This edition of CCR Focus provides critical reviews of several important areas in the field, including the application of findings from genomic investigations of brain tumors to improve diagnosis, clinical trial design, and ultimately optimizing individual patient treatment. Another article is a critical review provided by experts in the field that discusses the recent clinical trials using angiogenesis inhibitors, possible explanations for the results, and how to move forward. There is a concise discussion of the application of immunotherapy to brain tumors by key investigators in this field, reflecting the potential opportunities as well as the disease-specific challenges. Finally, leading pediatric brain tumor investigators provide an overview of the field and insights about the recent seminal discoveries in two pediatric brain tumors, supporting the paradigm that laboratory investigations lead to more precise diagnosis, prognosis, and ultimately better treatment. Herein, an overview of the recent advances and challenges in the area of clinical and translational brain tumor research is provided to set the stage for the contributions that follow.

See all articles in this CCR Focus section, “Discoveries, Challenges, and Progress in Primary Brain Tumors.”

Clin Cancer Res; 20(22); 5591–600. ©2014 AACR.

Malignant primary brain tumors remain an important area of clinical research. Most of these cancers remain incurable despite decades of laboratory and clinical investigation. These tumors are somewhat unique in that they rarely spread outside the central nervous system (CNS). Despite this finding, surgery is not curative even in low-grade (WHO grade 2) tumors because of their infiltrative nature (1). However, the extent of tumor resection is important as it has prognostic significance in many CNS cancers, and in some tumors, such as medulloblastoma and low-grade glioma, dictates subsequent therapies (2). Following surgery, radiotherapy remains the cornerstone of most brain tumor treatment regimens (3). Enhancing tumor targeting with new radiation modalities such as intensity-modulated radiotherapy or alternative radiation sources such as heavy particle therapy (i.e., proton, carbon ion) may improve the risk to benefit ratio in brain tumors (4).

The progress in developing effective chemotherapy regimens for primary brain tumors lags behind that of other cancers. For the most common and most aggressive brain tumor, glioblastoma (WHO grade 4), the only systemic chemotherapy that has proven survival benefit is temozolomide, an oral alkylating agent. Even with the first-line use of temozolomide, the survival benefit is only measured in months (5). Over the past several decades, many novel treatment agents have been tested; some of these studies are listed in Table 1. These included established and novel cytotoxic chemotherapy agents, antiangiogenic agents, signal transduction modulators, biologic agents, and immunotherapies (6). Early studies recognized that drug delivery across the blood–brain barrier may limit the efficacy of many therapeutic agents; novel routes of delivery have been tried. These include direct tumor injection, intra-arterial delivery, tumor perfusion using convection-enhanced delivery, and implantation of slow release vehicles (i.e., polifeprosan 20 with carmustine wafer; refs. 7, 8). Despite all of these efforts, the portfolio of effective agents remains quite limited.

Table 1.

Single-agent targeted therapies for recurrent glioblastoma

AgentTargetTrial phaseNumber of patients6-month PFS (%)
Imatinib (50) PDGFR, c-kit, and c-ABL II 51 16 
Gefitinib (51, 52) EGFR II 53 and 28 13 and 14 
Erlotinib (53, 54) EGFR II 110 and 38 11 and 3 
Pazopanib (55) VEGFR and PDGFR II 35 
Tipifarnib (56) Farnesyltransferase II 67 12 
Temsirolimus (57) mTOR II 65 and 41 8 and 3 
Cediranib (58, 59) VEGFR II and III 31 and 131 26 and 16 
Bevacizumab (60, 61) VEGF-A II 85 and 48 36 and 29 
Aflibercept (62) VEGF-A II 42 
Cilengitide (63, 64) αvβ3,αvβ5 integrins II 81 and 26 15 and 12 
Vorinostat (65) HDAC II 66 15 
Enzastaurin (66, 67) PKC II and III 72 and 174 7 and 11 
Cabozantinib (68) EGFR and C-MET II 124 21 
AgentTargetTrial phaseNumber of patients6-month PFS (%)
Imatinib (50) PDGFR, c-kit, and c-ABL II 51 16 
Gefitinib (51, 52) EGFR II 53 and 28 13 and 14 
Erlotinib (53, 54) EGFR II 110 and 38 11 and 3 
Pazopanib (55) VEGFR and PDGFR II 35 
Tipifarnib (56) Farnesyltransferase II 67 12 
Temsirolimus (57) mTOR II 65 and 41 8 and 3 
Cediranib (58, 59) VEGFR II and III 31 and 131 26 and 16 
Bevacizumab (60, 61) VEGF-A II 85 and 48 36 and 29 
Aflibercept (62) VEGF-A II 42 
Cilengitide (63, 64) αvβ3,αvβ5 integrins II 81 and 26 15 and 12 
Vorinostat (65) HDAC II 66 15 
Enzastaurin (66, 67) PKC II and III 72 and 174 7 and 11 
Cabozantinib (68) EGFR and C-MET II 124 21 

Abbreviations: c-kit, stem cell factor receptor; C-MET, met-protooncogene.

Adapted and updated from ref. (69): Seminars in Oncology, Volume 38, Supplement 4, Gilbert MR, pp. S21–33, Copyright 2011, with permission from Elsevier.

The challenges that exist in finding effective treatments for patients with brain tumors stem from inherent heterogeneity and continual genomic transformation of the cancer, the unique microenvironment consisting of glial cells, microglia, and others, and the variable integrity of the blood–brain barrier, leading to issues with drug delivery. Furthermore, although preclinical testing often demonstrates high rates of efficacy for new agents, this has not yet translated to more effective treatment regimens. Although preclinical models are improving, they do not fully recapitulate the challenging aspects of the human disease such as genomic heterogeneity and tumor invasiveness (9). The testing of new regimens in clinical trials in this patient population is further complicated by limitations in current assessment of treatment efficacy. Standard imaging technologies, such as MRI, can be used to quantify the area (or volume) of abnormality. However, treatment may evoke reactions within the brain parenchyma that emulate tumor and may be incorrectly interpreted as treatment failure (pseudoprogression, Fig. 1). Conversely, some therapies, such as agents that target angiogenesis, may alter blood–brain barrier permeability, decreasing diffusion of the imaging contrast agent leading to an “improvement” in imaging that may not reflect an overall improvement or decrease in tumor burden (pseudoresponse, Fig. 2). In addition, incorporating measures of patient outcomes such as neurocognitive function, symptom burden, and quality of life have been successfully done, but now need to be integrated into the overall metrics of treatment efficacy.

Figure 1.

Pseudoprogression: an example of spontaneous improvement of imaging findings after completion of concurrent temozolomide and radiotherapy for glioblastoma. A, before surgical resection. B, after surgery. C, after concurrent temozolomide and radiation. D, after three cycles of adjuvant temozolomide (4 months after completion of radiation). E, after nine cycles of temozolomide.

