose-dense TZM (21  days in combination with irinotecan.74 The response rate was

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


18 2.  TREATMENT OF PRIMARY BRAIN TUMORS

an impressive 43%, although there were two treatment- depending on EIAED or NEIAED), on an every 2-weeks
related deaths (intracranial hemorrhage, intestinal per- schedule. The primary endpoints for the study were
foration). Based on this promising preliminary work, 6-month PFS and objective response rate on follow-up
Vredenburgh and colleagues organized a prospec- MRI, with secondary endpoints of safety and overall
tive phase II trial of bevacizumab and irinotecan for survival. The 6-month PFS rates in the bevacizumab
patients with recurrent malignant gliomas.75 Thirty-two alone and bevacizumab plus irinotecan groups were
patients were enrolled (GBM 23; anaplastic tumors 9) 42.6% and 50.3%, respectively. Objective response rates
and received intravenous bevacizumab (10 mg/kg) and on follow-up MRI were 28.2% for the bevacizumab alone
irinotecan (340 mg/m2 on enzyme-inducing antiepilep- cohort and 37.8% for the bevacizumab plus irinotecan
tic drugs (EIAED), 125 mg/m2 not on EIAED) every 2 group. Median PFS times for the bevacizumab alone
weeks. Radiographic responses were noted in 20 of 32 and bevacizumab plus irinotecan cohorts were 4.2 and
patients (63%; CR 1, PR 19), including 14 of 23 in the 5.6 months, respectively. The overall survival times were
GBM cohort and 6 of 9 in the anaplastic glioma sub- also similar between the single-agent bevacizumab and
group. The median overall PFS was 23 weeks: 20 weeks the bevacizumab plus irinotecan groups (9.2 months vs
in GBM patients and 30 weeks in patients with anaplas- 8.7 months). There was a trend for patients on cortico-
tic tumors. The 6-month PFS and overall survival rates steroids at the beginning of the trial to remain on stable
were 38% and 72%, respectively. In updated reports or decreasing doses over time.86 Grade 3 or 4 toxicity
from 68 patients (GBM 35; anaplastic tumors 33) the was more frequent in the bevacizumab plus irinotecan
MRI response rate was similar (59%).76,77 For the GBM group (65.8%) in comparison to those receiving bevaci-
cohort the 6-month PFS was 46%, with a 6-month over- zumab alone (46.4%). The most common adverse events
all survival of 77%. For the anaplastic glioma subgroup, were hypertension, fatigue, neutropenia, and seizures.
the 6-month PFS rate was 61%, with a median PFS of Intracranial hemorrhage was uncommon in both groups
42 weeks. Eight patients were taken off the study for (2.4% bevacizumab alone, 3.8% bevacizumab plus irino-
thrombotic complications, including pulmonary emboli tecan; 5 total patients, only 1 was grade 4). The authors
(4), deep venous thrombosis (2), thrombotic throm- concluded that bevacizumab, alone or in combination
bocytopenic purpura (1), and thrombotic stroke (1). with irinotecan, was active against recurrent GBM, with
The preliminary use of bevacizumab and irinotecan at 6-month PFS rates that were far superior to the expected
