Glioblastoma Multiforme

Back

Practice Essentials

Glioblastoma multiforme (GBM) is the most common and most malignant of the glial tumors. See the image below.



View Image

Histopathologic slide demonstrating a glioblastoma multiforme (GBM).

See Brain Lesions: 9 Cases to Test Your Management Skills, a Critical Images slideshow, to review cases including meningiomas, glioblastomas and craniopharyngiomas, and to determine the best treatment options based on the case history and images.

Signs and symptoms

The clinical history of a patient with glioblastoma multiforme (GBM) is usually short (< 3 months in > 50% of patients). Common presenting symptoms include the following:

Neurologic symptoms and signs can be either general or focal and reflect the location of the tumor, as follows:

The etiology of GBM is unknown in most cases. Suggested causes include the following:

See Presentation for more detail.

Diagnosis

No specific laboratory studies are helpful in diagnosing GBM. Tumor genetics are useful for predicting response to adjuvant therapy.

Imaging studies of the brain are essential for making the diagnosis, including the following:

Other diagnostic measures that may be considered include the following:

In most cases, complete staging is neither practical nor possible. These tumors do not have clearly defined margins; they tend to invade locally and spread along white matter pathways, creating the appearance of multiple GBMs or multicentric gliomas on imaging studies.

See Workup for more detail.

Management

No current treatment is curative. Standard treatment consists of the following:

Key points regarding radiotherapy for GBM include the following:[7, 8]

The optimal chemotherapeutic regimen for GBM is not yet defined, but adjuvant chemotherapy appears to yield a significant survival benefit in more than 25% of patients.[15, 1, 16, 17, 18, 19]

Agents used include the following:

Surgical options include gross total resection (better survival), subtotal resection, and biopsy. Because GBM cannot be cured surgically, the surgical goals are as follows:

In some cases, stereotactic biopsy is followed by radiation therapy (eg, for patients with a tumor located in an eloquent area of the brain, patients whose tumors have minimal mass effect, and patients in poor medical condition who cannot undergo general anesthesia). The extent of surgery (biopsy vs resection) has been shown in a number of studies to affect length of survival. 

See Treatment and Medication for more detail.

For patient education resources, see the Cancer Center as well as the patient education article Brain Cancer.

Background

Glioblastoma multiforme (GBM) is by far the most common and most malignant of the glial tumors. Attention has been drawn to this form of brain cancer by the deaths of Senator Ted Kennedy and Senator John McCain from glioblastoma.

Of the estimated 17,000 primary brain tumors diagnosed in the United States each year, approximately 60% are gliomas. Gliomas comprise a heterogeneous group of neoplasms that differ in location within the central nervous system, in age and sex distribution, in growth potential, in extent of invasiveness, in morphological features, in tendency for progression, and in response to treatments.

See images below.



View Image

A T1-weighted axial MRI without intravenous contrast. This image demonstrates a hemorrhagic multicentric tumor (glioblastoma multiforme [GBM]) in the ....



View Image

A T1-weighted sagittal MRI with intravenous contrast in a patient with glioblastoma multiforme (GBM).

Composed of a heterogeneous mixture of poorly differentiated neoplastic astrocytes, glioblastomas primarily affect adults, and they are located preferentially in the cerebral hemispheres. Much less commonly, glioblastoma multiforme can affect the brainstem (especially in children) and the spinal cord. These tumors may develop from lower-grade astrocytomas (World Health Organization [WHO] grade II) or anaplastic astrocytomas (WHO grade III), but more frequently they manifest de novo, without any evidence of a less malignant precursor lesion. The treatment of glioblastomas is palliative and includes surgery, radiotherapy, and chemotherapy.[27, 28, 29]

Pathophysiology

Glioblastomas can be classified as primary or secondary. Primary glioblastoma multiforme accounts for the vast majority of cases (60%) in adults older than 50 years. These tumors manifest de novo (ie, without clinical or histopathologic evidence of a preexisting, less-malignant precursor lesion), and patients presenting after a short clinical history, usually less than 3 months.

Secondary glioblastoma multiformes (40%) typically develop in younger patients (< 45 y) through malignant progression from a low-grade astrocytoma (WHO grade II) or anaplastic astrocytoma (WHO grade III). The time required for this progression varies considerably, ranging from less than 1 year to more than 10 years, with a mean interval of 4-5 years. Increasing evidence indicates that primary and secondary glioblastomas constitute distinct disease entities that evolve through different genetic pathways, affect patients at different ages, and differ in response to some of the present therapies. Of all the astrocytic neoplasms, glioblastomas contain the greatest number of genetic changes, which, in most cases, result from the accumulation of multiple mutations.

Over the past decade, the concept of different genetic pathways leading to the common phenotypic endpoint (ie, glioblastoma multiforme) has gained general acceptance. Genetically, primary and secondary glioblastomas show little overlap and constitute different disease entities. Studies are beginning to assess the prognoses associated with different mutations. Some of the more common genetic abnormalities are described as follows:

Eckel-Passow and colleagues classified gliomas into groups on the basis of three tumor markers: mutations in the TERT promoter, mutations in IDH, and codeletion of chromosome arms 1p and 19q (1p/19q codeletion). The groups had different ages at onset, overall survival, and associations with germline variants, which implies that they are characterized by distinct mechanisms of pathogenesis. Findings included the following[37] :

Less frequent but more malignant mutations in glioblastomas include the following:

Additional genetic alterations in primary glioblastomas include p16 deletions (30-40%), p16INK4A, and retinoblastoma (RB) gene protein alterations. Progression of secondary glioblastomas often includes LOH at chromosome arm 19q (50%), RB protein alterations (25%), PTEN mutations (5%), deleted-in-colorectal-carcinoma gene (DCC) gene loss of expression (50%), and LOH at 10q.

Glioblastoma multiformes occur most often in the subcortical white matter of the cerebral hemispheres. In a series of 987 glioblastomas from University Hospital Zurich, the most frequently affected sites were the temporal (31%), parietal (24%), frontal (23%), and occipital (16%) lobes.[38] Combined frontotemporal location is particularly typical.

Tumor infiltration often extends into the adjacent cortex or the basal ganglia. When a tumor in the frontal cortex spreads across the corpus callosum into the contralateral hemisphere, it creates the appearance of a bilateral symmetric lesion, hence the term butterfly glioma. Sites for glioblastomas that are much less common are the brainstem (which often is found in affected children), the cerebellum, and the spinal cord.

Etiology

The etiology of glioblastoma remains unknown in most cases. Familial gliomas account for approximately 5% of malignant gliomas, and less than 1% of gliomas are associated with a known genetic syndrome (eg, neurofibromatosis, Turcot syndrome, or Li-Fraumeni syndrome).[1]

Although concerns have been raised regarding cell phone use as a potential risk factor for development of gliomas, study results have been inconsistent, and this possibility remains controversial. The largest studies have not supported cell phone use as a cancer risk factor.[39, 2, 3, 4, 5, 6]  

Studies of association with head injury, N-nitroso compounds, occupational hazards, and electromagnetic field exposure have been inconclusive.[39]

Epidemiology

Overall incidence is very similar among countries. Glioblastoma multiformes are slightly more common in the United States, Scandinavia, and Israel than in Asia. This may reflect differences in genetics, diagnosis and the healthcare system, and reporting practices. Glioblastoma multiforme is the most frequent primary brain tumor, accounting for approximately 12-15% of all intracranial neoplasms and 50-60% of all astrocytic tumors. In most European and North American countries, incidence is approximately 2-3 new cases per 100,000 people per year.

Within the United States, glioblastoma multiforme is slightly more common in whites.

In a review of 1003 glioblastoma biopsies from the University Hospital Zurich,[40]  males had a slight preponderance over females, with a male-to-female ratio of 3:2.

Glioblastoma multiforme may manifest in persons of any age, but it affects adults preferentially, with a peak incidence at 45-70 years. In the series from University Hospital Zurich (a review of 1003 glioblastoma biopsies), 70% of patients were in this age group, with a mean age of 53 years.[40]  In a series reported by Dohrman (1976), only 8.8% of glioblastoma multiformes occurred in children.[41]

Prognosis

Only modest advancements in the treatment of glioblastoma have occurred in the past 25 years. Although current therapies remain palliative, they have been shown to prolong quality survival. Mean survival is inversely correlated with age, which may reflect exclusion of older patients from clinical trials. Without therapy, patients with glioblastoma multiformes uniformly die within 3 months. Patients treated with optimal therapy, including surgical resection, radiation therapy, and chemotherapy, have a median survival of approximately 12 months, with fewer than 25% of patients surviving up to 2 years and fewer than 10% of patients surviving up to 5 years. Whether the prognosis of patients with secondary glioblastoma is better than or similar to the prognosis for those patients with primary glioblastoma remains controversial.

Brain tumor resection has an overall mortality rate of 1-2%.  Approximately 40% of patients have no or minimal deficits after surgery, 30% manifest no postoperative change relative to preoperative deficits, and 25% sustain an increased postoperative deficit that usually improves.

Despite extensive clinical trials, individual prediction of clinical outcome has remained an elusive goal. Glioblastomas are among the most malignant human neoplasms, with a median survival despite optimal treatment of less than 1 year. In a series of 279 patients receiving aggressive radiation and chemotherapy, only 5 of 279 patients (1.8%) survived longer than 3 years.[42]

Patient survival depends on a variety of clinical parameters. Younger age, higher Karnofsky performance scale (a standard measure of the ability of patients with cancer to perform daily tasks) score at presentation, radiotherapy, and chemotherapy all correlate with improved outcome. Clinical evidence also suggests that a greater extent of resection favors longer survival.[43, 44, 45, 46] Tumors that are deemed unresectable due to location (eg, in the brainstem) also portend a poorer prognosis.[47]

A review by Perrini et al of 48 patients with recurrent glioblastoma found that preoperative performance status at recurrence and subtotal versus gross-total repeat resection were independent predictors of survival. These authors concluded that gross-total resection at repeat craniotomy is associated with longer overall survival and should be performed whenever possible in patients with recurrent glioblastoma who have good performance status.[48]

Survival has not been shown to correlate with p53, EGFR, or MDM2 mutations.[49]

Two separate reviews of outcomes in elderly patients have been published. One found that although elderly patients have a poor prognosis, gross-total resection confers a modest survival benefit and treatment with bevacizumab significantly increased overall survival. Older age and preoperative Karnofsky Performance Scale score also were significant prognostic factors.[50]

The results of the second study concurred that there is a survival advantage for those who undergo maximal safe resection. The review also found that radiotherapy extends survival in selected patients and temozolomide chemotherapy is safe and extends the survival of patients with tumors that harbor O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation.[51]

A study by Li et al used an updated Radiation Therapy Oncology Group (RTOG) GBM database to produce a simplified original recursive partitioning analysis (RPA) model combining classes V and VI. This resulted in 3 distinct prognostic groups defined by performance status, age, neurologic function, and extent of resection. This classification will be used in future RTOG GBM trials.[52]

Clearly, new approaches for the management of glioblastomas are necessary. Enrollment of patients into clinical trials will generate new information regarding investigational therapies. Novel approaches, such as the use of gene therapy and immunotherapy, as well as improved methods for the delivery of antiproliferative, antiangiogenic, and noninvasive therapies, provide hope for the future.