Figure 1.

Pseudoprogression: an example of spontaneous improvement of imaging findings after completion of concurrent temozolomide and radiotherapy for glioblastoma. A, before surgical resection. B, after surgery. C, after concurrent temozolomide and radiation. D, after three cycles of adjuvant temozolomide (4 months after completion of radiation). E, after nine cycles of temozolomide.

Close modal
Figure 2.

Pseudoresponse: a patient treated with bevacizumab. Within 6 days of treatment there is a dramatic decrease in contrast enhancement, but the T2 abnormality remains. A, pretreatment T2 image. B, pretreatment after-contrast image. C, T2 image 6 days after bevacizumab. D, after-contrast image 6 days after bevacizumab.

Figure 2.

Pseudoresponse: a patient treated with bevacizumab. Within 6 days of treatment there is a dramatic decrease in contrast enhancement, but the T2 abnormality remains. A, pretreatment T2 image. B, pretreatment after-contrast image. C, T2 image 6 days after bevacizumab. D, after-contrast image 6 days after bevacizumab.

Close modal

Although progress has been made, particularly in our understanding of the complex biology of malignant brain tumors, these advances in tumor biology have not been translated into the clinic. Many studies in both newly diagnosed and recurrent brain tumors have been completed; some of the key studies are listed in Table 2. However, these efforts have only led to a few therapeutic successes that have resulted in new standards of care. Nonetheless, even the negative studies have helped to create clinical trial platforms that set the stage for the next generation of multifaceted studies that can include mandatory tumor tissue with real-time analysis for patient selection and stratification as well as incorporation of measures of patient outcome with successful longitudinal assessment of quality-of-life measures, symptom burden, and formal neurocognitive testing. The articles in this edition of CCR Focus highlight some of the advances that will be incorporated into the next generation of clinical and translational research. It is through these collaborative efforts, integrating the cutting edge scientific discoveries with novel clinical trial designs, that the therapeutic advances will occur.

Table 2.

Recent randomized trials for adult primary brain tumors

Trial nameDisease typeTreatment(s) testedTrial phaseOutcomes; Notes
EORTC 26981 (5) Newly diagnosed GBM Temozolomide-radiation vs. radiation III Temozolomide radiation with improved OS, PFS, and 2-year survival rate 
RTOG 0525 (70) Newly diagnosed GBM Dose dense vs. standard temozolomide III No improvement in OS or PFS with dose dense 
RTOG 0825 (38) Newly diagnosed GBM Standard chemoradiation + bevacizumab vs. standard chemoradiation III No improvement in OS; PFS longer but not significant; patient outcomes worse with bevacizumab 
AvaGLIO (46) Newly diagnosed GBM Standard chemoradiation + bevacizumab vs. standard chemoradiation III No improvement in OS; PFS significantly prolonged; QOL maintained longer with bevacizumab 
CENTRIC (71) Newly diagnosed GBM Standard treatment + cilengitide vs. standard treatment III No improvement in OS or PFS 
Enzastaurin (67) Recurrent GBM Enzastaurin vs. lomustine III No improvement in OS or PFS with enzastaurin 
REGAL (59) Recurrent GBM Cedirinib + lomustine vs. single-agent cedirinib vs. single-agent lomustine III No improvement in OS or PFS with cedirinib alone or in combination with lomustine 
BELOB (26) Recurrent GBM Bevacizumab + lomustine vs. single-agent bevacizumab vs. single-agent lomustine II Primary endpoint 9-month OS; only bevacizumab + lomustine reached efficacy threshold 
RTOG 9402 (23) Newly diagnosed AO (grade 3) Intensive PCV, then radiation vs. radiation alone III Improved OS with PCV only for tumors with 1p 19q LOH 
EORTC 26951 (22) Newly diagnosed AO (grade 3) Radiation, then standard PCV vs. radiation alone III Improved OS with PCV only for tumors with 1p 19q LOH 
RTOG 9802 (21) Newly diagnosed low-grade (2) glioma Radiation, then standard PCV vs. radiation alone III Improved OS with PCV; note: molecular subtyping not yet reported 
Trial nameDisease typeTreatment(s) testedTrial phaseOutcomes; Notes
EORTC 26981 (5) Newly diagnosed GBM Temozolomide-radiation vs. radiation III Temozolomide radiation with improved OS, PFS, and 2-year survival rate 
RTOG 0525 (70) Newly diagnosed GBM Dose dense vs. standard temozolomide III No improvement in OS or PFS with dose dense 
RTOG 0825 (38) Newly diagnosed GBM Standard chemoradiation + bevacizumab vs. standard chemoradiation III No improvement in OS; PFS longer but not significant; patient outcomes worse with bevacizumab 
AvaGLIO (46) Newly diagnosed GBM Standard chemoradiation + bevacizumab vs. standard chemoradiation III No improvement in OS; PFS significantly prolonged; QOL maintained longer with bevacizumab 
CENTRIC (71) Newly diagnosed GBM Standard treatment + cilengitide vs. standard treatment III No improvement in OS or PFS 
Enzastaurin (67) Recurrent GBM Enzastaurin vs. lomustine III No improvement in OS or PFS with enzastaurin 
REGAL (59) Recurrent GBM Cedirinib + lomustine vs. single-agent cedirinib vs. single-agent lomustine III No improvement in OS or PFS with cedirinib alone or in combination with lomustine 
BELOB (26) Recurrent GBM Bevacizumab + lomustine vs. single-agent bevacizumab vs. single-agent lomustine II Primary endpoint 9-month OS; only bevacizumab + lomustine reached efficacy threshold 
RTOG 9402 (23) Newly diagnosed AO (grade 3) Intensive PCV, then radiation vs. radiation alone III Improved OS with PCV only for tumors with 1p 19q LOH 
EORTC 26951 (22) Newly diagnosed AO (grade 3) Radiation, then standard PCV vs. radiation alone III Improved OS with PCV only for tumors with 1p 19q LOH 
RTOG 9802 (21) Newly diagnosed low-grade (2) glioma Radiation, then standard PCV vs. radiation alone III Improved OS with PCV; note: molecular subtyping not yet reported 

Abbreviations: AO, anaplastic oligodendroglioma; GBM, glioblastoma; PCV, procarbazine, CCNU (lomustine), vincristine; QOL, quality of life.

The past two decades have been remarkable for a dramatic increase in our knowledge of the tumor biology of these cancers. Using advanced molecular testing platforms, collaborative groups such as the one organized by The Cancer Genome Atlas and others have yielded important insights into the heterogeneity of these tumors (10–12). Classical histopathology provides a tumor grading system that has been traditionally used for classification and prognosis, but a variety of molecular markers have been described that are increasingly being used to modify and subtype tumors within each of the tumor grades. The article by Huse and Aldape (13) in this CCR Focus section provides a concise overview of the recent molecular findings and how these are affecting disease classification and, more importantly, how these markers are being used in prospective clinical trials. These authors offer extensive supporting data that molecular testing provides a critical advance in disease classification and at the current time should be used in conjunction with classical histopathology. These enhanced classification efforts may also provide potential therapeutic targets and better patient selection for specific treatments.