other institutions resulted in similar results, in terms 15% rate for salvage chemotherapy and irinotecan alone.
of MRI response rates, time to progression, and overall Clinicians have begun to evaluate the safety and
survival.73,78–82 The Duke group has also performed a feasibility of using bevacizumab in combination with
phase II study of bevacizumab and irinotecan that was temozolomide during chemoradiation in newly diag-
restricted to only patients with recurrent grade III glio- nosed patients with high-grade gliomas.73,87,88 An early
mas (25 AA, 8 AO).83 There were two cohorts of patients: pilot study of bevacizumab (10 mg/kg every 2 weeks)
the first group of 9 patients received the standard com- in combination with temozolomide (75 mg/m2 dur-
bination regimen every 2 weeks, whereas the second ing irradiation, followed by 150–200 mg/m2 × 5 days
group received bevacizumab (15 mg/kg every 3 weeks) every 4 weeks) was performed by Lai and colleagues
and irinotecan (days 1, 8, 22, 29 of each cycle; 340 mg/m2 in 10 patients with newly diagnosed GBM.87 The tox-
on EIAED, 125 mg/m2 on NEIAED) on a 6-week cycle. icity was considered to be acceptable and consisted of
Objective responses were noted in 29 patients (61% fatigue, myelotoxicity, wound breakdown, DVT/PE,
with at least a PR). The 6-month PFS and overall sur- and one case of radiation-induced optic neuropathy.
vival rates were 55% and 79%, respectively. There was Preliminary efficacy analysis suggested an encourag-
no difference in PFS or survival rates between the two ing mean PFS (range 15–45 weeks). A similar feasibil-
treatment cohorts. Although bevacizumab has been well ity study from Narayana et al. evaluated 15 patients
tolerated by the majority of patients, severe toxicity can with high-grade glioma, with the adjuvant therapy
occur, including hypertension, proteinuria, arterial and phase planned over 1 year (temozolomide 150 mg/m2/
venous thromboembolic events, hemorrhage, gastroin- day).88 Thirteen patients (86.6%) completed the entire
testinal perforation, and impaired wound healing.84 year of adjuvant treatment; radiographic responses
The results of the BRAIN trial were reported by Fried- were noted in 13 of 14 assessable patients (92.8%).
man and colleagues in 2009.73,85 This study was a phase The 1-year PFS and overall survival rates were 59.3%
II, multicenter, open-label, noncomparative trial evaluat- and 86.7%, respectively. Using a more aggressive
ing the efficacy of bevacizumab, alone or in combination approach, Vredenburgh et al. treated 75 patients with
with irinotecan, in a group of 167 patients with GBM in newly diagnosed GBM with standard chemoradia-
first or second relapse. Patients were randomly assigned tion plus bevacizumab (10 mg/kg every 2 weeks), fol-
to receive bevacizumab (10 mg/kg) as a single agent lowed by adjuvant temozolomide (200 mg/m2 × 5 days
or in combination with irinotecan (340 or 125 mg/m2, every month) in combination with bevacizumab