A study by Kaur et al determined that the presence of a large cyst in patients with GBM does not affect overall survival compared with those who do not have a cyst.[53]

Patient Education

For patient education information, see the Brain Cancer Health Center. In addition, information about glioblastoma (and other brain tumors) is available from the American Brain Tumor Association (ABTA) at Brain Tumor Information.

History

The clinical history of patients with glioblastoma multiformes (GBMs) usually is short, spanning less than 3 months in more than 50% of patients, unless the neoplasm developed from a lower-grade astrocytoma. Note the following:

Physical Examination

Neurologic symptoms and signs affecting patients with glioblastomas can be either general or focal and reflect the location of the tumor. General symptoms include headaches, nausea and vomiting, personality changes, and slowing of cognitive function. Note the following:

Laboratory Studies

Currently, no specific laboratory studies are helpful in making a diagnosis of glioblastoma.

Response to adjuvant therapy may be predicted based on the tumor's genetics.

Imaging Studies

Imaging studies of the brain are essential to make the diagnosis of glioblastoma multiforme (GBM). For complete discussion, see Imaging in Glioblastoma Multiforme.

On CT scans, glioblastomas usually appear as irregularly shaped hypodense lesions with a peripheral ringlike zone of contrast enhancement and a penumbra of cerebral edema.

MRI with and without contrast is the study of choice (see the images below). These lesions typically have an enhancing ring observed on T1-weighted images and a broad surrounding zone of edema apparent on T2-weighted images. The central hypodense core represents necrosis, the contrast-enhancing ring is composed of highly dense neoplastic cells with abnormal vessels permeable to contrast agents, and the peripheral zone of nonenhancing low attenuation is vasogenic edema containing varying numbers of invasive tumor cells. Several pathological studies have clearly shown that the area of enhancement does not represent the outer tumor border because infiltrating glioma cells can be identified easily within, and occasionally beyond, a 2-cm margin.[7]



View Image

A T1-weighted axial MRI without intravenous contrast. This image demonstrates a hemorrhagic multicentric tumor (glioblastoma multiforme [GBM]) in the ....



View Image

A T1-weighted axial MRI with intravenous contrast. Heterogenous enhancement of the lesion is present within the right temporal lobe. The hypointensity....



View Image

A T1-weighted coronal MRI with intravenous contrast. This image demonstrates the lesion (glioblastoma multiforme [GBM]) within the medial temporal lob....



View Image

A T1-weighted sagittal MRI with intravenous contrast in a patient with glioblastoma multiforme (GBM).



View Image

A T2-weighted axial MRI. The tumor (glioblastoma multiforme [GBM]) and surrounding white matter within the right temporal lobe show increased signal i....



View Image

A fluid-attenuated inversion recovery (FLAIR) axial MRI. This image is similar to the T2-weighted image and demonstrates extensive edema in a patient ....

Positron emission tomography (PET) scans and magnetic resonance (MR) spectroscopy can be helpful to identify glioblastomas in difficult cases, such as those associated with radiation necrosis or hemorrhage. On PET scans, increased regional glucose metabolism closely correlates with cellularity and reduced survival. MR spectroscopy demonstrates an increase in the choline-to-creatine peak ratio, an increased lactate peak, and decreased N- acetylaspartate (NAA) peak in areas with glioblastomas (see the image below).



View Image

Magnetic resonance (MR) spectroscopy is representative of a glioblastoma multiforme (GBM), demonstrating a high peak ratio of choline (CHO) to creatin....

A study by Piroth et al found that O-(2-[(18)F]fluoroethyl-l-tyrosine (FET) PET to measure tumor volume after surgery has a strong prognostic impact.[8]

Cerebral angiograms are not necessary for the diagnosis or clinical management of glioblastomas.

Other Tests

Electroencephalography (EEG) performed on a patient with glioblastoma multiforme may show generalized diffuse slowing and/or epileptogenic spikes over the area of the tumor. However, findings specific for glioblastoma cannot be observed on EEG.

Procedures

Lumbar puncture is generally contraindicated in the setting of a brain tumor because of the possibility of transtentorial herniation with increased intracranial pressure. However, if ruling out lymphoma, it may be necessary.

CSF studies do not aid significantly in the specific diagnosis of glioblastoma multiforme.

Histologic Findings

As its name suggests, the histopathology of glioblastoma multiforme is extremely variable. Glioblastoma multiformes are composed of poorly differentiated, often pleomorphic astrocytic cells with marked nuclear atypia and brisk mitotic activity. Necrosis is an essential diagnostic feature, and prominent microvascular proliferation is common. Macroscopically, glioblastomas are poorly delineated, with peripheral grayish tumor cells, central yellowish necrosis from myelin breakdown, and multiple areas of old and recent hemorrhages. Most glioblastomas of the cerebral hemispheres are clearly intraparenchymal with an epicenter in the white matter, but some extend superficially and contact the leptomeninges and dura.[9, 10, 11, 12, 13, 14, 15]

Despite the short duration of symptoms, these tumors are often surprisingly large at the time of presentation, occupying much of a cerebral lobe. Undoubtedly, glial fibrillary acidic protein (GFAP) remains the most valuable marker for neoplastic astrocytes. Although immunostaining is variable and tends to decrease with progressive dedifferentiation, many cells remain immunopositive for GFAP even in the most aggressive glioblastomas. Vimentin and fibronectin expression are common but less specific.[54]

The regional heterogeneity of glioblastomas is remarkable and makes histopathological diagnosis a serious challenge when it is based solely on stereotactic needle biopsies. Tumor heterogeneity is also likely to play a significant role in explaining the meager success of all treatment modalities, including radiation, chemotherapy, and immunotherapy.



View Image

Histopathologic slide demonstrating a glioblastoma multiforme (GBM).

Staging

Completely staging most glioblastomas is neither practical nor possible because these tumors do not have clearly defined margins. Rather, they exhibit well-known tendencies to invade locally and spread along compact white matter pathways, such as the corpus callosum, internal capsule, optic radiation, anterior commissure, fornix, and subependymal regions. Such spread may create the appearance of multiple glioblastomas or multicentric gliomas on imaging studies.

Careful histological analyses have indicated that only 2-7% of glioblastomas are truly multiple independent tumors rather than distant spread from a primary site. Despite its rapid infiltrative growth, the glioblastoma tends not to invade the subarachnoid space and, consequently, rarely metastasizes via cerebrospinal fluid (CSF). Hematogenous spread to extraneural tissues is very rare in patients who have not had previous surgical intervention, and penetration of the dura, venous sinuses, and bone is exceptional.[16, 17, 18, 19, 20, 21]

Approach Considerations

The treatment of glioblastomas remains difficult in that no contemporary treatments are curative.[22]  While overall mortality rates remain high, recent work leading to an understanding of the molecular mechanisms and gene mutations combined with clinical trials are leading to more promising and tailored therapeutic approaches. Multiple challenges remain, including tumor heterogeneity, tumor location in a region where it is beyond the reach of local control, and rapid, aggressive tumor relapse. Therefore, the treatment of patients with malignant gliomas still remains palliative and encompasses surgery, radiotherapy, and chemotherapy. See Brain Cancer Treatment Protocols for summarized information.

Upon initial diagnosis of glioblastoma multiforme (GBM), standard treatment consists of maximal surgical resection, radiotherapy, and concomitant and adjuvant chemotherapy with temozolomide.[23, 24]  At some institutions, transferring the patient to another facility may be necessary if the proper consultations cannot be obtained. In most cases, surgical resection can be performed on an urgent, but not emergent, basis. Patients with glioblastomas who undergo surgical resection typically spend the night after surgery in an intensive care unit, followed by an inpatient stay of 3-5 days. The final length of stay depends on each patient's neurological condition.

Postoperative antibiotics usually are continued for 24 hours, and deep vein thrombosis prophylaxis is continued until patients are ambulatory. Anticonvulsants are maintained at therapeutic levels throughout the inpatient stay, while steroids are reduced gradually, tailored to each patient's clinical status. Many patients benefit from occupational therapy and physical therapy or rehabilitation.

While patients are in the hospital, they should receive postoperative imaging to determine the extent of surgical resection. Surgical resection is evaluated best within 3 days of surgery by using contrast-enhanced MRI. Contrast enhancement during this period accurately reflects residual tumor. If not performed preoperatively, complete evaluations by consulting physicians, including a neurooncologist and radiation oncologist, should be considered postoperatively.

For patients older than 70 years, less aggressive therapy is sometimes employed, using radiation or temozolomide alone.[25, 26, 27]  A study by Scott et al found that elderly patients with glioblastoma who underwent radiotherapy had improved cancer-specific survival and overall survival compared with those who did not undergo radiotherapy treatment.[28]

Evidence suggests that in patients over 60 years old, treatment with temozolomide is associated with longer survival than treatment with standard radiotherapy, and for those over 70 years old, temozolomide or hypofractionated radiotherapy is associated with prolonged survival than treatment with standard fractionated radiotherapy. The improvement in survival with temozolomide is enhanced in patients with MGMT promoter methylation.[29]  Data from a a randomised phase 3 trial suggests that lomustine-temozolomide plus radiotherapy might be superior to temozolomide chemoradiotherapy in newly diagnosed glioblastoma with methylation of the MGMT promoter.[55]

Stupp et al reported the final results of the randomized phase III trial for patients with glioblastoma who were treated with adjuvant temozolomide and radiation with a median follow-up of more than 5 years. Stupp et al previously reported improved median and 2-year survival when temozolomide was added to radiation therapy in glioblastoma. Survival in the combined therapy group (ie, temozolomide and radiation) continued to exceed that of radiation alone throughout the 5-year follow-up (p< 0.0001). Survival of patients who received adjuvant temozolomide with radiotherapy for glioblastoma is superior to radiotherapy alone across all clinical prognostic subgroups.[56]   

Median time to recurrence after standard therapy is 6.9 months.[57]  For recurrent glioblastoma multiforme, surgery is appropriate in selected patients, and various radiotherapeutic, chemotherapeutic, biologic, or experimental therapies are also employed.[58, 35]

Approximately 90% of glioblastomas express cytomegalovirus (CMV) proteins, and Batich et al have reported benefit with a dendritic cell vaccine targeting CMV antigen pp65, using CMV as a proxy for glioblastoma. Patients are first treated with dose-intensified temozolomide, as the temozolomide induces lymphopenia, which provides an opportunity to retrain the immune system.