As with advancing treatment for other cancers, the foundation of determining efficacy continues to be the successful completion of robust clinical trials. These studies in brain tumors are proven to be challenging for several reasons.

Malignant primary brain tumors are relatively uncommon. Even the most common, glioblastoma, has fewer than 15,000 patients diagnosed in the United States annually (14). Most patients do not participate in clinical trials either because of limited interest or access or ineligibility due to their poor performance status. Increasingly, however, these accrual issues are being circumvented by collaborations among the major cooperative groups and/or centers.

Clinical trials will need to incorporate molecular profiling into the study design to account for differences in tumor prognosis or likelihood of response to specific treatments (predictive factors). Although molecular subtyping may make fewer patients eligible for a trial and make accrual issues even more challenging, these factors may be offset by the use of efficient clinical trial designs that provide such robust and tumor-specific treatment effects that smaller numbers of patients will be required to determine a measurable improvement.

Molecular classification of most primary brain tumors has identified many potential targets (Text Box 1); to date none have translated into specific treatments. This situation may improve with the implementation of clinical trials that incorporate mandatory tumor profiling and study designs that are hypothesis driven, based on assessing the presence of the target and tumor response.

Text Box 1. Potential molecular targets in glioblastoma

  • Cell surface growth factor receptors

    • Epidermal growth factor receptor (EGFR)

    • Platelet-derived growth factor receptor (PDGFR)

    • Fibroblast growth factor receptor (FGFR)

    • Transforming growth factor receptor (TGFR)

    • Vascular endothelial growth factor receptor (VEGFR)

    • Insulin-like growth factor receptor (IGFR)

  • Signaling transduction pathway and downstream effectors

    • PI3K/Akt

    • Ras/mitogen-activated protein kinase (MAPK)

    • Mammalian target of rapamycin (mTOR)

    • Protein kinase C (PKC)

    • TP53

    • MET (hepatocyte growth factor, Met-protooncogene)

    • Janus kinase/signal transducer and activator of transcription (JAK/STAT)

    • Smoothened

    • Notch

    • Retinoblastoma (Rb) signaling

    • Farnesyltransferase

  • Angiogenesis

    • VEGFR

    • Vascular endothelial growth factor (VEGF)

    • Integrins

    • Angiopoietin 1/2

  • Other targets

    • Histone deacetylase (HDAC)

    • Isocitrate dehydrogenase mutations (IDH1 and IDH2)

    • Poly (adenosine–diphosphase–ribose) polymerase (PARP)

For many targets, it is not known whether if the target was present in the newly diagnosed tumor, it will still be present at progression. This has important implications for patient management and targeted therapies and may require resampling tumor.

Drug delivery in the presence of a variably intact blood–brain barrier remains a challenge. Increasingly, preclinical testing has evaluated drug delivery and some agents have undergone early clinical testing that included measuring drug concentrations within tumor samples (pharmacokinetics) and the impact on the drug target (pharmacodynamics). These studies, however, require extensive resources and have been difficult to complete.

Accrual issues, the increasing portfolio of possible treatment agents, and the recognition that combination regimens will likely be required have led to increasing interest in developing novel trial designs that can efficiently screen for efficacy. Unfortunately, the results from single-arm phase II trials, particularly in patients with newly diagnosed tumors, have been difficult to interpret; therefore, randomized studies are being increasingly used (15). An overview of many of the currently used designs in brain tumor clinical trials was recently published (16). One of the novel designs, called the factorial design, permits simultaneous evaluation of multiple permutations of combinations of a set of agents. A study has been successfully completed in patients with glioblastoma that evaluated three experimental agents in combination with temozolomide during the adjuvant phase of treatment. For this trial, only 20 patients per arm were required to provide adequate power to determine the impact of each individual agent and to determine whether there was improved benefit of triplet therapy (temozolomide plus two experimental agents) compared with doublet treatment regimens (17).

The adaptive randomized design begins by completely random allocation of patients to each of the arms, but the weighting of randomization is altered by efficacy data (18). Treatment arms with the most success get a higher rating and are more likely to get patients. If there is a real efficacy difference, the randomization weighting will continue until one treatment is declared the winner. This design may reduce accrual needs while maintaining the structure of a randomized trial. Adaptively designed trials are in progress for brain tumors, but no results have been reported to date.

The randomized discontinuation design has not been used in brain tumor trials but may be particularly well suited for the brain tumor patient population as the design focuses on patients with “stable disease.” All patients are treated with the new regimen for a set period of time. Treatment responders continue therapy and patients with disease progression or unacceptable toxicity stop. Patients with no significant change (stable disease) are then randomized to active agent or placebo. At a set time later, a comparison of the progression-free survival (PFS) rate is made between agent and placebo. An improvement in PFS rate with the experimental agent is a strong evidence of treatment efficacy. This strategy, which requires far fewer patients than a traditional randomized phase II or III study, has been successfully used in a clinical trial in renal cell carcinoma (19).

Assessment of treatment response in brain tumor clinical trials has been challenging. The irregular and infiltrative nature of these cancers makes traditional measures often difficult and creates variability of quantitation and interpretation. There are ongoing discussions regarding the optimal endpoint measures with each of the conventional cancer clinical trial measures as described below.

Overall survival (OS) is definitive and universally recognized as a robust endpoint. However, crossover or salvage treatments may alter the ability to assess efficacy if a variety of treatments are used in salvage among the study patients at disease recurrence. This is most germane for the lower-grade gliomas in which effective salvage regimens have been developed that may, therefore, affect OS independent of the protocol treatment. In addition, survival studies require long follow-up for some brain tumors, for example in patients with low-grade (WHO grade 2) and some anaplastic (WHO grade 3) gliomas (20–23).

PFS has the advantage of not having the potential problem of crossover or varied use of salvage regimens. However, the definition of disease progression as determined by conventional imaging may be confounded by treatment-related changes. Concurrent assessment of clinical status may improve the determination of disease status, but as described below, there is no consensus on the optimal outcomes measures.

Objective response rate (ORR) has been the traditional measure in cancer studies as a screening tool for treatment activity. For most solid tumors, cross-sectional area has been replaced by single dimension (RECIST) measure, whereas in brain tumor studies, complex tumor shapes may require two-dimensional or even volume assessment (24). As with assessment of progression, some treatments may alter imaging, hampering the ability to accurately determine response.