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


Molecular or “Targeted” Treatment 19
and irinotecan (340  mg/m2 on EIAED, 125  mg/m2 HR = 0.79), but was not considered significant because
on NEIAED) every 2 weeks.89 The median PFS for the it did not reach the prespecified improvement target.
entire cohort was 14.2 months, with a median overall During the course of the study, the bevacizumab group
survival of 21.2 months. At 16-month follow-up, the showed an increased symptom burden, worse quality of
overall survival rate was 65%. Another update by the life, and a decline in neurocognitive function in compari-
Duke group reports the results from 125 newly diag- son to the placebo group.
nosed GBM patients who were treated with standard
chemoradiation and temozolomide, in addition to bev-
acizumab (10 mg/kg intravenously every 2 weeks).90 MOLECULAR OR “TARGETED”
Overall, the combination of temozolomide and bevaci- TREATMENT
zumab was tolerated well in the majority of patients,
with 96% completing the full course of chemoradiation As noted above, conventional chemotherapeutic
and 90% able to continue on with treatment into the approaches to treatment for malignant glioma are not
adjuvant chemotherapy phase. Toxicities associated predicated on the biology of the malignant phenotype.
with treatment discontinuation included pulmonary It has become apparent that the transformed pheno-
emboli, CNS hemorrhage, pancytopenia, wound dehis- type of brain tumor cells is highly complex and results
cence, and colonic perforation. from the dysfunction of a variety of interrelated regula-
Based on these intriguing preliminary studies, two tory pathways.93–95 The transformation process involves
large phase III clinical trials were performed to determine amplification or overexpression of oncogenes in combi-
the impact of adding bevacizumab to standard chemo- nation with loss or lack of expression of tumor suppres-
radiation with temozolomide in newly diagnosed GBM sor genes. Oncogenes and signal transduction molecules
patients. The first study was the Avastin in Glioblastoma that have been demonstrated to be important for glio-
Trial (AVAglio), an international effort based mainly magenesis include PDGF and its receptor (PDGFR),
in Europe, which enrolled 921 patients and randomly EGF and EGFR, CDK4, mdm-2, Ras, phosphoinositol-3
assigned them to receive either bevacizumab (10 mg/kg kinase (PI3K), Akt, and mTOR (mammalian target of
every 2 weeks) or placebo, plus standard radiotherapy rapamycin). Tumor suppressor genes of importance in
and concomitant temozolomide.91 The coprimary end- glial transformation include p53, retinoblastoma, p16
points of the study were PFS and overall survival. After and p15 (i.e., INK4a, INK4b), DMBT1, and PTEN. Most
chemoradiation was completed, patients received either of these tumor suppressor genes function as negative
bevacizumab or placebo every 2 weeks, along with adju- regulators of the cell cycle, whereas others are inhibitors
vant temozolomide (150–200 mg/m2/day) for up to six of important internal signal transduction pathways. The
cycles. After the completion of adjuvant temozolomide, net effect of these acquired abnormalities is dysregula-
patients would continue with single-agent bevacizumab tion of, and an imbalance between, the activity of the cell
(15 mg/kg every 3 weeks) or placebo until disease pro- cycle and apoptotic pathways.
gression or unacceptable toxicity. The median PFS was Because the survival of patients with high-grade
longer in the bevacizumab group than in the placebo gliomas has remained so poor using conventional che-
group (median 10.6  months vs 6.2  months; HR  = 064; motherapeutic approaches, new treatment modalities
p < 0.001). Overall survival was not significantly different are being investigated that have a more molecular, “tar-
between the groups (median 16.8 months vs 16.7 months; geted” mechanism of action, with the ability to overcome
HR = 0.88; p = 0.10). The overall survival rates of the bev- the transformed phenotype.59,60,96–98 Advances in growth
acizumab and placebo groups were 72.4% and 66.3% factor and signal transduction biology are now provid-
at 1 year (p = 0.049) and 33.9% and 30.1% at 2 years ing the background for the development of “molecu-
(p = 0.24), respectively. Health-related quality of life and lar therapeutics,” a new class of drugs that manipulate
performance status were maintained longer in the beva- and exploit these pathways. Molecular drugs targeting
cizumab group; in addition, they required a lower dose critical signal transduction pathway effectors, such as
of corticosteroids. The other phase III trial was the RTOG PDGFR, EGFR, Ras, PI3K, and mTOR, have entered clini-
0825 study, which enrolled and randomized 637 patients cal trials in brain tumor patients.59,60,96–98 To date, numer-
with newly diagnosed GBM and had design and treat- ous drugs have been tested in phase I, II, and III trials,
ment groups similar to those of the AVAglio study, except designed to target various key receptors, signal trans-
that during the adjuvant treatment phase, patients could duction proteins, and cell membrane-related proteins
continue temozolomide for up to 12 cycles.92 There was such as EGFR (gefitinib, erlotinib, cetuximab), PDGFR
no difference between the bevacizumab and the placebo (imatinib), Ras (tipifarnib, lonafarnib), mTOR (temsiroli-
groups in terms of overall survival (median 15.7 months mus), integrins (cilengitide), and protein kinase C (enza-
vs 16.1 months; HR = 1.13). The PFS was longer for the staurin), as well as multikinase drugs (e.g., lapatinib)
bevacizumab cohort (median 10.7 months vs 7.3 months; that can target combinations of the above. Preliminary

