Adult Gliomas
Adult Gliomas
Adult Gliomas
Adult Gliomas
C O N T I N UU M A UD I O By Howard Colman, MD, PhD, FAAN
I NT E R V I E W A V AI L A B L E
ONLINE
P6SJseWGqdq1qm8onTV2TE13o9Z1Akm74qifjXwQXy4w/knSw6EEt4q/Qw/6Dxcgpxzs8WFacSsJpZJ4o on 03/06/2023
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
G
grant support from the Alliance
for Clinical Trials in Oncology liomas are a heterogeneous group of primary brain tumors that
(LAPS-UT003), The Hope present multiple diagnostic and therapeutic challenges. Important
Foundation, the Huntsman issues facing the clinician evaluating a patient with possible or
Cancer Foundation, the
Musella Foundation confirmed glioma include the initial (prediagnosis) evaluation,
Continued on page 1475 need for neurosurgical referral, prognostic considerations, and
decisions about treatment. Recent developments have demonstrated the central
UNLABELED USE OF
PRODUCTS/INVESTIGATIONAL
role of molecular markers in the diagnosis, subtyping, and treatment of
USE DISCLOSURE: gliomas and their relationship with specific clinical and radiographic
Dr Colman discusses the presentations. This article highlights some of the important clinical,
unlabeled/investigational use of
agents targeting mutations/ radiographic, pathologic, and molecular considerations when evaluating a
alterations in BRAF (including patient with a possible or confirmed diagnosis of glioma.
vemurafenib), EGFR, FGFR, IDH,
and NTRK to treat gliomas that
harbor alterations in those genes PRESENTATION AND INITIAL EVALUATION
and the use of checkpoint Gliomas most commonly present with signs and symptoms related to a
inhibitors, chimeric antigen
receptor T cells, and
space-occupying lesion with symptoms often related to anatomic location. The
pembrolizumab to treat time course and nature of symptoms vary significantly based on growth rate,
glioblastoma. location, degree of associated edema, degree of increased intracranial pressure,
© 2020 American Academy
and whether seizures are part of the presentation. In general, lower-grade and
of Neurology. slower-growing tumors present with gradually progressive symptoms and more
CONTINUUMJOURNAL.COM 1453
One of the key decisions in the evaluation of a patient with a possible glioma is
whether to pursue immediate referral for neurosurgical intervention and tissue
diagnosis, pursue additional diagnostic evaluations, or initially observe with
follow-up MRI studies to better determine the natural history and refine the
differential diagnosis before pursuing a tissue diagnosis. Some of the key
considerations in whether a patient requires immediate biopsy or resection of an
P6SJseWGqdq1qm8onTV2TE13o9Z1Akm74qifjXwQXy4w/knSw6EEt4q/Qw/6Dxcgpxzs8WFacSsJpZJ4o on 03/06/2023
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
TABLE 1-1 Molecular and Clinical Features of Selected World Health Organization
2016 Glioma Diagnoses
World Health Organization Diagnostic Molecular Other Common Characteristic Clinical and
2016 Integrated Diagnosis Markers Genomic Alterations Radiographic Features
Diffuse astrocytoma and IDH-mutant, 1p/19q-intact ATRX, TP53 Younger adults, supratentorial,
anaplastic astrocytoma, frontal predominance
IDH-mutant (grade II and III)
Glioblastoma, IDH-mutant IDH-mutant, 1p/19q-intact ATRX, TP53; some Younger adults, supratentorial,
(grade IV) cases: CDKN2 loss frontal predominance,
enhancement common
Oligodendroglioma and IDH-mutant, 1p/19q CIC, FUBP1, TERT Younger adults, supratentorial,
anaplastic codeleted promoter frontal predominance
oligodendroglioma,
IDH-mutant and 1p/
19q-codeleted (grade II and III)
evaluation of a newly
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
CONTINUUMJOURNAL.COM 1455
earlier time point and was diagnosed only after malignant transformation to
● Loss of chromosomes
grade IV. However, while currently given the name glioblastoma and/or the 1p/19q is the molecular
same grade as histologically similar IDH-wildtype tumors, the IDH-mutant hallmark that distinguishes
tumors have a prognosis that is significantly better within any particular oligodendroglial from
grade.11–13 In addition, developing data suggest that the presence or absence of astrocytic gliomas within the
IDH-mutant group.