Finally, set time point rates report the percentage of patients reaching a preset milestone time. For example, in many studies in recurrent glioblastoma, a 6-month PFS rate has been used as a metric based on strong historical data that have provided this measure for ineffective treatments (expected probability; P0) in the statistical design (25). Some examples of brain tumor clinical trials that reported the 6-month PFS rate are listed in Table 1. Survival rate can also be used as an endpoint. In the BELOB trial the 9-month OS rate was the primary measure of efficacy (26). These measures have been primarily used for screening for efficacy, and the rate determinations are subject to the same limitations as described for PFS (imaging) and OS (crossover or salvage treatment) metrics reported above.

Until recently, most clinical trials for malignant primary brain tumors used measures of cross-sectional area of enhancing tissue as an indication of tumor burden (27). However, it has been increasingly recognized that certain treatments may alter the degree of contrast enhancement without necessarily affecting tumor size. This is particularly germane for antiangiogenic therapies in which normalization of blood–brain barrier permeability may reduce overall tumor enhancement. This concern led to the formation of a consensus group, Response Assessment in Neuro-oncology (RANO), which has published guidelines for image analysis that include assessment of corticosteroid use, neurologic function, and nonenhancing (T2 or T2FLAIR) abnormalities in addition to the enhancing lesion (28). A summary of these guidelines is provided in Table 3. Further work is ongoing to modify the RANO guidelines to accommodate imaging changes that are anticipated with the increasing use of immunotherapies and the potential for inflammatory changes that could be misinterpreted as tumor progression rather than a desired immune response.

Table 3.

Summary of RANO criteria

CriterionCRPRSDPD
T1 gadolinium–enhancing disease None ≥50% ↓ <50% ↓ but < 25% ↑ ≥25% ↑a 
T2/FLAIR Stable or ↓ Stable or ↓ Stable or ↓ a 
New lesion None None None Presenta 
Corticosteroids None Stable or ↓ Stable or ↓ NAb 
Clinical status Stable or ↑ Stable or ↑ Stable or ↑ a 
Requirement for response All All All Anya 
CriterionCRPRSDPD
T1 gadolinium–enhancing disease None ≥50% ↓ <50% ↓ but < 25% ↑ ≥25% ↑a 
T2/FLAIR Stable or ↓ Stable or ↓ Stable or ↓ a 
New lesion None None None Presenta 
Corticosteroids None Stable or ↓ Stable or ↓ NAb 
Clinical status Stable or ↑ Stable or ↑ Stable or ↑ a 
Requirement for response All All All Anya 

Abbreviations: CR, complete response; FLAIR, fluid-attenuated inversion recovery; NA, not applicable; PD, progressive disease; PR, partial response; SD, stable disease.

aProgression occurs when this criterion is present.

bIncrease in corticosteroids alone will not be taken into account in determining progression in the absence of persistent clinical deterioration.

Reprinted with permission from Wen et al. (28). © 2010 American Society of Clinical Oncology. All rights reserved. Wen PY, Macdonald DR, Reardon DA, Cloughesy TF, Sorensen AG, Galanis E, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group. J Clin Oncol 2010;28:1963–72.

Hindered by the difficulties associated with interpreting most conventional brain imaging (typically MRI), there is great interest in developing advanced imaging techniques to help determine whether imaging improvements represent true response to treatment or “pseudoresponse,” or conversely, whether worsening is due to growth or “pseudoprogression” (29). Advanced MRI techniques include magnetic resonance spectroscopy (MRS), which can quantify specific molecular compounds in an area of interest (reviewed in ref. 30). Most conventional scanners can provide measures of choline, creatine, lactate, N-acetyl aspartate (NAA), and other compounds. Characteristic signatures of active tumor (high choline to creatine) or necrosis (high lactate, low NAA) are used in clinical practice, although the ability to definitively determine tumor or necrosis may be limited. In addition, tumor metabolites such as 2-hydroxyglutarate in tumors with isocitrate dehydrogenase mutations can be quantified using MRS and may prove to be a noninvasive biomarker. Other advanced imaging techniques, including dynamic contrast–enhanced MRI and dynamic susceptibility contrast MRI, may provide additional information particularly regarding integrity of tumor vasculature and blood volume (30), and can be helpful in differentiating recurrent disease from “pseudoprogression.”

PET using 18-fluorodeoxyglucose has been performed for decades in patients with brain tumors (29). Initially thought to be highly specific for tumor, false-positive testing particularly with inflammatory changes, as well as high background activity within the cortex and other metabolically active structures, has limited the clinical utility of this test. However, other PET tracers are under active investigation, including 18F-fluorodopa-, fluorethyltyrosine-, methionine-, and thymidine-based tracers. PET technology may be particularly helpful in determining the ability of an agent to penetrate the blood–brain barrier and be delivered to the tumor.

There is increasing recognition of the importance of evaluating the impact of disease-modifying therapy on the patient as a measure of clinical benefit. Symptomatic benefit is recognized by the FDA as a measure of clinical benefit, and within the spectrum of oncology several examples demonstrate how these measures have been used to support the approval of treatments by the FDA (31). These measures, referred to as Clinical Outcome Assessments (COA), may include patient-reported outcomes, observed outcomes, or outcomes that are clinically assessed. Patient-reported outcomes were incorporated into one of the pivotal studies of ruxolinitib in myelofibrosis. In this trial, incorporation of a symptom scale specific to myelofibrosis provided compelling evidence of clinical improvement in the setting of disease response measured by spleen size (32). This sets the stage for the use of patient-reported outcomes in glioblastoma in which, depending on location, relatively small changes in tumor size can have a major impact on symptom burden and functional status.

Patient-reported outcomes

A variety of COAs have been used in brain tumor clinical trials (33). Measures of health-related quality of life have been incorporated into many of the randomized studies mainly using either the EORTC QLQ30 with the additional brain-specific BN-20 or the FACT-BR, which is also brain specific. These instruments are easy to administer and may provide important insights regarding impact of therapies and have prognostic implications, but a systematic review raises concerns that the value may be limited unless specific hypotheses are developed and an analysis plan is constructed before the implementation of the clinical trial (34, 35).

Several instruments that measure symptom burden in patients with cancer have been developed and are being increasingly used in the assessment of treatment efficacy (i.e., for ruxolinitib, discussed previously). However, currently the only symptom instrument designed specifically for the brain tumor patient population is the MD Anderson Symptom Inventory-Brain Tumor (MDASI-BT; ref. 36). This instrument contains 23 items, and patients rate the severity of their symptoms and the impact of the symptoms on activities of daily life (interference). Recent clinical trials have demonstrated that the MDASI-BT is sensitive to tumor progression, may predict OS and PFS, and is sensitive to differences in treatment arms (37, 38).

Neurocognitive testing

Formal neurocognitive testing provides quantitative, objective measures of patient outcomes that may be particularly germane in patients with brain tumors in which there is often pretreatment loss of function and in which longitudinal assessments may provide insights into the impact of treatment on function. Loss of neurocognitive function has also been reported to precede imaging evidence of tumor progression (39). The results from recent clinical trials in brain tumors demonstrate that baseline neurocognitive function may predict OS and PFS and, with the longitudinal assessment, can detect differences in treatment arms (37, 38, 40).