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


20 2.  TREATMENT OF PRIMARY BRAIN TUMORS

results suggest only modest activity against recurrent 19. Parney IF, Berger MS. Principles of brain tumor surgery. Handbk
high-grade gliomas when used as single agents. Cur- Clin Neurol. 2012;104:187–213.
20. Dunn IF, Black PM. The neurosurgeon as local oncologist: cellular
rent and subsequent clinical trials will investigate using and molecular neurosurgery in malignant glioma therapy. Neuro-
molecular drugs in combination with conventional che- surg. 2003;52:1411–1424.
motherapeutic agents (e.g., TZM, hydroxyurea), with 21. Choi BD, Mehta AI, Batich KA, et al. The use of motor map-
other molecular drugs that target different signal trans- ping to aid resection of eloquent gliomas. Neurosurg Clin N Am.
duction pathways, and with irradiation. 2012;23:215–225.
22. Orringer DA, Golby A, Jolesz F. Neuronavigation in the surgical
management of brain tumors: current and future trends. Expert Rev
Med Devices. 2012;9:491–500.
Acknowledgments 23. Liang D, Schulder M. The role of intraoperative magnetic
The authors thank Shawna Huckell and Nicole Ghaffari for research resonance imaging in glioma surgery. Surg Neurol Int. 2012;3:
assistance. Dr. Newton was supported in part by National Cancer S320–S327.
Institute Grant CA 16058 and the Dardinger Neuro-Oncology Center 24. Lacroix M, Abi-Said D, Fourney DR, et al. A multivariate analysis
Endowment Fund. of 416 patients with glioblastoma multiforme: prognosis, extent of
resection, and survival. J Neurosurg. 2001;95:190–198.
25. Bloch O, Han SJ, Cha S, et al. Impact of extent of resection for recur-
References rent glioblastoma on overall survival: clinical article. J Neurosurg.
2012;117:1032–1038.
1. Newton HB. Primary brain tumors: review of etiology, diagnosis, 26. Caruso C, Carcaterra M, Donato V. Role of radiotherapy for high
and treatment. Am Fam Phys. 1994;49:787–797. grade glioma management. J Neurosurg Sci. 2013;57:163–169.
2. Preston-Martin S. Epidemiology of primary CNS neoplasms. Neu- 27. Wind JJ, Young R, Saini A, Sherman JH. The role of adjuvant radia-
rol Clin. 1996;14:273–290. tion therapy in the management of high-grade gliomas. Neurosurg
3. Davis FG, McCarthy BJ. Current epidemiological trends and Clin N Am. 2012;23:247–258.
surveillance issues in brain tumors. Expert Rev Anticancer Ther. 28. Minniti G, Goldsmith C, Brada M. Radiotherapy. Handbk Clin
2001;1:395–401. Neurol. 2012;104:215–228.
4.  Osborne RH, Houben MPWA, Tijssen CC, Coebergh JWW, van Duijn 29. Kortmann RD, Jeremic B, Bamberg M. Radiotherapy in the manage-
CM. The genetic epidemiology of glioma. Neurol. 2001;57:1751–1755. ment of low-grade gliomas. In: Petrovich Z, Brady LW, Apuzzo ML,
5. Wrensch M, Minn Y, Chew T, Bondy M, Berger MS. Epidemiology Bamberg M, eds. Combined Modality Therapy of Central Nervous
of primary brain tumors: current concepts and review of the litera- System Tumors. Vol. 16. Berlin: Springer; 2001:317–326.
ture. Neuro-Oncol. 2002;4:278–299. 30. Chang EL, Yi W, Allen PK, Levin VA, Sawaya RE, Maor MH. Hypo-
6. Hess KR, Broglio DR, Bondy ML. Adult glioma incidence trends in fractionated radiotherapy for elderly or younger low-performance
the United States, 1977–2000. Cancer. 2004;101:2293–2299. status glioblastoma patients: outcome and prognostic factors. Int J
7. Rodriguez FJ, Giannini C. Diagnostic neuropathology of tumors of Rad Oncol Biol Phys. 2003;56:519–528.