In a study of 11 patients with newly diagnosed glioblastoma received temozolomide, 100 mg/m2/d × 21 days per cycle, and at least three pp65-directed vaccines admixed with granulocyte-macrophage colony-stimulating factor on day 23 ± 1 of each cycle. Despite increased proportions of regulatory T cells (Tregs), median progression-free survival was 25.3 months and overall survival was 41.1 months; three patients remained progression-free more than 7 years after diagnosis.[59]

Wernicke et al report that prostate-specific membrane antigen is expressed in the vasculature of GBM vessels and represents a potential novel therapeutic vascular target. Future clinical trials are planned.[60]

Medical Care

In an evidence-based clinical practice guideline formulated to address the impact of cytotoxic chemotherapy on disease control and survival in adults with progressive glioblastoma, Olson et al make the following recommendations[61] :

Anticonvulsant medications are usually maintained, and levels are checked intermittently. Steroids are tapered to lower doses for radiation therapy and then tapered further if possible. While taking steroids, patients should be maintained on an antiulcer agent.

Radiation therapy [62, 63, 64, 65]

Radiation therapy in addition to surgery or surgery combined with chemotherapy has been shown to prolong survival in patients with glioblastoma multiformes compared to surgery alone. The addition of radiotherapy to surgery has been shown to increase survival from 3-4 months to 7-12 months.[57, 66]

Dose response relationships for glioblastomas demonstrate that a radiation dose of less than 4500 cGy results in a median survival of 13 weeks compared with a median survival of 42 weeks with a dose of 6000 cGy. This is usually administered 5 days per week in doses of 1.8-2.0 Gy.

Jablonska et al reported that in patients with poor clinical factors other than advanced age, the combination of hypofractionated radiation therapy and temozolomide produced results comparable to those seen with standard fractionation. In the 17 patients in the study, poor clinical factors included postoperative neurological complications, high tumor burden, unresectable or multifocal lesions, and potential low treatment compliance due to social factors or rapidly progressive disease. Patients received 40, 45, and 50 Gy in 15 fractions to 95% of the planning target volume (PTV), clinical target volume (CTV), and gross tumor volume (GTV), respectively. Treatment was delivered using intensity-modulated radiation therapy (IMRT) or volumetric modulated arc therapy (VMAT).[67]

The responsiveness of glioblastoma multiformes to radiotherapy varies. In many instances, radiotherapy can induce a phase of remission, often marked with stability or regression of neurologic deficits as well as diminution in the size of the contrast-enhancing mass. Unfortunately, any period of response is short-lived because the tumor typically recurs within 1 year, resulting in further clinical deterioration and the appearance of an expansile region of contrast enhancement.[68, 69]

Two studies investigated tumor recurrence after whole-brain radiation therapy and found that the tumor recurred within 2 cm of the original site in 90% and 78% of patients, supporting the use of focal radiation therapy. Multifocal recurrence occurred in 6% of patients in one study and in 5% of patients in a second trial.

Interstitial brachytherapy is of limited use and is rarely used. By implantation of radioactive seeds, a large dose of radiation is delivered to the tumor volume, with rapid fall-off of radiation in surrounding tissue. The tumor must be unilateral and smaller than 5 cm in diameter. In one study, patients treated with interstitial brachytherapy had a significantly better median survival (2 mo) compared with the conventional focal external beam radiation therapy. Following interstitial brachytherapy, up to 40% of patients require another surgery for removal of tissue damaged by radiation necrosis.[70]

Experimental studies are underway in which focal radiation is delivered directly to tumors through an implanted balloon containing interstitial radiation. MRI and MR spectroscopy can be used to monitor therapy. Clinical outcomes from these studies are not yet available.

Radiosensitizers, such as newer chemotherapeutic agents,[71] targeted molecular agents,[40, 41] and antiangiogenic agents[41] may increase the therapeutic effect of radiotherapy.[72]

Radiotherapy for recurrent glioblastoma multiforme is controversial, though some studies have suggested a benefit to stereotactic radiosurgery or fractionated stereotactic reirradiation.[73, 74, 75]  In adult patients with progressive glioblastoma, American Association of Neurological Surgeons/Congress of Neurological Surgeons (AANS/CNS) guidelines recommend that when the target tumor is amenable for additional radiation, re-irradiation should be performed to improve local tumor control. This re-irradiation may take the form of conventional fractionation radiotherapy, fractionated radiosurgery, or single fraction radiosurgery.[76]

Fleischmann et al reported that in patients undergoing re-irradiation for recurrent glioblastoma, concomitant treatment with bevacizumab significantly reduced the rate of radiation toxicity, both in the short and the long term. Bevacizumab was given in a dose of 10 mg/kg on days 1 and 15 of re-irradiation therapy.[77]

Chemotherapy – Antineoplastic agents [78, 79, 80, 81, 82, 83]

Although the optimal chemotherapeutic regimen for glioblastoma is not defined at present, several studies have suggested that more than 25% of patients obtain a significant survival benefit from adjuvant chemotherapy. Meta-analyses have suggested that adjuvant chemotherapy results in a 6-10% increase in 1-year survival rate.[84, 85]

Temozolomide is an orally active alkylating agent that is used for persons newly diagnosed with glioblastoma multiforme. It was approved by the United States Food and Drug Administration (FDA) in March 2005. Studies have shown that the drug was well tolerated and provided a survival benefit. Adjuvant and concomitant temozolomide with radiation was associated with significant improvements in median progression-free survival over radiation alone (6.9 vs 5 mo), overall survival (14.6 vs 12.1 mo), and the likelihood of being alive in 2 years (26% vs 10%).

Nitrosoureas: BCNU (carmustine)-polymer wafers (Gliadel) were approved by the FDA in 2002. Though Gliadel wafers are used by some for initial treatment, they have shown only a modest increase in median survival over placebo (13.8 vs. 11.6 months) in the largest such phase III trial, and are associated with increased rates of CSF leak and increased intracranial pressure secondary to edema and mass effect.[86, 87]

MGMT is a DNA repair enzyme that contributes to temozolomide resistance. Methylation of the MGMT promoter, found in approximately 45% of glioblastoma multiformes, results in an epigenetic silencing of the gene, decreasing the tumor cell's capacity for DNA repair and increasing susceptibility to temozolomide.[88] Note the following:

Carmustine (BCNU) and cis -platinum (cisplatin) have been the primary chemotherapeutic agents used against malignant gliomas. All agents in use have no greater than a 30-40% response rate, and most fall into the range of 10-20%.

Data from the University of California at San Francisco indicate that, for the treatment of glioblastomas, surgery followed by radiation therapy leads to 1-, 3-, and 5-year survival rates of 44%, 6%, and 0%, respectively. By comparison, surgery followed by radiation and chemotherapy using nitrosourea-based regimens resulted in 1-, 3-, and 5-year survival rates of 46%, 18%, and 18%, respectively.

A major hindrance to the use of chemotherapeutic agents for brain tumors is the fact that the blood-brain barrier (BBB) effectively excludes many agents from the CNS. For this reason, novel methods of intracranial drug delivery are being developed to deliver higher concentrations of chemotherapeutic agents to the tumor cells while avoiding the adverse systemic effects of these medications.

Pressure-driven infusion of chemotherapeutic agents through an intracranial catheter, also known as convection-enhanced delivery (CED), has the advantage of delivering drugs along a pressure gradient rather than by simple diffusion. CED has shown promising results in animal models with agents including BCNU and topotecan.[91, 92, 93]

Initial attempts investigated the delivery of chemotherapeutic agents via an intraarterial route rather than intravenously. Unfortunately, no survival advantage was observed.

Chemotherapy for recurrent glioblastoma multiforme provides modest, if any, benefit, and several classes of agents are used. Carmustine wafers increased 6-month survival from 36% to 56% over placebo in one randomized study of 222 patients, though there was a significant association between the treatment group and serious intracranial infections.[94, 95]

Genotyping of brain tumors may have applications in stratifying patients for clinical trials of various novel therapies.

The anti-angiogenic agent bevacizumab was approved by the U.S. Food and Drug Administration for recurrent glioblastoma in May 2009.[96] When used with irinotecan, bevacizumab improved 6-month survival in recurrent glioma patients to 46% compared with 21% in patients treated with temozolomide.[97, 98] This bevacizumab and irinotecan combination for recurrent glioblastoma multiforme has been shown to improve survival over bevacizumab alone.[99] Anti-angiogenic agents also decrease peritumoral edema, potentially reducing the necessary corticosteroid dose.

A small proportion of glioblastomas responds to gefitinib or erlotinib (tyrosine kinase inhibitors). The simultaneous presence in glioblastoma cells of mutant EGFR (EGFRviii) and PTEN was associated with responsiveness to tyrosine kinase inhibitors, whereas increased p-akt predicts a decreased effect.[100, 101, 102] Other targets include PDGFR, VEGFR, mTOR, farnesyltransferase, and PI3K.