additional molecular features including CDKN2 loss and others may be a better
predictor of prognosis within IDH-mutant tumors than traditional histologic ● For the diagnosis of either
grading.14–16 Furthermore, growing data suggest that traditional histologic astrocytoma or anaplastic
features of grading, such as atypia and proliferation rate, may be less important astrocytoma with IDH
mutation, the presence or
in IDH-mutant tumors compared with IDH-wildtype tumors.16 absence of other molecular
alterations including CDKN2
IDH -WILDTYPE GLIOMAS AND GLIOBLASTOMAS. The vast majority of glioblastomas loss may be more important
in older patients are glioblastoma, IDH-wildtype and represent the so-called than the actual WHO grade
for prognosis.
primary glioblastoma (that do not arise from a known lower-grade precursor). In
contrast to lower-grade gliomas, the majority of these glioblastomas demonstrate ● The vast majority of
no mutation in IDH (IDH-wildtype). In general, patients with IDH-wildtype primary glioblastomas
tumors have a worse prognosis than those with IDH-mutant tumors of the same in older adults are
histology and grade. IDH-wildtype astrocytomas and glioblastomas often share a IDH-wildtype, and even
lower-grade IDH-wildtype
mutational profile that can include alterations in EGFR, NF1, TP53, PTEN, TERT astrocytic tumors with
promoter, CDKN2 loss, chromosome 7 gain, chromosome 10 loss, and others. A appropriate molecular
2018 publication by the cIMPACT-NOW group specifically recommended that alterations (EGFR or
infiltrating gliomas that are IDH-wildtype and demonstrate (1) EGFR chromosome 7 gain/
chromosome 10 loss or TERT
amplification, or (2) combined whole chromosome 7 gain and whole chromosome promoter) can be classified
10 loss, or (3) TERT promoter mutation should be more appropriately given the as diffuse astrocytic glioma,
diagnosis of diffuse astrocytic glioma, IDH-wildtype, with molecular features of IDH-wildtype, with
glioblastoma, WHO grade IV.17 Thus, even if histopathology is found to be lower molecular features of
glioblastoma, WHO grade IV.
grade (grades II and III), astrocytomas that harbor these glioblastomalike
alterations tend to have a poor prognosis and behave similarly to histologic grade
IV tumors. The recommended cIMPACT-NOW diagnosis for these patients (with
diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma,
WHO grade IV) is an attempt to assign a diagnosis appropriate with that
prognosis, despite the lower-grade histologic features.17–19 An example of a patient
with lower-grade histology but molecular findings consistent with glioblastoma is
shown in CASE 1-2.
CONTINUUMJOURNAL.COM 1457
CASE 1-1 A 39-year-old woman presented with a new-onset seizure. Brain MRI
demonstrated a nonenhancing mass in the left lateral frontoparietal
regions, with fluid-attenuated inversion recovery (FLAIR) and T1
postcontrast images at diagnosis shown in FIGURES 1-1A and 1-1B,
respectively. She underwent resection with pathology of diffuse
astrocytoma, World Health Organization (WHO) grade II. Molecular
testing at that time showed the tumor was mutant for IDH1, ATRX, and
TP53, and chromosome 1p/19q was intact. By using the new WHO
diagnostic criteria, this tumor would be classified as diffuse astrocytoma,
IDH-mutant, WHO grade II. After surgery, she was treated with radiation
to a dose of 54 Gy with concurrent temozolomide and six cycles of
adjuvant temozolomide. She had been off treatment since, and MRI
8 years after diagnosis is shown in FIGURES 1-1C and 1-1D (FLAIR and T1
postcontrast, respectively) with no evidence of progression since the
initial treatment. At her most recent visit, she had a Karnofsky
Performance Status Scale score of 100, was working full time, and was
seizure free.
COMMENT This case illustrates some of the common clinical, radiographic, and
diagnostic molecular features of IDH-mutant astrocytomas. The patient’s
good response to treatment, long progression-free interval, and excellent
clinical status highlight the potential for good outcomes with this tumor
type (as well as oligodendrogliomas, IDH-mutant and 1p/19q codeleted).