the central nervous system. Handbk Clin Neurol. 2012;104:77–107. 31. Roa W, Brasher PMA, Bauman G, et al. Abbreviated course of ra-
8. Kleihues P, Louis DN, Scheithauer BW, et al. The WHO classifi- diation therapy in older patients with glioblastoma multiforme:
cation of tumors of the nervous system. J Neuropathol Exp Neurol. a prospective randomized clinical trial. J Clin Oncol. 2004;22:
2002;61:215–225. 1583–1588.
9. McLendon RE, Enterline DS, Tien RD, Thorstad WL, Bruner JM. 32. Selker RG, Shapiro WR, Burger P, et al. The Brain Tumor Coopera-
Tumors of central neuroepithelial origin. In: Bigner DD, McLendon tive Group NIH trial 87-01: a randomized comparison of surgery,
RE, Bruner JM, eds. Russell & Rubinstein’s Pathology of Tumors of the external radiotherapy, and carmustine versus surgery, interstitial
Nervous System. Sixth Edition. Volume 1. London: Arnold; 1998;9: radiotherapy boost, external radiation therapy, and carmustine.
307–571. Neurosurg. 2002;51:343–357.
10. Li J, Sulman E, Aldape K. Molecular biology of brain tumors. Hand- 33. Elaimy AL, Mackay AR, Lamoreaux WT, et al. Clinical outcomes
bk Clin Neurol. 2012;104:23–34. of gamma knife radiosurgery in the salvage treatment of patients
11. Sharma A, Nand Sharma D, Kumar JP, Kishor RG. Pleomorphic with recurrent high-grade glioma. World Neurosurg. 2013;80:872–
xanthoastrocytoma – a clinico-pathological review. Neurol Neuro- 878.
chir Pol. 2011;45:379–386. 34. Koga T, Saito N. Efficacy and limitations of stereotactic radiosur-
12. Matthews S, Succar P, Jelinek H, et al. Diagnosis of oligodendrogli- gery in the treatment of glioblastoma. Neurol Clin Chir (Tokyo).
oma: molecular and classical histological assessment in the twenty- 2012;52:548–552.
first century. Asia Pac J Clin Oncol. 2012;8:213–216. 35. Tsao MN, Mehta MP, Whelan TJ, et al. The American Society for
13. Jenkinson MD, Walker C, Brodbelt AR, et al. Molecular genetics, Therapeutic Radiology and Oncology (ASTRO) evidence-based
imaging and treatment of oligodendroglial tumours. Acta Neuro- review of the role of radiosurgery for malignant glioma. Int J Rad
chir (Wien). 2010;152:1815–1825. Oncol Biol Phys. 2005;63:47–55.
14. Bromberg JE, van den Bent MJ. Oligodendrogliomas: molecular 36. Souhami L, Seiferheld W, Brachman D, et al. Randomized com-
biology and treatment. Oncologist. 2009;14:155–163. parison of stereotactic radiosurgery followed by conventional
15. Eberhart CG, Burger PC. Anaplasia and grading in medulloblasto- radiotherapy with carmustine to conventional radiotherapy with
mas. Brain Pathol. 2003;13:376–385. carmustine for patients with glioblastoma multiforme: report of
16. Lamszus K. Meningioma pathology, genetics, and biology. J Neuro- Radiation Therapy Oncology Group 93-05 protocol. Int J Rad Oncol
pathol Exp Neurol. 2004;63:275–286. Biol Phys. 2004;60:853–860.
17. Perry A, Gutmann DH, Reifenberger G. Molecular pathogenesis of 37. Newton HB. Chemotherapy of high-grade astrocytomas.
meningiomas. J Neuro-Oncol. 2004;70:183–202. In: Newton HB, ed. Handbook of Brain Tumor Chemotherapy. Vol. 24.
18. Deckert M, Engert A, Bruck W, et al. Modern concepts in the bi- London: Elsevier Medical Publishers-Academic Press; 2006:347–363.
ology, diagnosis, differential diagnosis, and treatment of primary 38. Blakeley J, Grossman SA. Chemotherapy with cytotoxic and cyto-
central nervous system lymphoma. Leukemia. 2011;25:1797–1807. static agents in brain cancer. Handbk Clin Neurol. 2012;104:229–265.