Signal transduction modulators

The current standard of care for patients with malignant gliomas remains some permutation of surgery for diagnosis and cytoreduction followed by radiation and cytotoxic chemotherapy. For glioblastoma, concurrent temozolomide with radiation followed by 6 to 12 months of maintenance temozolomide remains the international standard. For anaplastic gliomas (grade 3), a molecular test looking for allelic loss of chromosome arm 1p and 19q determines whether a combination of radiotherapy and chemotherapy is needed (1p 19q codeleted) or if the addition of chemotherapy provides no survival benefit (1p19q not codeleted). Recent results also suggest that poor- risk low-grade gliomas (age >40 or incomplete resection) also benefit from radiotherapy and chemotherapy (21). More specific recommendations based on molecular profiling will be forthcoming.

Despite the extensive knowledge about the molecular profiles of malignant glioma, no successful treatments have been developed for primary brain tumors using existing signal transduction modulators. This lack has been ascribed to the large number of pathway abnormalities and the inter-networking of signaling pathways. For most malignant gliomas, by the time a diagnosis is made there is such intratumoral heterogeneity that identifying a driver mutation is very difficult (41). The recent discovery of a chromosomal translocation of coding domains of fibroblast growth factor receptor (FGFR) to transforming acidic coiled-coil (TACC) coding domains resulting in a specific fusion protein, FGFR–TACC, that is likely a tumor driver is encouraging (42). Although this fusion occurs in only 3% of glioblastoma, if the ongoing clinical trials specifically targeting this unique protein are successful, it will provide proof of principle of this approach and encourage continued investigations.

Combination treatment regimens that incorporate agents to either target multiple signaling pathways or use multiple agents to inhibit one pathway may have more efficacy than the single-agent regimens. These studies are logistically difficult as they require the integration of molecular analysis of contemporary tumor samples coupled with an efficient selection design and often require cooperation between pharmaceutical companies (43). These studies either need to treat only tumors with specific profiles or have several treatment arms to accommodate a variety of tumor subtypes. In addition, despite a typically modest toxicity profile when a drug is used as a single agent, combination regimens have demonstrated more severe toxicity than expected. Therefore, safety testing (phase I) has been required for these combination regimens.

Antiangiogenic therapy

Vascular proliferation, or angiogenesis, is one of the hallmarks of glioblastoma and accounts for the contrast enhancement on imaging studies. The VEGF pathway is the most prominent mechanism for angiogenesis in glioblastoma and VEGF-A most commonly expressed in response to tumor hypoxia (44). Angiogenesis was therefore a logical therapeutic target and after initial concerns regarding the risk of intracranial hemorrhage were allayed by early clinical trials, a variety of antiangiogenic agents were tested in high-grade glioma. The article in this CCR Focus by Batchelor and colleagues (45) provides an overview of the putative mechanisms of angiogenesis and a review of the clinical trials that have been undertaken. Importantly, the authors also provide insights into the potential mechanisms of resistance and treatment failure. This is particularly important in light of the recent phase III trials testing bevacizumab in patients with newly diagnosed glioblastoma that showed no survival benefit (38, 46), raising questions about whether, despite its prominent role in glioblastoma biology, angiogenesis should remain a prime therapeutic target.

Immunotherapy

Immunotherapy for malignant primary brain tumors has been tried for decades. Early studies using brain tumor extracts as vaccines were not successful. Other studies involved cytokines, including interferons, which showed only minimal efficacy, or interleukins, which led to major toxicity, particularly increases in intracranial pressure. With the recent substantial increase in the understanding of tumor immunology and successes in other cancers, particularly metastatic melanoma, there has been a marked surge of interest in investigating immunotherapies for primary brain tumors. The article by Fecci and colleagues (47) in this CCR Focus reviews the increasing evidence that the brain is not “immunoprivileged” and that pursuing immunotherapy may be a very important therapeutic strategy. These authors provide a summary of the approaches that have been used as immunotherapies for brain tumors in the context of the increasing understanding of tumor-related immunosuppression. They also present an overview of the ongoing and planned clinical trials providing the additional perspective of immunotherapy research in other cancers.

Pediatric brain tumors, although uncommon, represent the second most common cancer of childhood and a leading cause of cancer-related death. The development of new treatments faces accrual challenges given the relative rarity of the disease, issues with response assessments, and often limited access to new agents because of safety concerns (48). In addition to the typical issues of treatment-related toxicity with cancer treatment, impact on the developing nervous system must be considered. For example, treatment of CNS cancers in young patients typically avoids or delays radiotherapy because of the profound effect on brain development.

Despite these limitations and challenges, recent seminal discoveries in childhood brain tumors highlight the importance of basic research and the potential to translate these laboratory findings into the clinical treatment decision making process. The article by Gajjar and colleagues (49) in this CCR Focus section highlights the culmination of collaborative efforts in two major pediatric brain tumors, medulloblastoma and ependymoma. These authors review the seminal findings for each disease and discuss the potential clinical implications for prognosis, treatment decisions, and ultimately for devising disease subtype–specific therapies.

Despite decades of research, most primary brain tumors remain incurable. However, there has been a marked increase in our knowledge and understanding of the underlying tumor biology along with the complexity and heterogeneity of these cancers. Parallel efforts are ongoing to develop multifaceted and efficient clinical–translational studies that use tumor molecular profiles for hypothesis-based clinical trials and incorporate advanced imaging and patient outcomes measures to fully realize the potential from each trial. These goals are realistic because the recent surge of collaborations within the field indicate that a global effort is needed to actuate a real improvement in outcomes for these patients. The articles in this CCR Focus provide a snapshot of some of the exciting work that is under way.

M.R. Gilbert reports receiving commercial research grants from Genentech, GlaxoSmithKline, and Merck, and is a consultant/advisory board member for AbbVie, Genentech, Heron Therapeutics, and Merck. T.S. Armstrong reports receiving a commercial research grant from Merck, other commercial research support from Genentech, and is a consultant/advisory board member for Roche. W.B. Pope is a consultant/advisory board member for Roche. M.J. van den Bent reports receiving commercial research grants from AbbVie and Roche; speakers bureau honoraria from Merck; and is a consultant/advisory board member for AbbVie, Amgen, CellDex Therapeutics, and Roche. P.Y. Wen reports receiving commercial research grants from Agios, Angiochem, AstraZeneca, Genentech, GlaxoSmithKline, Karyopharm Therapeutics, Novartis, and Sanofi; speakers bureau honoraria from Merck; and is a consultant/advisory board member for AbbVie, CellDex Therapeutics, Midatech, Novartis, Roche, Sigma-Tau Pharmaceuticals, and Vascular Biogenics. No other potential conflicts of interest were disclosed.