GRADES WITH BRAF GENE ALTERATIONS. This group of tumors can have variable
histologic findings, with some indistinguishable from more typical IDH-mutant
FIGURE 1-1
Imaging of the patient in CASE 1-1. Axial fluid-attenuated inversion recovery (FLAIR) (A) and
postcontrast T1-weighted (B) brain MRI demonstrates a nonenhancing mass in the left
lateral frontoparietal region at diagnosis. FLAIR (C) and postcontrast T1-weighted (D)
images 8 years after diagnosis show a resection cavity with no evidence of progression
since the initial treatment.
CONTINUUMJOURNAL.COM 1459
and BRAF gene fusions (BRAF-KIAA1549), and these alterations are mutually
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
CASE 1-2 A 45-year-old man presented with a new-onset seizure and some subtle
but progressive speech difficulty. Brain MRI demonstrated a
nonenhancing mass in the medial left temporal lobe, with
fluid-attenuated inversion recovery (FLAIR) and postcontrast
T1-weighted images at diagnosis as shown in FIGURES 1-2A and 1-2B,
respectively. He underwent almost complete resection of the tumor in
the left temporal lobe with pathology of diffuse astrocytoma, World
Health Organization (WHO) grade II. The tumor was determined to be
IDH-wildtype by immunohistochemistry and sequencing. Next-generation
DNA sequencing showed that the tumor was EGFR amplified and had
chromosome 7 gain, PTEN loss, and a TERT promoter mutation. If the
patient had been diagnosed by using the revised 2016 WHO criteria, this
tumor would have been classified as diffuse astrocytoma, IDH-wildtype,
WHO grade II. O-6-methylguanine-DNA methyltransferase (MGMT)
promoter was unmethylated.
He received radiation to 60 Gy with concurrent temozolomide followed
by six cycles of adjuvant temozolomide. Despite treatment typical for
higher-grade astrocytoma (due to the unfavorable molecular findings), the
patient experienced rapid recurrence involving the left insula, frontal
lobe, and corpus callosum approximately 2 years later
(FIGURES 1-2C and 1-2D).
COMMENT This case illustrates the importance of the molecular alterations in diffuse
gliomas relative to prognosis. Although age, presentation, imaging,
pathology, and histologic grade were similar to those in CASE 1-1, this patient
experienced a very different and much worse outcome despite receiving
similar treatment. The findings of EGFR mutation, chromosome 7 gain, TERT
promoter mutation, and others in a histologically low-grade IDH-wildtype
astrocytoma are generally associated with a prognosis that is similar to
glioblastoma, and the cIMPACT-NOW (Consortium to Inform Molecular and
Practical Approaches to CNS Tumor) group recently published
recommendations that these tumors are given the diagnosis diffuse
astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma,
WHO grade IV.16 This case highlights the need for clinicians to be aware of
these subtleties in the diagnosis and treatment of diffuse gliomas.
FIGURE 1-2
Imaging of the patient in CASE 1-2. Axial fluid-attenuated inversion recovery (FLAIR) (A) and
postcontrast T1-weighted (B) brain MRI demonstrates a nonenhancing mass in the medial left
temporal lobe at diagnosis. FLAIR (C) and postcontrast T1-weighted (D) images show aggressive
recurrence approximately 2 years after initial treatment with radiation and temozolomide.
CONTINUUMJOURNAL.COM 1461
distinct cell morphology in which greater than 50% of tumors harbor the BRAF
V600E mutation. Some debate exists about the overlap of epithelioid
glioblastoma and anaplastic pleomorphic xanthoastrocytoma and whether these
really represent distinct diagnoses.20 Case reports and small series in the
literature describe radiographic responses of various glioma subtypes harboring
BRAF alterations to drugs targeting either BRAF V600E mutation and drugs that
P6SJseWGqdq1qm8onTV2TE13o9Z1Akm74qifjXwQXy4w/knSw6EEt4q/Qw/6Dxcgpxzs8WFacSsJpZJ4o on 03/06/2023
COMMENT As in the previous cases, this case demonstrates another example of the
spectrum of molecular findings and prognoses that can occur in diffuse
gliomas, sometimes with similar clinical and radiographic findings. Although
this patient’s tumor has a relatively “benign” and well-circumscribed
appearance on the initial imaging, which might suggest a diagnosis of
several grade I tumors such as pilocytic astrocytoma, dysembryoplastic
neuroepithelial tumor (DNET), or ganglioglioma based on imaging alone, the
molecular findings here are diagnostic of the grade IV H3K27M midline
tumor, and accurately predicted the poor prognosis of this patient despite
aggressive treatment.