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


References 21
39. Viaccoz A, Lekoubou A, Ducray F. Chemotherapy in low-grade 61. Jensen RL. Growth factor-mediated angiogenesis in the ma-
gliomas. Curr Opin Oncol. 2012;24:694–701. lignant progression of glial tumors: a review. Surg Neurol.
40. Malkin MG. Chemotherapy of low-grade astrocytomas. In: Newton 1998;49:189–196.
HB, ed. Handbook of Brain Tumor Chemotherapy. Vol. 25. London: 62. Dunn IF, Heese O, Black PM. Growth factors in glioma angio-
Elsevier Medical Publishers-Academic Press; 2006:364–370. genesis: FGFs, PDGF, EGF, and TGFs. J Neuro-Oncol. 2000;50:
41. Fine HA, Dear KBG, Loeffler JS, Black PM, Canellos GP. Meta-analysis 121–137.
of radiation therapy with and without adjuvant chemotherapy for 63. Zagzag D, Zhong H, Scalzitti JM, et al. Expression of hypoxia-
malignant gliomas in adults. Cancer. 1993;71:2585–2597. inducible factor 1α in brain tumors. Association with angiogenesis,
42. Steward LA, Burdett S, Souhami RL, Stenning S. Chemothera- invasion, and progression. Cancer. 2000;88:2606–2618.
py in adult high-grade glioma: a systematic review and meta- 64. Jiang BH, Jiang G, Zheng JZ, et al. Phosphatidylinositol 3-kinase
analysis of individual patient data from 12 randomised trials. signaling controls levels of hypoxia-inducible factor 1. Cell Growth
Lancet. 2002;359:1011–1018. Differ. 2001;12:363–369.
43. El Kamar FG, Abrey LE. Chemotherapy for primary central ner- 65. Wen S, Stolarov J, Myers MP, et al. PTEN controls tumor-induced
vous system lymphoma. In: Newton HB, ed. Handbook of Brain angiogenesis. Proc Natl Acad Sci USA. 2001;98:4622–4627.
Tumor Chemotherapy. Vol. 28. London: Elsevier Medical Publishers- 66. Hicklin DJ, Ellis LM. Role of the vascular endothelial growth
Academic Press; 2006:395–406. factor pathway in tumor growth and angiogenesis. J Clin Oncol.
44. Stevens MFG, Newlands ES. From triazines and triazenes to temo- 2005;23:1011–1027.
zolomide. Eur J Cancer. 1993;20A:1045–1047. 67. Levine AM, Tulpule A, Quinn DI, et al. Phase I study of antisense
45. Newlands ES, O’Reilly SM, Glaser MG, et al. The Charing Cross oligonucleotide against vascular endothelial growth factor: de-
Hospital experience with temozolomide in patients with gliomas. crease in plasma vascular endothelial growth factor with potential
Eur J Cancer. 1996;2A:2236–2241. clinical efficacy. J Clin Oncol. 2006;24:1712–1719.
46. Newlands ES, Stevens MFG, Wedge SR, Wheelhouse RT, Brock C. 68. Wachsberger PR, Burd R, Cardi C, et al. VEGF trap in combination
Temozolomide: a review of its discovery, chemical properties, with radiotherapy improves tumor control in U87 glioblastoma.
pre-clinical development and clinical trials. Cancer Treat Rev. Int J Rad Oncol Biol Phys. 2007;67:1526–1537.
1997;23:35–61. 69. Ranieri G, Patruno R, Ruggieri E, Montemurro S, Valerio P, Ribatti D.
47. Hvizdos KM, Goa KL. Temozolomide. CNS Drugs. 1999;12:237–243. Vascular endothelial growth factor (VEGF) as a target of bevaci-
48. Yung WKA, Prados MD, Yaya-Tur R, et al. Multicenter phase II tri- zumab in cancer: from the biology to the clinic. Curr Med Chem.
al of temozolomide in patients with anaplastic astrocytoma or ana- 2006;13:1845–1857.
plastic oligoastrocytoma at first relapse. J Clin Oncol. 1999;17:2762– 70. Shih T, Lindley C. Bevacizumab: an angiogenesis inhibitor for the
2771. treatment of solid malignancies. Clin Ther. 2006;28:1779–1802.
49. Yung WKA, Albright RE, Olson J, et al. A phase II study of temo- 71. Kunkel P, Ulbricht U, Bohlen P, et al. Inhibition of intracranial gli-
zolomide vs. procarbazine in patients with glioblastoma multi- oma angiogenesis and growth in vivo by systemic treatment with