Other therapy modalities under investigation include gene therapy, peptide and dendritic cell vaccines, synthetic chlorotoxins, and radiolabeled drugs and antibodies.[103, 104, 105, 106, 107, 108]

A population-based analysis of 5607 adult patients with glioblastoma in the SEER (Surveillance Epidemiology and End Results) database found that bevacizumab therapy may improve survival. In the study, glioblastoma patients who died in 2010 (after the FDA approved bevacizumab for this condition) survived significantly longer than those who died of the disease in 2008. Median survival was 8 months for patients who died in 2006, 7 months in 2008, and 9 months in 2010. This difference in survival was highly significant between 2008 (pre-bevacizumab) and 2010 (post-bevacizumab). This survival difference was unlikely due to improvements in supportive care during this time interval, because there was no significant difference between those who died in 2006 and patients who died 2 years later, in 2008.[109, 110]

Electric-field therapy

The Optune device uses low-intensity, intermediate-frequency, alternating electric fields (tumor- treating fields) to target dividing cells in glioblastoma multiforme while generally not harming normal cells. The tumor-treating fields are generated via electrodes placed directly on the scalp. To target the tumor, array placement is based on the individual patient's magnetic resonance imaging results.[111]

Optune, also known as the NovoTTF-100A System, was initially approved in 2011 for use in glioblastoma multiforme that had recurred or progressed after treatment. In October 2015, the FDA expanded approval to include use of the device in conjunction with temozolomide chemotherapy in the first-line setting. Approval was based on an open-label, randomized phase 3 trial in 700 patients, in which median overall survival was 19.4 months with use of the device plus temozolomide, versus 16.6 months with chemotherapy only.[111]

In a randomized, open-label trial in 695 patients with glioblastoma, the addition of  tumor-treating fields to treatment with temozolomide improved median progression-free survival from 4.0 months to 6.7 months (hazard ratio [HR], 0.63; 95% confidence index [CI], 0.52-0.76; P < 0.001). Median overall survival improved from 16.0 months to 20.9 months (HR, 0.63; 95% CI, 0.53-0.76; P <  0.001).[112, 113]

Modified polio vaccine therapy

The poliovirus receptor CD155 is broadly upregulated on the surface of malignant solid tumors, and a preliminary study of intratumoral infusion of a modified poliovirus vaccine has demonstrated benefit in some cases of recurrent  malignant glioma. In a dose-finding and toxicity study, 61 patients with recurrent supratentorial WHO grade IV malignant glioma received seven doses of a live attenuated poliovirus type 1 vaccine with its cognate internal ribosome entry site replaced with that of human rhinovirus type 2. The recombinant nonpathogenic polio–rhinovirus chimera was infused into the glioma via an implanted catheter.[114]

In contrast to overall survival rates in a historical control group, which declined steadily to 14% at 24 months and 4% at 36 months, overall survival in the study patients stabilized at 21% at 24 months, remaining at that rate through 36 months. Adverse events that affected more than 20% of the study patients in the dose-expansion phase included headache (52%), hemiparesis (50%), seizure (45%), dysphasia (28%), and cognitive disturbance (25%).[114]

Surgical Care

Oral aminolevulinic acid (ALA; Gleolan) was approved by the FDA in June 2017 as an adjunct for visualization of malignant tissue during surgery in patients with malignant glioma (suspected WHO grades III or IV on preoperative imaging). During surgery, an operating microscope adapted with a blue-emitting light source and filters for excitation light of wavelength 375-440 nm, and observation at wavelengths of 620-710 nm is used to visualize PpIX (an ALA metabolite) accumulation in tumor cells that shows up as red fluorescence.[115]

Fluorescence-guided surgery (FGS), an emerging technology that combines detection devices with fluorescent contrast agents, may provide more complete and precise resection of gliomas. Tozuleristide (BLZ-100), a near-infrared imaging agent composed of the peptide chlorotoxin and a near-infrared fluorophore indocyanine green, is a candidate for FGS of glioma and other tumor types. In a phase 1 study, tozuleristide (BLZ-100) provided a viable fluorescence signal in both high- and low-grade glial tumors, but did not bind to normal tissues. Signal intensity in high-grade tumors was found to improve with increasing doses of tozuleristide, regardless of the time of dosing relative to surgery.[116, 117]

The extent of surgery (biopsy vs resection) has been shown in a number of studies to affect length of survival. In a study by Ammirati and colleagues (1987), patients with high-grade gliomas who had a gross total resection had a 2-year survival rate of 19%, while those with a subtotal resection had a 2-year survival rate of 0%.[118]

In another study of 416 patients, gross total resection, defined as >98% on MRI, conferred a survival advantage over subtotal resection (13 vs 8.8 mo).[46]

In another study of 92 patients, a total tumor resection without any residual disease resulted in a median survival of 93 weeks, whereas the smallest percent of resection (< 25%) and greatest volume of residual tumor (>20 cm3) gradually shortened the survival to 31 weeks and 50 weeks, respectively.[45]

An analysis of 28 studies found a mean duration of survival advantage of total over subtotal resection for glioblastoma multiforme (14 vs 11 mo).[44, 119]

Li and colleagues compared the survival of patients having 100% removal of the contrast-enhancing tumor, with or without additional resection of the surrounding FLAIR abnormality region to that of patients undergoing 78% to < 100% extent of resection of the enhancing mass. The median survival time for patients acheiving complete resection (15.2 months) was significantly longer than that for patients undergoing less than complete resection (9.8 months; p < 0.001). The patients who underwent resection of ≥ 53.21% of the surrounding FLAIR abnormality beyond the 100% resection achieved significant prolongation of survival (median survival times 20.7).[120]

Because these tumors cannot be cured with surgery, the surgical goals are to establish a pathological diagnosis, relieve mass effect, and, if possible, achieve a gross total resection to facilitate adjuvant therapy.[121] Most glioblastomas recur in and around the original tumor bed, but contralateral and distant recurrences are not uncommon, especially with lesions near the corpus callosum. The indications for reoperation of malignant astrocytomas after initial treatment with surgery, radiation therapy, and chemotherapy are not firmly established. Reoperation is generally considered in the face of a life-threatening recurrent mass, particularly if radionecrosis rather than recurrent tumor is suspected as the cause of clinical and radiographic deterioration. PET scans and MR spectroscopy have proven useful in discriminating between these 2 entities.

See the images below.



View Image

Axial CT scan without intravenous contrast. This image reveals a large right temporal intraaxial mass (glioblastoma multiforme [GBM]). Extensive surro....



View Image

A T1-weighted axial MRI without intravenous contrast. This image demonstrates a hemorrhagic multicentric tumor (glioblastoma multiforme [GBM]) in the ....



View Image

A T1-weighted axial MRI with intravenous contrast. Heterogenous enhancement of the lesion is present within the right temporal lobe. The hypointensity....



View Image

A T1-weighted coronal MRI with intravenous contrast. This image demonstrates the lesion (glioblastoma multiforme [GBM]) within the medial temporal lob....



View Image

A T1-weighted sagittal MRI with intravenous contrast in a patient with glioblastoma multiforme (GBM).



View Image

A T2-weighted axial MRI. The tumor (glioblastoma multiforme [GBM]) and surrounding white matter within the right temporal lobe show increased signal i....



View Image

A fluid-attenuated inversion recovery (FLAIR) axial MRI. This image is similar to the T2-weighted image and demonstrates extensive edema in a patient ....



View Image

Histopathologic slide demonstrating a glioblastoma multiforme (GBM).

Although no formal studies have been performed, observations indicate that variables, such as young age, prolonged interval between operations, and extent of the second surgical resection, have prognostic significance.[122]

A study by El Hindy et al found that a common regulatory single-nucleotide polymorphism (-938C>A) is a survival prognosticator and a marker for high-risk in patients with glioblastoma multiforme who undergo gross total resection.[123]

Stereotactic biopsy followed by radiation therapy may be considered in certain circumstances. These include patients with a tumor located in an eloquent area of the brain, patients whose tumors have minimal mass effect, and patients in poor medical condition, precluding general anesthesia. Median survival after stereotactic biopsy and radiation therapy is reported to be from 27-47 weeks.[124]

A study by Jakola et al found that surgical procedures may not significantly alter the quality of life (QOL) in the average patient, however, the use of intraoperative ultrasonography may be associated with a preservation of QOL in that it helps avoid introducing new deficits.[125]

Consultations

Patients with glioblastomas should be evaluated by a team of specialists, including a neurologist, neurosurgeon, neurooncologist, and radiation oncologist, in order to develop a coordinated treatment strategy.

Activity

No universal restrictions on activity are necessary for patients with glioblastomas. The patient's activity depends on his or her overall neurologic status. The presence of seizures may prevent the patient from driving. In many circumstances, physical therapy and/or rehabilitation are extremely beneficial. Activity is encouraged to reduce the risk of deep venous thrombosis.

Guidelines Summary

The National Comprehensive Cancer Network (NCCN) has released guidelines on central nervous system (CNS) cancers which includes recommendations for the diagnosis and treatment of glioblastomas (grade IV gliomas). The goals of surgery are to obtain a diagnosis, alleviate symptoms of increased intracrainial pressure or compression, increase survival, and decrease the need for corticosteroids. Adjuvant treatment options depend on the patient performance status (PS), age and MGMT promoter methylation status.[126]  

Category 1 recommendations for first-line treatment[126] :

Medication Summary

No specific medications exist to treat glioblastomas. However, certain conditions require medical treatment. For seizures, the patient usually is started on levetiracetam (Keppra), phenytoin (Dilantin), or carbamazepine (Tegretol). Levetiracetam is often used because it lacks the effects on the P450 system seen with phenytoin and carbamazepine, which can interfere with antineoplastic therapy. Vasogenic cerebral edema is typically managed with corticosteroids (eg, dexamethasone), usually in combination with some form of antiulcer agent (eg, famotidine, ranitidine). The American Academy of Neurology's practice parameters state that prophylactic antiepileptic drugs (AEDs) should not be administered routinely to patients with newly diagnosed brain tumors (standard) and should be discontinued in the first postoperative week in patients who have not experienced a seizure.[127]

Temozolomide (Temodar)

Clinical Context:  Oral alkylating agent converted to MTIC at physiologic pH; 100% bioavailable; approximately 35% crosses the blood-brain barrier. Indicated for glioblastoma multiforme combined with radiotherapy. Significant overall survival improvement was demonstrated in patients treated with temozolomide and radiation compared with radiotherapy alone.

Carmustine (BiCNU)

Clinical Context:  Alkylates and cross-links DNA strands, inhibiting cell proliferation.

Cisplatin (Platinol)

Clinical Context:  Inhibits DNA synthesis and, thus, cell proliferation by causing DNA crosslinks and denaturation of double helix.

Erlotinib (Tarceva)

Clinical Context:  Pharmacologically classified as a human epidermal growth factor receptor type 1/epidermal growth factor receptor (HER1/EGFR) tyrosine kinase inhibitor. EGFR is expressed on the cell surface of normal cells and cancer cells. Indicated for locally advanced or metastatic non-small cell lung cancer after failure of at least one prior chemotherapy regimen.

Gefitinib (Iressa)

Clinical Context:  An anilinoquinazoline. Indicated as monotherapy to treat locally advanced or metastatic non-small cell lung cancer after failure of both platinum-based and docetaxel chemotherapies. The mechanism is not fully understood. Inhibits tyrosine kinases intracellular phosphorylation associated with transmembrane cell surface receptors.

Class Summary

Although the optimal chemotherapeutic regimen for glioblastoma is not yet defined, several studies have suggested significant survival benefit from adjuvant chemotherapy.

Levetiracetam (Keppra)

Clinical Context:  Used as adjunct therapy for partial seizures and myoclonic seizures. Also indicated for primary generalized tonic-clonic seizures. Mechanism of action is unknown.