EPENDYMAL TUMORS. Ependymal tumors are glial neoplasms that are distinct from
the diffuse gliomas and can present in both childhood and adulthood.6 In contrast
to the diffuse gliomas, this group of tumors is thought to arise from the
ependymal lining of the ventricular cavities or from radial glia precursors in these
regions.23 Ependymal tumors comprise different histologies including
FIGURE 1-3
Imaging of the patient in CASE 1-3. Axial fluid-attenuated inversion recovery (FLAIR) (A) and
postcontrast T1-weighted (B) brain MRIs demonstrate a relatively well-circumscribed,
nonenhancing mass involving the hypothalamus and inferior basal ganglia bilaterally. A
repeat brain postcontrast T1-weighted sequence (C) approximately 2.5 years after initial
treatment with radiation and temozolomide shows aggressive recurrence in the midline and
anteriorly and superiorly in the frontal lobe on the right.
CONTINUUMJOURNAL.COM 1463
Surgery
associated with poor
Initial treatment for all grades of gliomas starts with maximal safe resection. In prognosis.
addition to providing definitive histologic and molecular diagnosis, surgery
also can provide a clinical benefit from a reduction of mass effect, and more ● Initial treatment for most
extensive resection has been associated with better survival in several glioma diffuse gliomas starts with
maximal safe resection.
subtypes. For newly diagnosed glioblastoma (and by extension higher-grade
IDH-wildtype gliomas), a greater extent of resection of an enhancing tumor has
been associated with better overall survival. In some retrospective studies,
resection of greater than 95% of presurgical enhancement is needed for
significant improvement in survival,28,29 although in one analysis from a large
retrospective series the volume of residual enhancement after resection was
more important than the percentage of resection.30
Only recently have outcomes relative to the extent of resection in gliomas been
analyzed based on molecular subtypes. In the case of lower-grade gliomas, a
greater extent of resection and a smaller volume of residual (often
nonenhancing) tumor have been found to be positive prognostic factors in
several retrospective analyses.31 In analyses of gliomas that compared surgical
outcomes of IDH-mutant versus IDH-wildtype gliomas, the extent of resection
of enhancing disease (but not nonenhancing tumor) was prognostic in
IDH-wildtype tumors, whereas the extent of resection of total tumor volume
(enhancing and nonenhancing disease) was prognostic in IDH-mutant tumors.32
These findings suggest that the role and surgical target for maximal resection
may differ based on the molecular subtype of glioma.
In suspected lower-grade gliomas, the timing of surgery is also an important
clinical decision. The effect of early resection versus initial observation of
presumed low-grade gliomas has not been evaluated in a randomized trial.
Uncontrolled and retrospective analyses have suggested a survival benefit of
early and more complete resection.31–33 To date, no studies have convincingly
defined a population of patients in which delayed or incomplete resection has
demonstrated clinical benefit. As a result, the most common approach in
specialized centers is to proceed with maximal safe resection in patients with
suspected lower-grade gliomas, as recommended in National Comprehensive
Cancer Network guidelines for suspected low-grade gliomas.17,34
CONTINUUMJOURNAL.COM 1465
T2-weighted sequences (gross tumor volume) plus a margin that can range from
1 cm to 3 cm. The therapeutic effect of radiation is generally related to dose, not
modality (eg, photons versus protons), with current clinical trials comparing
intensity-modulated radiation therapy to proton therapy mainly aimed at determining
if there is any benefit of proton therapy in terms of reduced neurotoxicity.
The role of chemotherapy as a standard part of treatment in newly diagnosed
P6SJseWGqdq1qm8onTV2TE13o9Z1Akm74qifjXwQXy4w/knSw6EEt4q/Qw/6Dxcgpxzs8WFacSsJpZJ4o on 03/06/2023
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
predictive marker of a
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
of the patient, and the electric fields are delivered via a battery pack that is worn
better outcome with
as a shoulder or backpack and attached to the electrodes via a wiring harness. temozolomide.