forme at first relapse. Br J Cancer. 2000;83:588–593. a monoclonal antibody against vascular endothelial growth factor
50. Stupp R, Dietrich PY, Kraljevic SO, et al. Promising survival for receptor-2. Cancer Res. 2001;61:6624–6628.
patients with newly diagnosed glioblastoma treated with concomi- 72. Stefanik DF, Fellows WK, Rizkalla LR, et al. Monoclonal antibodies
tant radiation plus temozolomide followed by adjuvant temozolo- to vascular endothelial growth factor (VEGF) and the VEGF recep-
mide. J Clin Oncol. 2002;20:1375–1382. tor, FLT-1, inhibit the growth of C6 glioma in a mouse xenograft.
51. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus J Neuro-Oncol. 2001;55:91–100.
concomitant and adjuvant temozolomide for glioblastoma. New 73. Newton HB. Overview of angiogenesis and the use of bevacizum-
Engl J Med. 2005;352:987–996. ab in patients with malignant gliomas. Curr Signal Transduct Ther.
52. Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with 2013;8:25–35.
concomitant and adjuvant temozolomide versus radiothera- 74. Stark-Vance V. Bevacizumab and CPT-11 in the treatment of re-
py alone on survival in glioblastoma in a randomized phase III lapsed malignant glioma. In: World Federation of Neuro-Oncology
study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. Meeting. 2005:91.
2009;10:459–466. 75. Vredenburgh JJ, Desjardins A, Herndon JE, et al. Phase II trial of
53. Strik HM, Marosi C, Kaina B, Neyns B. Temozolomide dosing regi- bevacizumab and irinotecan in recurrent malignant glioma. Clin
mens for glioma patients. Curr Neurol Neurosci Rep. 2012;12:286–293. Cancer Res. 2007;13:1253–1259.
54. Gilbert MR, Wang M, Aldape KD, et al. Dose-dense temozolomide 76. Goli KJ, Desjardins A, Herndon JE, et al. Phase II trial of bevaci-
for newly diagnosed glioblastoma: a randomized phase III clinical zumab and irinotecan in the treatment of malignant gliomas (ab-
trial. J Clin Oncol. 2013;31:4085–4091. stract). J Clin Oncol (Suppl). 2007;18S:75s.
55. Cavallaro U, Christofori G. Molecular mechanisms of tumor angio- 77. Vredenburgh JJ, Desjardins A, Herndon JE, et al. Bevacizumab
genesis and tumor progression. J Neuro-Oncol. 2000;50:63–70. plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol.
56. Webb CP, Vande Woude GF. Genes that regulate metastasis and 2007;25:4722–4729.
angiogenesis. J Neuro-Oncol. 2000;50:71–87. 78. Kang T, Jin T, Elinzano H, Peereboom D. Irinotecan and bevaci-
57. Plate KH. Mechanisms of angiogenesis in the brain. J Neuropath Exp zumab in progressive primary brain tumors, an evaluation of ef-
Neurol. 1999;58:313–320. ficacy and safety. J Neuro-Oncol. 2008;89:113–118.
58. Hardee ME, Zagzag D. Mechanisms of glioma-associated neovas- 79. Gilbert MR, Wang M, Aldape K, Lassman A, Sorensen AG,
cularization. Am J Pathol. 2012;181:1126–1141. Mikkelson T, et al. RTOG 0625: a phase II study of bevacizumab
59. Newton HB. Molecular neuro-oncology and the development of and irinotecan in recurrent glioblastoma (GBM) (abstract). J Clin
“targeted” therapeutic strategies for brain tumors. Part 2 – PI3K/ Oncol (Suppl). 2009;27:89s.
Akt/PTEN, mTOR, SHH/PTCH, and angiogenesis. Expert Rev An- 80. Ali SA, McHayleh WM, Ahmad A, Sehgal R, Braffet M, Rahman M,
ticancer Ther. 2004;4:105–128. et al. Bevacizumab and irinotecan in glioblastoma multiforme:
60. Newton HB. Molecular neuro-oncology and the development of a series of 13 cases. J Neurosurg. 2008;109:268–272.
“targeted” therapeutic strategies for brain tumors. Part 1 – growth 81. Bokstein F, Shpigel S, Blumenthal DT. Treatment with bevacizum-
factor and ras signaling pathways. Expert Rev Anticancer Ther. ab and irinotecan for recurrent high-grade glial tumors. Cancer.
2003;3:595–614. 2008;112:2267–2273.