Phenytoin (Dilantin)

Clinical Context:  Acts to block sodium channels and prevent repetitive firing of action potentials. As such, it is a very effective anticonvulsant. First-line agent in patients with partial and generalized tonic-clonic seizures.

Carbamazepine (Tegretol)

Clinical Context:  Like phenytoin, acts by interacting with sodium channels and blocking repetitive neuronal firing. First-line agent in patients with partial and tonic-clonic seizures. Serum levels should be checked and should be approximately 4-8 mcg/mL.

Class Summary

These agents are used to treat and prevent seizures.

Dexamethasone (Decadron)

Clinical Context:  Postulated mechanisms of action in brain tumors include reduction in vascular permeability, cytotoxic effects on tumors, inhibition of tumor formation, and decreased CSF production.

Class Summary

These agents reduce edema around the tumor, frequently leading to symptomatic and objective improvement.

What is glioblastoma multiforme (GBM)?What are common symptoms of glioblastoma multiforme (GBM)?What are the neurologic signs and symptoms of glioblastoma multiforme (GBM)?What causes glioblastoma multiforme (GBM)?How is glioblastoma multiforme (GBM) diagnosed?How is glioblastoma multiforme (GBM) treated?What is the role of radiotherapy in the treatment of glioblastoma multiforme (GBM)?What is the efficacy of chemotherapy for the treatment of glioblastoma multiforme (GBM)?Which medications are used in the treatment of glioblastoma multiforme (GBM)?What are surgical options for the treatment of glioblastoma multiforme (GBM)?What is glioblastoma multiforme (GBM)?What is the pathophysiology of glioblastoma multiforme (GBM)?What is the role of genetics in the pathophysiology of glioblastoma multiforme (GBM)?Which genetic tumor markers have been used to classify glioblastoma multiforme (GBM)?Which genetic abnormalities result in more malignant glioblastomas?Which genetic alterations are associated with primary glioblastomas?What is the pathophysiology of glioblastoma multiforme (GBM) in cerebral hemispheres?What is the etiology of glioblastoma multiforme (GBM)?What is the prevalence of glioblastoma multiforme (GBM)?What is the prognosis of glioblastoma multiforme (GBM)?What is included in patient education about glioblastoma multiforme (GBM)?How does the incidence of glioblastoma multiforme (GBM) vary geographically?What is the prevalence of glioblastoma multiforme (GBM)?What are the survival rates for glioblastoma multiforme (GBM)?Which clinical history findings are characteristic of glioblastoma multiforme (GBM)?What are physical findings characteristic of glioblastoma multiforme (GBM)?Which conditions should be considered in the differential diagnosis of glioblastoma multiforme (GBM)?What is the role of lab studies in the workup of glioblastoma multiforme (GBM)?What is the role of imaging studies in the diagnosis of glioblastoma multiforme (GBM)?What is the role of MRI in the diagnosis of glioblastoma multiforme (GBM)?What is the role of positron emission tomography (PET) scans and magnetic resonance (MR) spectroscopy in the diagnosis of glioblastoma multiforme (GBM)?What is the role of EEG in the workup of glioblastoma multiforme (GBM)?What is the role of lumbar puncture and CSF studies in the workup of glioblastoma multiforme (GBM)?Which histologic findings are characteristic of glioblastoma multiforme (GBM)?How is glioblastoma multiforme (GBM) staged?What are the treatment options for glioblastoma multiforme (GBM)?What is included in standard treatment of glioblastoma multiforme (GBM)?What is the role of temozolomide in the treatment of glioblastoma multiforme (GBM)?What is the role of antineoplastic chemotherapy in the treatment of glioblastoma multiforme (GBM)?What are the AANS/CNS clinical practice guidelines for use of cytotoxic chemotherapy in the treatment of glioblastoma multiforme (GBM)?What is the role of radiation therapy in the treatment of glioblastoma multiforme (GBM)?What is the role of interstitial brachytherapy in the treatment of glioblastoma multiforme (GBM)?Which radiation therapies for glioblastoma multiforme (GBM) are under investigation?What is the efficacy of radiotherapy for the treatment of recurrent glioblastoma multiforme (GBM)?What are the benfits of concomitant threatment with bevacizumab for patients undergoing re-irradiation for recurrent glioblastoma?What is the role of convection-enhanced delivery (CED) of chemotherapy in the treatment of glioblastoma multiforme (GBM)?What is the role of chemotherapy for treatment of recurrent glioblastoma multiforme (GBM)?Which therapies are being investigated for the treatment of glioblastoma multiforme (GBM)?What is the role of electric-field therapy for the treatment of glioblastoma multiforme (GBM)?What aids are used to visualize glioblastoma multiforme (GBM) during surgery?What are survival rates for surgery for glioblastoma multiforme (GBM)?What are the goals for surgery in glioblastoma multiforme (GBM)?Which factors have prognostic significance in glioblastoma multiforme (GBM)?What is the role of stereotactic biopsy in the treatment of glioblastoma multiforme (GBM)?Which specialist consultations are needed for the treatment of glioblastoma multiforme (GBM)?What are activity restrictions for patients with glioblastoma multiforme (GBM)?Which medications are used in the treatment of glioblastoma multiforme (GBM)?Which medications in the drug class Corticosteroids are used in the treatment of Glioblastoma Multiforme?Which medications in the drug class Anticonvulsants are used in the treatment of Glioblastoma Multiforme?Which medications in the drug class Antineoplastic agents are used in the treatment of Glioblastoma Multiforme?

Author

Jeffrey N Bruce, MD, Edgar M Housepian Professor of Neurological Surgery Research, Vice-Chairman and Professor of Neurological Surgery, Director of Brain Tumor Tissue Bank, Director of Bartoli Brain Tumor Laboratory, Department of Neurosurgery, Columbia University College of Physicians and Surgeons

Disclosure: Received grant/research funds from NIH for other.

Coauthor(s)

Benjamin C Kennedy, MD, Assistant Professor of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania; Director of Epilepsy and Functional Neurosurgery, The Children’s Hospital of Philadelphia

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Chief Editor

Herbert H Engelhard, III, MD, PhD, FACS, FAANS, Professor of Clinical Neurosurgery and Bioengineering, Chief, Division of Neuro-Oncology, Medical Director, UIC Neurosurgery Clinic, Department of Neurosurgery, University of Illinois at Chicago College of Medicine

Disclosure: Nothing to disclose.

Additional Contributors

Robert C Shepard, MD, FACP, Associate Professor of Medicine in Hematology and Oncology at University of North Carolina at Chapel Hill; Vice President of Scientific Affairs, Therapeutic Expertise, Oncology, at PRA International

Disclosure: Nothing to disclose.

Acknowledgements

We would like to acknowledge previous contributions to this chapter from Katharine Cronk, MD,PhD; Richard C Anderson, MD; Chris E Mandigo, MD; Andrew T Parsa MD, PhD; Patrick B Senatus, MD, PhD; and Allen Waziri, MD.