The randomized study examining the effect of tumor treating fields in newly
diagnosed glioblastoma involved random assignment of patients after they ● Standard treatment
completed radiation and temozolomide to either temozolomide alone (control) or options with survival benefit
in randomized studies for
temozolomide and tumor treating fields (experimental) groups. This was not a newly diagnosed
placebo-controlled study, and thus, it is not possible to determine whether glioblastoma include
differences in supportive care or follow-up between arms played some role in radiation, temozolomide,
the outcomes.40 However, the study did demonstrate a survival benefit in the and tumor treating fields.
tumor treating fields group versus control (20.9 months versus 16.0 months,
respectively).39 Tumor treating fields are FDA approved for use in newly diagnosed
glioblastoma along with temozolomide after radiation based on these data.
Recurrent Glioblastoma
Despite initial therapy, glioblastoma progresses in virtually every patient. While
clinical and molecular prognostic markers such as IDH mutation and MGMT
promoter methylation have been associated with better outcomes in recurrent
tumors in at least some studies, the survival of recurrent glioblastoma is generally
poor, with one randomized study showing a median survival of approximately
9 months from the time of first progression.41 Patients with poor prognostic
factors (eg, older age, poor performance status) have even worse survival
outcomes. Despite the testing of many different agents with various mechanisms
of action in recurrent glioblastoma, no treatment has been shown to improve
overall survival in this group of patients. Cytotoxic chemotherapies have very
low radiographic response rates in recurrent glioblastoma.42 Thus, the
antiangiogenic agent bevacizumab (a humanized monoclonal antibody against
vascular endothelial growth factor [VEGF]) created some excitement when it
was found to have a high response rate (40% to 60%) in initial single-arm
studies, as well as improved progression-free survival compared with historical
controls.43 The observation of a high response rate and improved progression-
free survival with bevacizumab led to accelerated approval by the FDA for the
treatment of recurrent glioblastoma. However, the provisional FDA approval in
2009 stipulated that confirmatory phase 3 studies be performed. Two phase 3
randomized, placebo-controlled studies of radiation, temozolomide, plus
bevacizumab versus radiation and temozolomide alone were performed with
patients with newly diagnosed glioblastoma,44,45 and subsequently a separate
phase 3 study of bevacizumab plus lomustine versus lomustine alone in recurrent
glioblastoma.41 The results of all of these studies were similar (despite the
different patient populations) in that all confirmed a high proportion of
radiographic responses to bevacizumab in glioblastoma and improvement of
progression-free survival in the 3- to 5-month range. However, none of these
studies demonstrated a significant benefit on overall survival. Despite these
findings, it was concluded that the net clinical benefit observed in terms of
response rate, progression-free survival, and maintenance of performance status
CONTINUUMJOURNAL.COM 1467
was important, and this led to conversion of bevacizumab to full approval by the
FDA in 2019. Several other agents including lomustine, tumor treating fields,
carmustine, carboplatin, and others have shown some activity in recurrent
glioblastoma and are listed in National Comprehensive Cancer Network
guidelines,34 but as with bevacizumab, none of these have demonstrated an overall
survival benefit in recurrent glioblastoma in a randomized study.
P6SJseWGqdq1qm8onTV2TE13o9Z1Akm74qifjXwQXy4w/knSw6EEt4q/Qw/6Dxcgpxzs8WFacSsJpZJ4o on 03/06/2023
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
progression-free survival in
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
CONTINUUMJOURNAL.COM 1469
Targeted Therapy
One of the modern breakthroughs in oncology was the identification of multiple
genomic alterations that drive tumor initiation or maintenance, rendering such
tumors to functional blockade by drugs that target the mutant protein or kinase
or one of its downstream effectors. Examples of such targetable alterations in
oncology are numerous but began with ones such as BCR-ABL fusions
(in chronic myelogenous leukemia), HER2 amplification (in breast cancer), and
EGFR mutations (in lung cancer). There are several potentially targetable
alterations observed in different subtypes of lower-grade glioma and
glioblastoma. In IDH-mutant tumors, the IDH mutation itself has been a
potential target with a number of drugs developed to inhibit the neomorphic
function of the mutant IDH enzyme to block the production of 2-hydroxyglutarate.