Conception and design: M.R. Gilbert, T.S. Armstrong, W.B. Pope

Development of methodology: M.R. Gilbert, W.B. Pope

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M.R. Gilbert

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M.R. Gilbert, W.B. Pope

Writing, review, and/or revision of the manuscript: M.R. Gilbert, T.S. Armstrong, W.B. Pope, M.J. van den Bent, P.Y. Wen

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M.R. Gilbert

1.
Sanai
N
,
Berger
MS
. 
Recent surgical management of gliomas
.
Adv Exp Med Biol
2012
;
746
:
12
25
.
2.
Sanai
N
,
Berger
MS
. 
Extent of resection influences outcomes for patients with gliomas
.
Rev Neurol
2011
;
167
:
648
54
.
3.
Nabors
LB
,
Ammirati
M
,
Bierman
PJ
,
Brem
H
,
Butowski
N
,
Chamberlain
MC
, et al
Central nervous system cancers
.
J Natl Compr Canc Netw
2013
;
11
:
1114
51
.
4.
Amelio
D
,
Lorentini
S
,
Schwarz
M
,
Amichetti
M
. 
Intensity-modulated radiation therapy in newly diagnosed glioblastoma: a systematic review on clinical and technical issues
.
Radiother Oncol
2010
;
97
:
361
9
.
5.
Stupp
R
,
Mason
WP
,
van den Bent
MJ
,
Weller
M
,
Fisher
B
,
Taphoorn
MJ
, et al
Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma
.
N Engl J Med
2005
;
352
:
987
96
.
6.
Omuro
A
,
DeAngelis
LM
. 
Glioblastoma and other malignant gliomas: a clinical review
.
JAMA
2013
;
310
:
1842
50
.
7.
Bruce
JN
,
Fine
RL
,
Canoll
P
,
Yun
J
,
Kennedy
BC
,
Rosenfeld
SS
, et al
Regression of recurrent malignant gliomas with convection-enhanced delivery of topotecan
.
Neurosurgery
2011
;
69
:
1272
9
;
discussion 1279–80
.
8.
Lesniak
MS
,
Langer
R
,
Brem
H
. 
Drug delivery to tumors of the central nervous system
.
Curr Neurol Neurosci Rep
2001
;
1
:
210
6
.
9.
Rankin
SL
,
Zhu
G
,
Baker
SJ
. 
Review: insights gained from modelling high-grade glioma in the mouse
.
Neuropathol Appl Neurobiol
2012
;
38
:
254
70
.
10.
Verhaak
RG
,
Hoadley
KA
,
Purdom
E
,
Wang
V
,
Wilkerson
MD
,
Miller
CR
, et al
Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1
.
Cancer Cell
2010
;
17
:
98
110
.
11.
Brennan
CW
,
Verhaak
RG
,
McKenna
A
,
Campos
B
,
Noushmehr
H
,
Salama
SR
, et al
The somatic genomic landscape of glioblastoma
.
Cell
2013
;
155
:
462
77
.
12.
Yan
H
,
Parsons
DW
,
Jin
G
,
McLendon
R
,
Rasheed
BA
,
Yuan
W
, et al
IDH1 and IDH2 mutations in gliomas
.
N Engl J Med
2009
;
360
:
765
73
.
13.
Huse
JT
,
Aldape
KD
. 
The evolving role of molecular markers in the diagnosis and management of diffuse glioma
.
Clinical Cancer Res
2014
;
20
:
5601
11
.
14.
Ostrom
QT
,
Gittleman
H
,
Farah
P
,
Ondracek
A
,
Chen
Y
,
Wolinsky
Y
, et al
CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2006–2010
.
Neuro Oncol
2013
;
15
:
ii1
56
.
15.
Grossman
SA
,
Ye
X
,
Piantadosi
S
,
Desideri
S
,
Nabors
LB
,
Rosenfeld
M
, et al
Survival of patients with newly diagnosed glioblastoma treated with radiation and temozolomide in research studies in the United States
.
Clin Cancer Res
2010
;
16
:
2443
9
.
16.
Galanis
E
,
Wu
W
,
Cloughesy
T
,
Lamborn
K
,
Mann
B
,
Wen
PY
, et al
Phase 2 trial design in neuro-oncology revisited: a report from the RANO group
.
Lancet Oncol
2012
;
13
:
e196
204
.
17.
Penas-Prado
M
,
Hess
KR
,
Fisch
MJ
,
Lagrone
LW
,
Groves
MD
,
Levin
VA
, et al
Randomized phase II adjuvant factorial study of dose-dense temozolomide alone and in combination with isotretinoin, celecoxib, and/or thalidomide for glioblastoma
.
Neuro Oncol
2014 Sep 19
.
[Epub ahead of print]
.
18.
Berry
DA
. 
Bayesian clinical trials
.
Nat Rev Drug Discov
2006
;
5
:
27
36
.
19.
Ratain
MJ
,
Eisen
T
,
Stadler
WM
,
Flaherty
KT
,
Kaye
SB
,
Rosner
GL
, et al
Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma
.
J Clin Oncol
2006
;
24
:
2505
12
.
20.
Reardon
DA
,
Galanis
E
,
DeGroot
JF
,
Cloughsey
TF
,
Wefel
JS
,
Lamborn
KR
, et al
Clinical trial end points for high-grade glioma: the evolving landscape
.
Neuro Oncol
2011
;
13
:
353
61
.
21.
Buckner
JC
,
Pugh
SL
,
Shaw
EG
,
Gilbert
MR
,
Barger
G
,
Coons
S
, et al
Phase III study of radiation therapy (RT) with or without procarbazine, CCNU, and vincristine (PCV) in low-grade glioma: RTOG 9802 with Alliance, ECOG and SWOG
.
JClin Oncol
2014
;
32
:
5s
, (
suppl; abstr 2000
).
22.
van den Bent
MJ
,
Brandes
AA
,
Taphoorn
MJ
,
Kros
JM
,
Kouwenhoven
MC
,
Delattre
JY
, et al
Adjuvant procarbazine, lomustine, and vincristine chemotherapy in newly diagnosed anaplastic oligodendroglioma: long-term follow-up of EORTC brain tumor group study 26951
.
J Clin Oncol
2013
;
31
:
344
50
.
23.
Cairncross
G
,
Wang
M
,
Shaw
E
,
Jenkins
R
,
Brachman
D
,
Buckner
J
, et al
Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402
.
J Clin Oncol
2013
:
31
:
337
43
.
24.
Ellingson
BM
,
Cloughesy
TF
,
Lai
A
,
Nghiemphu
PL
,
Mischel
PS
,
Pope
WB
. 
Quantitative volumetric analysis of conventional MRI response in recurrent glioblastoma treated with bevacizumab
.
Neuro Oncol
2011
;
13
:
401
9
.
25.
Wong
ET
,
Hess
KR
,
Gleason
MJ
,