References

  1. Farrell CJ, Plotkin SR. Genetic causes of brain tumors: neurofibromatosis, tuberous sclerosis, von Hippel-Lindau, and other syndromes. Neurol Clin. 2007 Nov. 25(4):925-46, viii. [View Abstract]
  2. Hardell L, Carlberg M. Mobile phone and cordless phone use and the risk for glioma - Analysis of pooled case-control studies in Sweden, 1997-2003 and 2007-2009. Pathophysiology. 2015 Mar. 22 (1):1-13. [View Abstract]
  3. Lahkola A, Auvinen A, Raitanen J, et al. Mobile phone use and risk of glioma in 5 North European countries. Int J Cancer. 2007 Apr 15. 120(8):1769-75. [View Abstract]
  4. Inskip PD, Tarone RE, Hatch EE, et al. Cellular-telephone use and brain tumors. N Engl J Med. 2001 Jan 11. 344(2):79-86. [View Abstract]
  5. Weintraub MI. Glioblastoma multiforme and the cellular telephone scare. J Neurosurg. 1994 Jan. 80(1):169-70. [View Abstract]
  6. Kan P, Simonsen SE, Lyon JL, Kestle JR. Cellular phone use and brain tumor: a meta-analysis. J Neurooncol. 2008 Jan. 86(1):71-8. [View Abstract]
  7. Mukundan S, Holder C, Olson JJ. Neuroradiological assessment of newly diagnosed glioblastoma. J Neurooncol. 2008 Sep. 89(3):259-69. [View Abstract]
  8. Piroth MD, Holy R, Pinkawa M, et al. Prognostic impact of postoperative, pre-irradiation (18)F-fluoroethyl-l-tyrosine uptake in glioblastoma patients treated with radiochemotherapy. Radiother Oncol. 2011 May. 99(2):218-24. [View Abstract]
  9. Russell DS, Rubinstein LJ. Pathology of tumors of the nervous system. 6th ed. London, England: Edward Arnold; 1998. 426-52.
  10. Daumas-Duport C, Scheithauer B, O'Fallon J, Kelly P. Grading of astrocytomas. A simple and reproducible method. Cancer. 1988 Nov 15. 62(10):2152-65. [View Abstract]
  11. Kim TS, Halliday AL, Hedley-Whyte ET, Convery K. Correlates of survival and the Daumas-Duport grading system for astrocytomas. J Neurosurg. 1991 Jan. 74(1):27-37. [View Abstract]
  12. Pedersen PH, Rucklidge GJ, Mork SJ, et al. Leptomeningeal tissue: a barrier against brain tumor cell invasion. J Natl Cancer Inst. 1994 Nov 2. 86(21):1593-9. [View Abstract]
  13. Nagashima G, Suzuki R, Hokaku H, et al. Graphic analysis of microscopic tumor cell infiltration, proliferative potential, and vascular endothelial growth factor expression in an autopsy brain with glioblastoma. Surg Neurol. 1999 Mar. 51(3):292-9. [View Abstract]
  14. Pompili A, Calvosa F, Caroli F, et al. The transdural extension of gliomas. J Neurooncol. 1993 Jan. 15(1):67-74. [View Abstract]
  15. Brat DJ, Prayson RA, Ryken TC, Olson JJ. Diagnosis of malignant glioma: role of neuropathology. J Neurooncol. 2008 Sep. 89(3):287-311. [View Abstract]
  16. Lampl Y, Eshel Y, Gilad R, Sarova-Pinchas I. Glioblastoma multiforme with bone metastase and cauda equina syndrome. J Neurooncol. 1990 Apr. 8(2):167-72. [View Abstract]
  17. Hulbanni S, Goodman PA. Glioblastoma multiforme with extraneural metastases in the absence of previous surgery. Cancer. 1976 Mar. 37(3):1577-83. [View Abstract]
  18. Hoffman HJ, Duffner PK. Extraneural metastases of central nervous system tumors. Cancer. 1985 Oct 1. 56(7 Suppl):1778-82. [View Abstract]
  19. Barnard RO, Geddes JF. The incidence of multifocal cerebral gliomas. A histologic study of large hemisphere sections. Cancer. 1987 Oct 1. 60(7):1519-31. [View Abstract]
  20. Batzdorf U, Malamud N. The Problem of Multicentric Gliomas. J Neurosurg. 1963 Feb. 20:122-36. [View Abstract]
  21. Pasquier B, Pasquier D, N'Golet A, Panh MH, Couderc P. Extraneural metastases of astrocytomas and glioblastomas: clinicopathological study of two cases and review of literature. Cancer. 1980 Jan 1. 45(1):112-25. [View Abstract]
  22. Preusser M, de Ribaupierre S, Wohrer A, et al. Current concepts and management of glioblastoma. Ann Neurol. 2011 Jul. 70(1):9-21. [View Abstract]
  23. Sathornsumetee S, Reardon DA, Desjardins A, Quinn JA, Vredenburgh JJ, Rich JN. Molecularly targeted therapy for malignant glioma. Cancer. 2007 Jul 1. 110(1):13-24. [View Abstract]
  24. Furnari FB, Fenton T, Bachoo RM, et al. Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 2007 Nov 1. 21(21):2683-710. [View Abstract]
  25. Keime-Guibert F, Chinot O, Taillandier L, et al. Radiotherapy for glioblastoma in the elderly. N Engl J Med. 2007 Apr 12. 356(15):1527-35. [View Abstract]
  26. Roa W, Brasher PM, Bauman G, et al. Abbreviated course of radiation therapy in older patients with glioblastoma multiforme: a prospective randomized clinical trial. J Clin Oncol. 2004 May 1. 22(9):1583-8. [View Abstract]
  27. Glantz M, Chamberlain M, Liu Q, Litofsky NS, Recht LD. Temozolomide as an alternative to irradiation for elderly patients with newly diagnosed malignant gliomas. Cancer. 2003 May 1. 97(9):2262-6. [View Abstract]
  28. Scott J, Tsai YY, Chinnaiyan P, Yu HH. Effectiveness of radiotherapy for elderly patients with glioblastoma. Int J Radiat Oncol Biol Phys. 2011 Sep 1. 81(1):206-10. [View Abstract]
  29. Malmstrom A, Gronberg BH, Marosi C, et al; Nordic Clinical Brain Tumour Study Group (NCBTSG). Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. Lancet Oncol. 2012 Sep. 13(9):916-26. [View Abstract]
  30. Waugh MG. Chromosomal Instability and Phosphoinositide Pathway Gene Signatures in Glioblastoma Multiforme. Mol Neurobiol. 2014 Dec 15. [View Abstract]
  31. von Deimling A, Louis DN, von Ammon K, et al. Association of epidermal growth factor receptor gene amplification with loss of chromosome 10 in human glioblastoma multiforme. J Neurosurg. 1992 Aug. 77(2):295-301. [View Abstract]
  32. Ohgaki H, Kleihues P. Genetic pathways to primary and secondary glioblastoma. Am J Pathol. 2007 May. 170(5):1445-53. [View Abstract]
  33. Wong AJ, Ruppert JM, Bigner SH, et al. Structural alterations of the epidermal growth factor receptor gene in human gliomas. Proc Natl Acad Sci U S A. 1992 Apr 1. 89(7):2965-9. [View Abstract]
  34. Desai R, Suryadevara CM, Batich KA, Farber SH, Sanchez-Perez L, Sampson JH. Emerging immunotherapies for glioblastoma. Expert Opin Emerg Drugs. 2016 Jun. 21 (2):133-45. [View Abstract]
  35. Shapiro WR, Green SB, Burger PC, et al. Randomized trial of three chemotherapy regimens and two radiotherapy regimens and two radiotherapy regimens in postoperative treatment of malignant glioma. Brain Tumor Cooperative Group Trial 8001. J Neurosurg. 1989 Jul. 71(1):1-9. [View Abstract]
  36. Duerr EM, Rollbrocker B, Hayashi Y, et al. PTEN mutations in gliomas and glioneuronal tumors. Oncogene. 1998 Apr 30. 16(17):2259-64. [View Abstract]
  37. Warren KE, Gururangan S, Geyer JR, McLendon RE, Poussaint TY, Wallace D, et al. A phase II study of O6-benzylguanine and temozolomide in pediatric patients with recurrent or progressive high-grade gliomas and brainstem gliomas: a Pediatric Brain Tumor Consortium study. J Neurooncol. 2012 Feb. 106 (3):643-9. [View Abstract]
  38. Kleihues P, Burger PC, Cavenee WK. Glioblastoma. WHO Classification: Pathology and genetics of tumors of the nervous system. 1st ed. Lyon, France: International Agency for Research on Cancers; 1997. 16-24.
  39. Fisher JL, Schwartzbaum JA, Wrensch M, Wiemels JL. Epidemiology of brain tumors. Neurol Clin. 2007 Nov. 25(4):867-90, vii. [View Abstract]
  40. Chi AS, Wen PY. Inhibiting kinases in malignant gliomas. Expert Opin Ther Targets. 2007 Apr. 11(4):473-96. [View Abstract]
  41. Duda DG, Jain RK, Willett CG. Antiangiogenics: the potential role of integrating this novel treatment modality with chemoradiation for solid cancers. J Clin Oncol. 2007 Sep 10. 25(26):4033-42. [View Abstract]
  42. Scott JN, Rewcastle NB, Brasher PM, et al. Long-term glioblastoma multiforme survivors: a population-based study. Can J Neurol Sci. 1998 Aug. 25(3):197-201. [View Abstract]
  43. Sneed PK, Prados MD, McDermott MW, et al. Large effect of age on the survival of patients with glioblastoma treated with radiotherapy and brachytherapy boost. Neurosurgery. 1995 May. 36(5):898-903; discussion 903-4. [View Abstract]
  44. Sanai N, Berger MS. Glioma extent of resection and its impact on patient outcome. Neurosurgery. 2008 Apr. 62(4):753-64; discussion 264-6. [View Abstract]
  45. Keles GE, Anderson B, Berger MS. The effect of extent of resection on time to tumor progression and survival in patients with glioblastoma multiforme of the cerebral hemisphere. Surg Neurol. 1999 Oct. 52(4):371-9. [View Abstract]
  46. Lacroix M, Abi-Said D, Fourney DR, et al. A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. J Neurosurg. 2001 Aug. 95(2):190-8. [View Abstract]
  47. Salmon I, Dewitte O, Pasteels JL, et al. Prognostic scoring in adult astrocytic tumors using patient age, histopathological grade, and DNA histogram type. J Neurosurg. 1994 May. 80(5):877-83. [View Abstract]
  48. Perrini P, Gambacciani C, Weiss A, Pasqualetti F, Delishaj D, Paiar F, et al. Survival outcomes following repeat surgery for recurrent glioblastoma: a single-center retrospective analysis. J Neurooncol. 2016 Nov 14. [View Abstract]
  49. Newcomb EW, Cohen H, Lee SR, et al. Survival of patients with glioblastoma multiforme is not influenced by altered expression of p16, p53, EGFR, MDM2 or Bcl-2 genes. Brain Pathol. 1998 Oct. 8(4):655-67. [View Abstract]
  50. Babu R, Komisarow JM, Agarwal VJ, Rahimpour S, Iyer A, Britt D, et al. Glioblastoma in the elderly: the effect of aggressive and modern therapies on survival. J Neurosurg. 2016 Apr. 124 (4):998-1007. [View Abstract]
  51. Jordan JT, Gerstner ER, Batchelor TT, Cahill DP, Plotkin SR. Glioblastoma care in the elderly. Cancer. 2016 Jan 15. 122 (2):189-97. [View Abstract]
  52. Li J, Wang M, Won M, et al. Validation and simplification of the Radiation Therapy Oncology Group recursive partitioning analysis classification for glioblastoma. Int J Radiat Oncol Biol Phys. 2011 Nov 1. 81(3):623-30. [View Abstract]
  53. Kaur G, Bloch O, Jian BJ, et al. A critical evaluation of cystic features in primary glioblastoma as a prognostic factor for survival. J Neurosurg. 2011 Oct. 115(4):754-9. [View Abstract]
  54. Caccamo DV, Rubenstein LJ. Tumors: Applications of immunohistochemical methods. Neuropathology: The diagnostic approach. St Louis, Mo: Mosby-Year Book; 1997. 193-218.
  55. Herrlinger U, Tzaridis T, Mack F, et al, Neurooncology Working Group of the German Cancer Society. Lomustine-temozolomide combination therapy versus standard temozolomide therapy in patients with newly diagnosed glioblastoma with methylated MGMT promoter (CeTeG/NOA-09): a randomised, open-label, phase 3 trial. Lancet. 2019 Feb 16. 393 (10172):678-688. [View Abstract]
  56. Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009 May. 10(5):459-66. [View Abstract]
  57. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005 Mar 10. 352(10):987-96. [View Abstract]
  58. Chamberlain MC, Kormanik PA. Practical guidelines for the treatment of malignant gliomas. West J Med. 1998 Feb. 168(2):114-20. [View Abstract]
  59. Batich KA, Reap EA, Archer GE, Sanchez-Perez L, Nair SK, Schmittling RJ, et al. Long-term Survival in Glioblastoma with Cytomegalovirus pp65-Targeted Vaccination. Clin Cancer Res. 2017 Apr 15. 23 (8):1898-1909. [View Abstract]
  60. Wernicke AG, Edgar MA, Lavi E, et al. Prostate-specific membrane antigen as a potential novel vascular target for treatment of glioblastoma multiforme. Arch Pathol Lab Med. 2011 Nov. 135(11):1486-9. [View Abstract]
  61. [Guideline] Olson JJ, Nayak L, Ormond DR, Wen PY, Kalkanis SN, AANS/CNS Joint Guidelines Committee. The role of cytotoxic chemotherapy in the management of progressive glioblastoma : a systematic review and evidence-based clinical practice guideline. J Neurooncol. 2014 Jul. 118 (3):501-55. [View Abstract]
  62. Barker FG, Prados MD, Chang SM, et al. Radiation response and survival time in patients with glioblastoma multiforme. J Neurosurg. 1996 Mar. 84(3):442-8. [View Abstract]
  63. Leibel SA, Scott CB, Loeffler JS. Contemporary approaches to the treatment of malignant gliomas with radiation therapy. Semin Oncol. 1994 Apr. 21(2):198-219. [View Abstract]
  64. Liang BC, Thornton AF Jr, Sandler HM, Greenberg HS. Malignant astrocytomas: focal tumor recurrence after focal external beam radiation therapy. J Neurosurg. 1991 Oct. 75(4):559-63. [View Abstract]
  65. Buatti J, Ryken TC, Smith MC, et al. Radiation therapy of pathologically confirmed newly diagnosed glioblastoma in adults. J Neurooncol. 2008 Sep. 89(3):313-37. [View Abstract]
  66. Huang J, Samson P, Perkins SM, Ansstas G, Chheda MG, DeWees TA, et al. Impact of concurrent chemotherapy with radiation therapy for elderly patients with newly diagnosed glioblastoma: a review of the National Cancer Data Base. J Neurooncol. 2016 Nov 14. 49(3):333-43. [View Abstract]