Two of these IDH inhibitors are FDA approved in acute myeloid leukemia with
higher frequencies, as well as other alterations that are seen at lower frequencies
proven more effective for
including FGFR gene fusions, NTRK gene fusions, and BRAF mutations and prolonging survival than
fusions. For NTRK and BRAF mutations and fusions, drugs that are FDA approved radiation alone in lower-
in other tumor types have potential efficacy in glioma. Case reports and small grade diffuse gliomas with
IDH mutation.
series suggest clinical benefit of specific FGFR, BRAF, and NTRK inhibitors, but
the rarity of these alterations in glioblastoma has complicated attempts at larger
studies to verify activity in this tumor type. In the case of inhibitors of the BRAF
V600E mutation, initial data from a larger basket-type trial of multiple histologies,
unfortunately, suggest that the activity of one drug, vemurafenib, may be less in
higher-grade gliomas harboring this mutation than in other tumor types.20
Multiple attempts to target EGFR alterations have been made with targeted
tyrosine kinase inhibitors, vaccines, and EGFR-specific antibodies, but in all cases,
the phase 3 studies of these drugs or vaccines have not demonstrated any survival
benefit. Despite these disappointments, development continues on other
approaches including newer kinase inhibitors, bifunctional antibodies, as well as
chimeric antigen receptor (CAR) T cells engineered to target EGFRvIII mutation
and other glioblastoma alterations.
Another breakthrough in oncology was the demonstration in multiple tumor
types that drugs that modulate the immune response can have significant clinical
efficacy. In some cases, these immunotherapy agents are possibly curative in
subsets of patients who were previously incurable. Most of the currently
FDA-approved immunotherapy agents are humanized monoclonal antibodies
that act by blocking cell surface signaling proteins that inhibit activation of
immune responses against tumor cells and/or tumor neoantigens. Several phase 2
and 3 studies have tested the activity of immune checkpoint inhibitors in newly
diagnosed and recurrent glioblastoma. To date, all studies of single-agent
checkpoint inhibitors in glioblastoma have been negative for a survival benefit. A
small randomized phase 2 study of the checkpoint inhibitor pembrolizumab
given before and after surgery for recurrent glioblastoma was the first to
demonstrate a potential survival benefit in glioblastoma.59 The fact that the
benefit was observed in combination with surgery raises the hope that
immunotherapy can be used successfully in the treatment of glioblastoma but
also suggests that additional factors beyond the presence of a checkpoint
inhibitor alone are necessary for efficacy in glioblastoma (in contrast to other
solid tumors). However, the benefit of surgery plus pembrolizumab in a limited
number of patients requires validation in larger studies.
Clinical Trials
To date, the only treatments that have demonstrated a survival benefit in diffuse
gliomas and glioblastoma are radiation, cytotoxic chemotherapy, and tumor
treating fields. Despite the growing evidence and information regarding genomic
and epigenetic alterations that define molecular subgroups of glioma, no
FDA-approved drug targeting a specific mutation or molecular alteration
(beyond bevacizumab), or any immunomodulatory drug is FDA approved
CONTINUUMJOURNAL.COM 1471
KEY POINT specifically for gliomas. This situation highlights the need to increase enrollment
of all histologies and grades of gliomas in clinical trials. In the current National
● Participation in a clinical
trial is an important
Comprehensive Cancer Network guidelines, clinical trial enrollment is the
consideration in the preferred option for the treatment of both newly diagnosed and recurrent glioma
treatment of all histologies, for eligible patients.33
grades, and molecular
P6SJseWGqdq1qm8onTV2TE13o9Z1Akm74qifjXwQXy4w/knSw6EEt4q/Qw/6Dxcgpxzs8WFacSsJpZJ4o on 03/06/2023
subtypes of glioma.
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
CONCLUSION
Adult gliomas represent a clinically and molecularly heterogeneous group of
tumors. An understanding of the differences in presentation, diagnosis, and
treatment is important for neurologists. The recent advances in the
understanding of molecular subtypes of tumors have led to a dramatic revision of
the approach to the diagnosis and treatment of adult gliomas. Much of this
new knowledge is described in the 2016 World Health Organization
Classification of Tumors of the Central Nervous System, although this field
continues to evolve rapidly, and some diagnostic entities, such as the diagnosis of
diffuse astrocytic glioma, IDH-wildtype, with molecular features of
glioblastoma, WHO grade IV, remain consensus recommendations and will
presumably be incorporated into future WHO classifications. Despite the
dramatic advances in the biologic understanding and classification of adult
gliomas, the majority of these tumors remain incurable with standard approaches
of surgery, radiation, chemotherapy, and tumor treating fields, and efforts
aimed at the development and testing of new therapies for specific glioma
subtypes in clinical trials are ongoing.