Jaeckle
KA
,
Kyritsis
AP
,
Prados
MD
, et al
Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials
.
J CLin Oncol
1999
;
17
:
2572
.
26.
Taal
W
,
Oosterkamp
HM
,
Walenkamp
AM
,
Dubbink
HJ
,
Beerepoot
LV
,
Hanse
MC
, et al
Single-agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial
.
Lancet Oncol
2014
;
15
:
943
53
.
27.
Macdonald
DR
,
Cascino
TL
,
Schold
SC
 Jr
,
Cairncross
JG
. 
Response criteria for phase II studies of supratentorial malignant glioma
.
J Clin Oncol
1990
;
8
:
1277
80
.
28.
Wen
PY
,
Macdonald
DR
,
Reardon
DA
,
Cloughesy
TF
,
Sorensen
AG
,
Galanis
E
, et al
Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group
.
J Clin Oncol
2010
;
28
:
1963
72
.
29.
Peet
AC
,
Arvanitis
TN
,
Leach
MO
,
Waldman
AD
. 
Functional imaging in adult and paediatric brain tumours
.
Nat Rev Clin Oncol
2012
;
9
:
700
11
.
30.
Kalpathy-Cramer
J
,
Gerstner
ER
,
Emblem
KE
,
Andronesi
OC
,
Rosen
B
. 
Advanced magnetic resonance imaging of the physical processes in human glioblastoma
.
Cancer Res
2014
;
74
:
4622
37
.
31.
Deisseroth
A
,
Kaminskas
E
,
Grillo
J
,
Chen
W
,
Saber
H
,
Lu
HL
, et al
U.S. Food and Drug Administration approval: ruxolitinib for the treatment of patients with intermediate and high-risk myelofibrosis
.
Clin Cancer Res
2012
;
18
:
3212
7
.
32.
Verstovsek
S
,
Mesa
RA
,
Gotlib
J
,
Levy
RS
,
Gupta
V
,
DiPersio
JF
, et al
A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis
.
N Engl J Med
2012
;
366
:
799
807
.
33.
Armstrong
TS
,
Gilbert
MR
. 
Patient reported endpoints for measuring clinical benefit in (high grade glioma) primary brain tumor patients
.
Curr Treat Options Oncol
2014 Aug 31
.
[Epub ahead of print]
.
34.
Mauer
ME
,
Taphoorn
MJ
,
Bottomley
A
,
Coens
C
,
Efficace
F
,
Sansom
M
, et al
Prognostic value of health-related quality-of-life data in predicting survival in patients with anaplastic oligodendrogliomas, from a phase III EORTC brain cancer group study
.
J Clin Oncol
2007
;
25
:
5731
7
.
35.
Dirven
L
,
Taphoorn
MJ
,
Reijneveld
JC
,
Blazeby
J
,
Jacobs
M
,
Pusic
A
, et al
The level of patient-reported outcome reporting in randomised controlled trials of brain tumour patients: a systematic review
.
Eur J Cancer
2014
;
50
:
2432
48
.
36.
Armstrong
TS
,
Mendoza
T
,
Gning
I
,
Coco
C
,
Cohen
MZ
,
Eriksen
L
, et al
Validation of the M.D. Anderson Symptom Inventory Brain Tumor Module (MDASI-BT)
.
J Neurooncol
2006
;
80
:
27
35
.
37.
Armstrong
TS
,
Wefel
JS
,
Wang
M
,
Gilbert
MR
,
Won
M
,
Bottomley
A
, et al
Net clinical benefit analysis of radiation therapy oncology group 0525: a phase III trial comparing conventional adjuvant temozolomide with dose-intensive temozolomide in patients with newly diagnosed glioblastoma
.
J Clin Oncol
2013
;
31
:
4076
84
.
38.
Gilbert
MR
,
Dignam
JJ
,
Armstrong
TS
,
Wefel
JS
,
Blumenthal
DT
,
Vogelbaum
MA
, et al
A randomized trial of bevacizumab for newly diagnosed glioblastoma
.
N Engl J Med
2014
;
370
:
699
708
.
39.
Meyers
CA
,
Hess
KR
. 
Multifaceted end points in brain tumor clinical trials: cognitive deterioration precedes MRI progression
.
Neuro Oncol
2003
;
5
:
89
95
.
40.
Meyers
CA
,
Hess
KR
,
Yung
WK
,
Levin
VA
. 
Cognitive function as a predictor of survival in patients with recurrent malignant glioma
.
J Clin Oncol
2000
;
18
:
646
50
.
41.
Sottoriva
A
,
Spiteri
I
,
Piccirillo
SG
,
Toulomis
A
,
Collins
VP
,
Marioni
JC
, et al
Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics
.
Proc Natl Acad Sci U S A
2013
;
110
:
4009
14
.
42.
Singh
D
,
Chan
JM
,
Zoppoli
P
,
Niola
F
,
Sullivan
R
,
Castano
A
, et al
Transforming fusions of FGFR and TACC genes in human glioblastoma
.
Science
2012
;
337
:
1231
5
.
43.
Alexander
BM
,
Galanis
E
,
Yung
WK
,
Ballman
KV
,
Boyett
JM
,
Cloughesy
TF
, et al
Brain Malignancy Steering Committee clinical trials planning workshop: report from the Targeted Therapies Working Group.
Neuro Oncol
2014 Aug 26
.
[Epub ahead of print]
.
44.
Chi
AS
,
Sorensen
AG
,
Jain
RK
,
Batchelor
TT
. 
Angiogenesis as a therapeutic target in malignant gliomas
.
Oncologist
2009
;
14
:
621
36
.
45.
Batchelor
TT
,
Reardon
DA
,
de Groot
JF
,
Wick
W
,
Weller
M
. 
Antiangiogenic therapy for glioblastoma: current status and future prospects
.
Clin Cancer Res
2014
;
20
:
5612
9
.
46.
Chinot
OL
,
Wick
W
,
Mason
W
,
Henriksson
R
,
Saran
F
,
Nishikawa
R
, et al
Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma
.
N Engl J Med
2014
;
370
:
709
22
.
47.
Fecci
PE
,
Heimberger
AB
,
Sampson
JH
. 
Immunotherapy for primary brain tumors: no longer a matter of privilege
.
Clin Cancer Res
2014
;
20
:
5620
9
.
48.
Warren
KE
,
Poussaint
TY
,
Vezina
G
,
Hargrave
D
,
Packer
RJ
,
Goldman
S
, et al
Challenges with defining response to antitumor agents in pediatric neuro-oncology: a report from the response assessment in pediatric neuro-oncology (RAPNO) working group
.
Pediatr Blood Cancer
2013
;
60
:
1397
401
.
49.
Gajjar
A
,
Pfister
SM
,
Taylor
MD
,
Gilbertson
RJ
. 