  67. Jablonska PA, Diez-Valle R, Pérez-Larraya JG, Moreno-Jiménez M, Idoate MÁ, Arbea L, et al. Hypofractionated radiation therapy and temozolomide in patients with glioblastoma and poor prognostic factors. A prospective, single-institution experience. PLoS One. 2019. 14 (6):e0217881. [View Abstract]
  68. Gzell C, Back M, Wheeler H, Bailey D, Foote M. Radiotherapy in Glioblastoma: the Past, the Present and the Future. Clin Oncol (R Coll Radiol). 2016 Oct 13. Vol 3:867-82. [View Abstract]
  69. Hochberg FH, Pruitt A. Assumptions in the radiotherapy of glioblastoma. Neurology. 1980 Sep. 30(9):907-11. [View Abstract]
  70. Waters JD, Rose B, Gonda DD, et al. Immediate post-operative brachytherapy prior to irradiation and temozolomide for newly diagnosed glioblastoma. J Neurooncol. 2013 Jul. 113(3):467-77. [View Abstract]
  71. Stupp R, Hegi ME, Gilbert MR, Chakravarti A. Chemoradiotherapy in malignant glioma: standard of care and future directions. J Clin Oncol. 2007 Sep 10. 25(26):4127-36. [View Abstract]
  72. Rodrigus P. Motexafin gadolinium: a possible new radiosensitiser. Expert Opin Investig Drugs. 2003 Jul. 12(7):1205-10. [View Abstract]
  73. Butowski NA, Sneed PK, Chang SM. Diagnosis and treatment of recurrent high-grade astrocytoma. J Clin Oncol. 2006 Mar 10. 24(8):1273-80. [View Abstract]
  74. Combs SE, Thilmann C, Edler L, Debus J, Schulz-Ertner D. Efficacy of fractionated stereotactic reirradiation in recurrent gliomas: long-term results in 172 patients treated in a single institution. J Clin Oncol. 2005 Dec 1. 23(34):8863-9. [View Abstract]
  75. Tsao MN, Mehta MP, Whelan TJ, et al. The American Society for Therapeutic Radiology and Oncology (ASTRO) evidence-based review of the role of radiosurgery for malignant glioma. Int J Radiat Oncol Biol Phys. 2005 Sep 1. 63(1):47-55. [View Abstract]
  76. [Guideline] Ryu S, Buatti JM, Morris A, Kalkanis SN, Ryken TC, Olson JJ, et al. The role of radiotherapy in the management of progressive glioblastoma : a systematic review and evidence-based clinical practice guideline. J Neurooncol. 2014 Jul. 118 (3):489-99. [View Abstract]
  77. Fleischmann DF, Jenn J, Corradini S, Ruf V, Herms J, Forbrig R, et al. Bevacizumab reduces toxicity of reirradiation in recurrent high-grade glioma. Radiother Oncol. 2019 Jun 25. 138:99-105. [View Abstract]
  78. Kornblith PL. The role of cytotoxic chemotherapy in the treatment of malignant brain tumors. Surg Neurol. 1995 Dec. 44(6):551-2. [View Abstract]
  79. Kornblith PL, Walker M. Chemotherapy for malignant gliomas [published erratum appears in J Neurosurg 1988 Oct;69(4):645]. J Neurosurg. 1988 Jan. 68(1):1-17. [View Abstract]
  80. Lesser GJ, Grossman S. The chemotherapy of high-grade astrocytomas. Semin Oncol. 1994 Apr. 21(2):220-35. [View Abstract]
  81. Levin VA. Chemotherapy of primary brain tumors. Frank BD, ed. Symposium on Neuro-Oncology. 4th ed. New York, NY: Neurologic Clinics; 1985. Vol 3: 855-66.
  82. Levin VA, Silver P, Hannigan J, et al. Superiority of post-radiotherapy adjuvant chemotherapy with CCNU, procarbazine, and vincristine (PCV) over BCNU for anaplastic gliomas: NCOG 6G61 final report. Int J Radiat Oncol Biol Phys. 1990 Feb. 18(2):321-4. [View Abstract]
  83. Fadul CE, Wen PY, Kim L, Olson JJ. Cytotoxic chemotherapeutic management of newly diagnosed glioblastoma multiforme. J Neurooncol. 2008 Sep. 89(3):339-57. [View Abstract]
  84. Fine HA, Dear KB, Loeffler JS, Black PM, Canellos GP. Meta-analysis of radiation therapy with and without adjuvant chemotherapy for malignant gliomas in adults. Cancer. 1993 Apr 15. 71(8):2585-97. [View Abstract]
  85. Stewart LA. Chemotherapy in adult high-grade glioma: a systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet. 2002 Mar 23. 359(9311):1011-8. [View Abstract]
  86. Westphal M, Ram Z, Riddle V, Hilt D, Bortey E. Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial. Acta Neurochir (Wien). 2006 Mar. 148(3):269-75; discussion 275. [View Abstract]
  87. Gutenberg A, Bock HC, Bruck W, et al. MGMT promoter methylation status and prognosis of patients with primary or recurrent glioblastoma treated with carmustine wafers. Br J Neurosurg. 2013 Dec. 27(6):772-8. [View Abstract]
  88. Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005 Mar 10. 352(10):997-1003. [View Abstract]
  89. Hegi ME, Liu L, Herman JG, et al. Correlation of O6-methylguanine methyltransferase (MGMT) promoter methylation with clinical outcomes in glioblastoma and clinical strategies to modulate MGMT activity. J Clin Oncol. 2008 Sep 1. 26(25):4189-99. [View Abstract]
  90. Broniscer A, Gururangan S, MacDonald TJ, et al. Phase I trial of single-dose temozolomide and continuous administration of o6-benzylguanine in children with brain tumors: a pediatric brain tumor consortium report. Clin Cancer Res. 2007 Nov 15. 13(22 Pt 1):6712-8. [View Abstract]
  91. Kaiser MG, Parsa AT, Fine RL, Hall JS, Chakrabarti I, Bruce JN. Tissue distribution and antitumor activity of topotecan delivered by intracerebral clysis in a rat glioma model. Neurosurgery. 2000 Dec. 47(6):1391-8; discussion 1398-9. [View Abstract]
  92. Bruce JN, Falavigna A, Johnson JP, et al. Intracerebral clysis in a rat glioma model. Neurosurgery. 2000 Mar. 46(3):683-91. [View Abstract]
  93. Lopez KA, Waziri AE, Canoll PD, Bruce JN. Convection-enhanced delivery in the treatment of malignant glioma. Neurol Res. 2006 Jul. 28(5):542-8. [View Abstract]
  94. Brem H, Piantadosi S, Burger PC, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-brain Tumor Treatment Group. Lancet. 1995 Apr 22. 345(8956):1008-12. [View Abstract]
  95. Bota DA, Desjardins A, Quinn JA, Affronti ML, Friedman HS. Interstitial chemotherapy with biodegradable BCNU (Gliadel) wafers in the treatment of malignant gliomas. Ther Clin Risk Manag. 2007 Oct. 3(5):707-15. [View Abstract]
  96. FDA. Avastin Approval History. U.S. Food and Drug Administration. Available at http://www.accessdata.fda.gov/drugsatfda_docs/label/2009/125085s0169lbl.pdf. Accessed: June 15, 2019.
  97. Vredenburgh JJ, Desjardins A, Herndon JE 2nd, et al. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007 Feb 15. 13(4):1253-9. [View Abstract]
  98. Vredenburgh JJ, Desjardins A, Herndon JE 2nd, et al. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007 Oct 20. 25(30):4722-9. [View Abstract]
  99. Cloughesy TF, Prados MD, Wen PY. A phase II, randomized, non-comparative clinical trial of the effect of bevacizumab (BV) alone or in combinationwith irinotecan (CPT) on 6-month progressionfree survival (PFS6) in recurrent, treatment-refractory glioblastoma (GBM). J Clin Oncol. 2008. 26:Suppl:91s.
  100. Rich JN, Rasheed BK, Yan H. EGFR mutations and sensitivity to gefitinib. N Engl J Med. 2004 Sep 16. 351(12):1260-1; author reply 1260-1. [View Abstract]
  101. Rich JN, Reardon DA, Peery T, Dowell JM, Quinn JA, Penne KL. Phase II trial of gefitinib in recurrent glioblastoma. J Clin Oncol. 2004 Jan 1. 22(1):133-42. [View Abstract]
  102. Mellinghoff IK, Wang MY, Vivanco I, Haas-Kogan DA, Zhu S, Dia EQ. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med. 2005 Nov 10. 353(19):2012-24. [View Abstract]
  103. Fulci G, Chiocca EA. The status of gene therapy for brain tumors. Expert Opin Biol Ther. 2007 Feb. 7(2):197-208. [View Abstract]
  104. Reardon DA, Akabani G, Coleman RE, et al. Salvage radioimmunotherapy with murine iodine-131-labeled antitenascin monoclonal antibody 81C6 for patients with recurrent primary and metastatic malignant brain tumors: phase II study results. J Clin Oncol. 2006 Jan 1. 24(1):115-22. [View Abstract]
  105. Mamelak AN, Rosenfeld S, Bucholz R, et al. Phase I single-dose study of intracavitary-administered iodine-131-TM-601 in adults with recurrent high-grade glioma. J Clin Oncol. 2006 Aug 1. 24(22):3644-50. [View Abstract]
  106. Ferguson S, Lesniak MS. Convection enhanced drug delivery of novel therapeutic agents to malignant brain tumors. Curr Drug Deliv. 2007 Apr. 4(2):169-80. [View Abstract]
  107. Quang TS, Brady LW. Radioimmunotherapy as a novel treatment regimen: (125)I-labeled monoclonal antibody 425 in the treatment of high-grade brain gliomas. Int J Radiat Oncol Biol Phys. 2004 Mar 1. 58(3):972-5. [View Abstract]
  108. Rich JN, Bigner DD. Development of novel targeted therapies in the treatment of malignant glioma. Nat Rev Drug Discov. 2004 May. 3(5):430-46. [View Abstract]
  109. Nelson R. Bevacizumab May Boost Survival in Glioblastoma. Medscape Medical News. Available at http://www.medscape.com/viewarticle/809962. August 23, 2013; Accessed: June 15, 2019.
  110. Johnson DR, Leeper HE, Uhm JH. Glioblastoma survival in the United States improved after Food and Drug Administration approval of bevacizumab: a population-based analysis. Cancer. 2013 Oct 1. 119(19):3489-95. [View Abstract]
  111. Nelson R. FDA Expands Indication for Optune Device in Glioblastoma. Medscape Medical News. Available at http://www.medscape.com/viewarticle/852196. October 6, 2015; Accessed: June 15, 2019.
  112. Stupp R, Taillibert S, Kanner A, Read W, Steinberg DM, et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA. 2017 Dec 19. 318 (23):2306-2316. [View Abstract]
  113. Castellino AM. Tumor-Treating Fields: Significant Survival in Glioblastoma. Medscape Medical News. Available at https://www.medscape.com/viewarticle/890317. December 19, 2017; Accessed: June 15, 2019.
  114. Desjardins A, Gromeier M, Herndon JE 2nd, Beaubier N, Bolognesi DP, Friedman AH, et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N Engl J Med. 2018 Jun 26. [View Abstract]
  115. Gleolan (aminolevulinic acid oral) [package insert]. Lexington, KY: NX Development Corp. 2017 June. Available at
  116. Patil CG, Walker DG, Miller DM, Butte P, Morrison B, Kittle DS, et al. Phase 1 Safety, Pharmacokinetics, and Fluorescence Imaging Study of Tozuleristide (BLZ-100) in Adults With Newly Diagnosed or Recurrent Gliomas. Neurosurgery. 2019 May 9. [View Abstract]
  117. Vlessides M. Imaging Agent from Scorpion Venom May Improve Brain Tumor Resection. Medscape Medical News. May 30, 2019. Available at https://www.medscape.com/viewarticle/913701
  118. Ammirati M, Vick N, Liao YL, et al. Effect of the extent of surgical resection on survival and quality of life in patients with supratentorial glioblastomas and anaplastic astrocytomas. Neurosurgery. 1987 Aug. 21(2):201-6. [View Abstract]
  119. Fadul C, Wood J, Thaler H, et al. Morbidity and mortality of craniotomy for excision of supratentorial gliomas. Neurology. 1988 Sep. 38(9):1374-9. [View Abstract]
  120. Li YM, Suki D, Hess K, Sawaya R. The influence of maximum safe resection of glioblastoma on survival in 1229 patients: Can we do better than gross-total resection?. J Neurosurg. 2016 Apr. 124 (4):977-88. [View Abstract]
  121. Ryken TC, Frankel B, Julien T, Olson JJ. Surgical management of newly diagnosed glioblastoma in adults: role of cytoreductive surgery. J Neurooncol. 2008 Sep. 89(3):271-86. [View Abstract]
  122. Ciric I, Rovin R, Cozzens JW. Role of surgery in the treatment of malignant cerebral gliomas. Malignant Cerebral Glioma. Park Ridge, Ill: American Association of Neurological Surgeons; 1990. 141-53.
  123. El Hindy N, Bachmann HS, Lambertz et al. Association of the CC genotype of the regulatory BCL2 promoter polymorphism (-938C>A) with better 2-year survival in patients with glioblastoma multiforme. J Neurosurg. 2011 Jun. 114(6):1631-9. [View Abstract]
  124. Coffey RJ, Lunsford LD, Taylor FH. Survival after stereotactic biopsy of malignant gliomas. Neurosurgery. 1988 Mar. 22(3):465-73. [View Abstract]
  125. Jakola AS, Unsgard G, Solheim O. Quality of life in patients with intracranial gliomas: the impact of modern image-guided surgery. J Neurosurg. 2011 Jun. 114(6):1622-30. [View Abstract]
  126. [Guideline] NCCN Clinical Practice Guidelines in Oncology: Central Nervous System Cancers--V. 1.2019. National Comprehensive Cancer Network. Available at https://www.nccn.org/professionals/physician_gls/pdf/cns.pdf. March 5, 2019; Accessed: June 15, 2019.
  127. Glantz MJ, Cole BF, Forsyth PA, et al. Practice parameter: anticonvulsant prophylaxis in patients with newly diagnosed brain tumors. Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2000 May 23. 54(10):1886-93. [View Abstract]
  128. Chaichana KL, Jusue-Torres I, Navarro-Ramirez R, et al. Establishing percent resection and residual volume thresholds affecting survival and recurrence for patients with newly diagnosed intracranial glioblastoma. Neuro Oncol. 2014 Jan. 16(1):113-22. [View Abstract]