REFERENCES
1 Klein JP, Dietrich J. Neuroradiologic pearls for 7 Giannini C, Scheithauer BW, Weaver AL, et al.
neuro-oncology. Continuum (Minneap Minn) Oligodendrogliomas: reproducibility and
2017;23(6, Neuro-oncology):1619–1664. prognostic value of histologic diagnosis and
doi:10.1212/CON.0000000000000543 grading. J Neuropathol Exp Neurol 2001;60(3):
248–262. doi:10.1093/jnen/60.3.248
2 Omuro AM, Leite CC, Mokhtari K, Delattre JY.
Pitfalls in the diagnosis of brain tumours. Lancet 8 Louis DN, Perry A, Reifenberger G, et al. The 2016
Neurol 2006;5(11):937–498. doi:10.1016/S1474- World Health Organization Classification of
4422(06)70597-X Tumors of the Central Nervous System: a
summary. Acta Neuropathol 2016;131(6):803–820.
3 Law M, Yang S, Wang H, et al. Glioma grading:
doi:10.1007/s00401-016-1545-1
sensitivity, specificity, and predictive values of
perfusion MR imaging and proton MR 9 Louis DN, Ohgaki H, Wiestler OD, et al.
spectroscopic imaging compared with WHO Classification of Tumours of the Central
conventional MR imaging. AJNR Am J Neuroradiol Nervous System. 4th ed. Lyon, France:
2003;24(10):1989–1998. International Agency for Research on
Cancer; 2016.
4 Arrillaga I, Singhal A, Jarhult S, et al. HCP-03 newly
diagnosed single brain mass–implementation 10 Louis DN, Aldape K, Brat DJ, et al. Announcing
and performance of a hospital-wide cIMPACT-NOW: the Consortium to Inform
management pathway. Neuro Oncol 2015; Molecular and Practical Approaches to CNS
17(suppl 5):v101. doi:10.1093/neuonc/nov216.03 Tumor Taxonomy. Acta Neuropathol 2017;133(1):
1–3. doi:10.1007/s00401-016-1646-x
5 Partap S, Monje M. Pediatric brain tumors.
Continuum (Minneap Minn) 2020; 11 Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2
26(6, Neuro-oncology):1553–1583. mutations in gliomas. N Engl J Med 2009;360(8):
765–773. doi:10.1056/NEJMoa0808710
6 Ostrom QT, Gittleman H, Liao P, et al. CBTRUS
Statistical report: primary brain and other central 12 Cancer Genome Atlas Research Network.
nervous system tumors diagnosed in the United Comprehensive, integrative genomic analysis of
States in 2010–2014. 2017;19(suppl 5):v1–v88. diffuse lower-grade gliomas. N Engl J Med 2015;
doi:10.1093/neuonc/nox158 372(26):2481–2498. doi:10.1056/NEJMoa1402121
CONTINUUMJOURNAL.COM 1473
37 Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene 49 Perry JR, Laperriere N, O'Callaghan CJ, et al.
silencing and benefit from temozolomide in Short-course radiation plus temozolomide in
glioblastoma. N Engl J Med 2005;352(10): elderly patients with glioblastoma. N Engl J Med
997–1003. doi:10.1056/NEJMoa043331 2017;376(11):1027–1037. doi:10.1056/
NEJMoa1611977
38 Gorlia T, van den Bent MJ, Hegi ME, et al.
Nomograms for predicting survival of patients 50 Cairncross JG, Ueki K, Zlatescu MC, et al. Specific
with newly diagnosed glioblastoma: prognostic genetic predictors of chemotherapeutic
factor analysis of EORTC and NCIC Trial response and survival in patients with anaplastic
P6SJseWGqdq1qm8onTV2TE13o9Z1Akm74qifjXwQXy4w/knSw6EEt4q/Qw/6Dxcgpxzs8WFacSsJpZJ4o on 03/06/2023
Downloaded from http://journals.lww.com/continuum by gAuij8Aa4+A071iuQoJbgrwM8/2Un/wWxPpuO6xEGplU7+p
CONTINUUMJOURNAL.COM 1475