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS


22 2.  TREATMENT OF PRIMARY BRAIN TUMORS

82. Poulsen HS, Grunnet K, Sorensen M, et al. Bevacizumab plus irino- 90. Vredenburgh JJ, Adejardins A, Kirkpatrick JP, et al. Addition of
tecan in the treatment of patients with progressive recurrent malig- bevacizumab to standard radiation therapy and daily temozolo-
nant brain tumours. Acta Oncol. 2009;48:52–58. mide is associated with minimal toxicity in newly diagnosed
83. Desjardins A, Reardon DA, Herndon JE, et al. Bevacizumab plus glioblastoma multiforme. Int J Rad Oncol Biol Phys. 2012;82:
irinotecan in recurrent WHO grade 3 malignant gliomas. Clin Can- 58–66.
cer Res. 2008;14:7068–7073. 91. Chinot OL, Wick W, Mason W, et al. Bevacizumab plus radiotherapy-
84. Gordon MS, Cunningham D. Managing patients treated with beva- temozolomide for newly diagnosed glioblastoma. New Engl J Med.
cizumab combination therapy. Oncology. 2005;69:25–33. 2014;370:709–722.
85. Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and 92. Gilbert MR, Dignam JJ, Armstrong TS, et al. A randomized trial of
in combination with irinotecan in recurrent glioblastoma. J Clin bevacizumab for newly diagnosed glioblastoma. New Engl J Med.
Oncol. 2009;27:4733–4740. 2014;370:699–708.
86. Vredenburgh JJ, Cloughesy T, Samant M, et al. Corticosteroid use 93. Marumoto T, Saya H. Molecular biology of glioma. Adv Exp Med
in patients with glioblastoma at first or second relapse treated with Biol. 2012;746:2–11.
bevacizumab in the BRAIN study. Oncology. 2010;15:1329–1334. 94. Li J, Sulman E, Aldape K. Molecular biology of brain tumors. Hand-
87. Lai A, Filka E, McGibbon B, Nghiemphu PL, et al. Phase II pilot bk Clin Neurol. 2012;104:23–34.
study of bevacizumab in combination with temozolomide and 95. Westphal M, Lamszus K. The neurobiology of gliomas: from cell
regional radiation therapy for up-front treatment of patients with biology to the development of therapeutic approaches. Nat Rev
newly diagnosed glioblastoma mutiforme: Interim analysis of safe- Neurosci. 2011;12:495–508.
ty and tolerability. Int J Rad Oncol Biol Phys. 2008;71:1372–1380. 96. Newton HB. Molecular neuro-oncology and the development of
88. Narayana A, Golfinos JG, Fischer I, et al. Feasibility of using beva- “targeted” therapeutic strategies for brain tumors. Part 5 – apoptosis
cizumab with radiation therapy and temozolomide in newly diag- and cell cycle. Expert Rev Anticancer Ther. 2005;5:355–378.
nosed high-grade glioma. Int J Rad Oncol Biol Phys. 2008;72:383–389. 97. Sathornsumetee S, Rich JN. Molecularly targeted therapy in neuro-
89. Vredenburgh JJ, Desjardins A, Reardon DA, et al. The addition of oncology. Handbk Clin Neurol. 2012;104:255–278.
bevacizumab to standard radiation therapy and temozolomide fol- 98. Hamza MA, Gilbert M. Targeted therapy in gliomas. Curr Oncol
lowed by bevacizumab, temozolomide, and irinotecan for newly Rep. 2014;16(4):379.
diagnosed glioblastoma. Clin Cancer Res. 2011;17:4119–4124.

I.  OVERVIEW OF NEURO-ONCOLOGICAL DISORDERS

You might also like