Histopathologic slide demonstrating a glioblastoma multiforme (GBM).

A T1-weighted axial MRI without intravenous contrast. This image demonstrates a hemorrhagic multicentric tumor (glioblastoma multiforme [GBM]) in the right temporal lobe. Effacement of the ventricular system is present on the right, and mild impingement of the right medial temporal lobe can be observed on the midbrain.

A T1-weighted sagittal MRI with intravenous contrast in a patient with glioblastoma multiforme (GBM).

A T1-weighted axial MRI without intravenous contrast. This image demonstrates a hemorrhagic multicentric tumor (glioblastoma multiforme [GBM]) in the right temporal lobe. Effacement of the ventricular system is present on the right, and mild impingement of the right medial temporal lobe can be observed on the midbrain.

A T1-weighted axial MRI with intravenous contrast. Heterogenous enhancement of the lesion is present within the right temporal lobe. The hypointensity circumscribed within the enhancement is suggestive of necrosis. This radiologic appearance is typical of a multicentric glioblastoma multiforme (GBM).

A T1-weighted coronal MRI with intravenous contrast. This image demonstrates the lesion (glioblastoma multiforme [GBM]) within the medial temporal lobe and the stereotypical pattern of contrast enhancement.

A T1-weighted sagittal MRI with intravenous contrast in a patient with glioblastoma multiforme (GBM).

A T2-weighted axial MRI. The tumor (glioblastoma multiforme [GBM]) and surrounding white matter within the right temporal lobe show increased signal intensity compared to a healthy brain, suggesting extensive tumorigenic edema.

A fluid-attenuated inversion recovery (FLAIR) axial MRI. This image is similar to the T2-weighted image and demonstrates extensive edema in a patient with glioblastoma multiforme (GBM).

Magnetic resonance (MR) spectroscopy is representative of a glioblastoma multiforme (GBM), demonstrating a high peak ratio of choline (CHO) to creatine (CR), a decreased N-acetylaspartate (NAA) peak, and an increased lactate (LAC) peak.

Histopathologic slide demonstrating a glioblastoma multiforme (GBM).

Axial CT scan without intravenous contrast. This image reveals a large right temporal intraaxial mass (glioblastoma multiforme [GBM]). Extensive surrounding edema is present, as demonstrated by the peritumoral hypodensity, and a moderate right-to-left midline shift can be noted. All of the radiologic studies in this article are of the same patient.

A T1-weighted axial MRI without intravenous contrast. This image demonstrates a hemorrhagic multicentric tumor (glioblastoma multiforme [GBM]) in the right temporal lobe. Effacement of the ventricular system is present on the right, and mild impingement of the right medial temporal lobe can be observed on the midbrain.

A T1-weighted axial MRI with intravenous contrast. Heterogenous enhancement of the lesion is present within the right temporal lobe. The hypointensity circumscribed within the enhancement is suggestive of necrosis. This radiologic appearance is typical of a multicentric glioblastoma multiforme (GBM).

A T1-weighted coronal MRI with intravenous contrast. This image demonstrates the lesion (glioblastoma multiforme [GBM]) within the medial temporal lobe and the stereotypical pattern of contrast enhancement.

A T1-weighted sagittal MRI with intravenous contrast in a patient with glioblastoma multiforme (GBM).

A T2-weighted axial MRI. The tumor (glioblastoma multiforme [GBM]) and surrounding white matter within the right temporal lobe show increased signal intensity compared to a healthy brain, suggesting extensive tumorigenic edema.

A fluid-attenuated inversion recovery (FLAIR) axial MRI. This image is similar to the T2-weighted image and demonstrates extensive edema in a patient with glioblastoma multiforme (GBM).

Histopathologic slide demonstrating a glioblastoma multiforme (GBM).

Axial CT scan without intravenous contrast. This image reveals a large right temporal intraaxial mass (glioblastoma multiforme [GBM]). Extensive surrounding edema is present, as demonstrated by the peritumoral hypodensity, and a moderate right-to-left midline shift can be noted. All of the radiologic studies in this article are of the same patient.

A T1-weighted axial MRI without intravenous contrast. This image demonstrates a hemorrhagic multicentric tumor (glioblastoma multiforme [GBM]) in the right temporal lobe. Effacement of the ventricular system is present on the right, and mild impingement of the right medial temporal lobe can be observed on the midbrain.

A T1-weighted axial MRI with intravenous contrast. Heterogenous enhancement of the lesion is present within the right temporal lobe. The hypointensity circumscribed within the enhancement is suggestive of necrosis. This radiologic appearance is typical of a multicentric glioblastoma multiforme (GBM).

A T1-weighted coronal MRI with intravenous contrast. This image demonstrates the lesion (glioblastoma multiforme [GBM]) within the medial temporal lobe and the stereotypical pattern of contrast enhancement.

A T1-weighted sagittal MRI with intravenous contrast in a patient with glioblastoma multiforme (GBM).

A T2-weighted axial MRI. The tumor (glioblastoma multiforme [GBM]) and surrounding white matter within the right temporal lobe show increased signal intensity compared to a healthy brain, suggesting extensive tumorigenic edema.

A fluid-attenuated inversion recovery (FLAIR) axial MRI. This image is similar to the T2-weighted image and demonstrates extensive edema in a patient with glioblastoma multiforme (GBM).

Histopathologic slide demonstrating a glioblastoma multiforme (GBM).

Magnetic resonance (MR) spectroscopy is representative of a glioblastoma multiforme (GBM), demonstrating a high peak ratio of choline (CHO) to creatine (CR), a decreased N-acetylaspartate (NAA) peak, and an increased lactate (LAC) peak.