Molecular insights into pediatric brain tumors have the potential to transform therapy
.
Clin Cancer Res
2014
;
20
:
5630
40
.
50.
Raymond
E
,
Brandes
A
,
Van Oosterom
A
,
Dittrich
C
,
Fumoleau
P
,
Coudert
B
, et al
Multicentre phase II study of imatinib mesylate in patients with recurrent glioblastoma: an EORTC: NDDG/BTG Intergroup Study
.
J Clin Oncol
22
:
14s
, 
2004
(
suppl; abstr 1501
).
51.
Rich
JN
,
Reardon
DA
,
Peery
T
,
Dowell
JM
,
Quinn
JA
,
Penne
KL
, et al
Phase II trial of gefitinib in recurrent glioblastoma
.
J Clin Oncol
2004
;
22
:
133
42
.
52.
Franceschi
E
,
Cavallo
G
,
Lonardi
S
,
Magrini
E
,
Tosoni
A
,
Grosso
D
, et al
Gefitinib in patients with progressive high-grade gliomas: a multicentre phase II study by Gruppo Italiano Cooperativo di Neuro-Oncologia (GICNO)
.
Br J Cancer
2007
;
96
:
1047
51
.
53.
van den Bent
MJ
,
Brandes
AA
,
Rampling
R
,
Kouwenhoven
MC
,
Kros
JM
,
Carpentier
AF
, et al
Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC brain tumor group study 26034
.
J Clin Oncol
2009
;
27
:
1268
74
.
54.
Raizer
JJ
,
Abrey
LE
,
Lassman
AB
,
Chang
SM
,
Lamborn
KR
,
Kuhn
JG
, et al
A phase II trial of erlotinib in patients with recurrent malignant gliomas and nonprogressive glioblastoma multiforme postradiation therapy
.
Neuro Oncol
2010
;
12
:
95
103
.
55.
Iwamoto
FM
,
Lamborn
KR
,
Robins
HI
,
Mehta
MP
,
Chang
SM
,
Butowski
NA
, et al
Phase II trial of pazopanib (GW786034), an oral multi-targeted angiogenesis inhibitor, for adults with recurrent glioblastoma (North American Brain Tumor Consortium Study 06–02)
.
Neuro Oncol
2010
;
12
:
855
61
.
56.
Cloughesy
TF
,
Wen
PY
,
Robins
HI
,
Chang
SM
,
Groves
MD
,
Fink
KL
, et al
Phase II trial of tipifarnib in patients with recurrent malignant glioma either receiving or not receiving enzyme-inducing antiepileptic drugs: a North American Brain Tumor Consortium Study
.
J Clin Oncol
2006
;
24
:
3651
6
.
57.
Chang
SM
,
Wen
P
,
Cloughesy
T
,
Greenberg
H
,
Schiff
D
,
Conrad
C
, et al
Phase II study of CCI-779 in patients with recurrent glioblastoma multiforme
.
Invest New Drugs
2005
;
23
:
357
61
.
58.
Batchelor
TT
,
Duda
DG
,
di Tomaso
E
,
Ancukiewicz
M
,
Plotkin
SR
,
Gerstner
E
, et al
Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma
.
J Clin Oncol
2010
;
28
:
2817
23
.
59.
Batchelor
TT
,
Mulholland
P
,
Neyns
B
,
Nabors
LB
,
Campone
M
,
Wick
A
, et al
Phase III randomized trial comparing the efficacy of cediranib as monotherapy, and in combination with lomustine, versus lomustine alone in patients with recurrent glioblastoma
.
J Clin Oncol
2013
;
31
:
3212
8
.
60.
Friedman
HS
,
Prados
MD
,
Wen
PY
,
Mikkelsen
T
,
Schiff
D
,
Abrey
LE
, et al
Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma
.
J Clin Oncol
2009
;
27
:
4733
40
.
61.
Kreisl
TN
,
Kim
L
,
Moore
K
,
Duic
P
,
Royce
C
,
Stroud
I
, et al
Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma
.
J Clin Oncol
2009
;
27
:
740
5
.
62.
de Groot
JF
,
Lamborn
KR
,
Chang
SM
,
Gilbert
MR
,
Cloughesy
TF
,
Aldape
K
, et al
Phase II study of aflibercept in recurrent malignant glioma: a North American Brain Tumor Consortium study
.
J Clin Oncol
2011
;
29
:
2689
95
.
63.
Reardon
DA
,
Fink
KL
,
Mikkelsen
T
,
Cloughesy
TF
,
O'Neill
A
,
Plotkin
S
, et al
Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme
.
J Clin Oncol
2008
;
26
:
5610
7
.
64.
Gilbert
MR
,
Kuhn
J
,
Lamborn
KR
,
Lieberman
F
,
Wen
PY
,
Mehta
M
, et al
Cilengitide in patients with recurrent glioblastoma: the results of NABTC 03–02, a phase II trial with measures of treatment delivery
.
J Neurooncol
2011
;
106
:
147
53
.
65.
Galanis
E
,
Jaeckle
KA
,
Maurer
MJ
,
Reid
JM
,
Ames
MM
,
Hardwick
JS
, et al
Phase II trial of vorinostat in recurrent glioblastoma multiforme: a north central cancer treatment group study
.
J Clin Oncol
2009
;
27
:
2052
8
.
66.
Kreisl
TN
,
Kotliarova
S
,
Butman
JA
,
Albert
PS
,
Kim
L
,
Musib
L
, et al
A phase I/II trial of enzastaurin in patients with recurrent high-grade gliomas
.
Neuro Oncol
2010
;
12
:
181
9
.
67.
Wick
W
,
Puduvalli
VK
,
Chamberlain
MC
,
van den Bent
MJ
,
Carpentier
AF
,
Cher
LM
, et al
Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma
.
J Clin Oncol
2010
;
28
:
1168
74
.
68.
Wen
PY
,
Prados
M
,
Schiff
D
,
Reardon
DA
,
Cloughesy
T
,
Mikkelsen
T
, et al
Phase II study of XL184 (BMS 907351), an inhibitor of MET, VEGFR2, and RET, in patients (pts) with progressive glioblastoma (GB)
.
J Clin Oncol
28
:
15s
, 
2010
(
suppl; abstr 2006
).
69.
Gilbert
MR
. 
Recurrent glioblastoma: a fresh look at current therapies and emerging novel approaches
.
Semin Oncol
2011
;
38
:
S21
33
.
70.
Gilbert
MR
,
Wang
M
,
Aldape
KD
,
Stupp
R
,
Hegi
ME
,
Jaeckle
KA
, et al
Dose-dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial
.
J Clin Oncol
2013
;
31
:
4085
91
.
71.
Stupp
R
,
Hegi
ME
,
Gorlia
T
,
Erridge
SC
,
Perry
J
,
Hong
YK
, et al
Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071–22072 study): a multicentre, randomised, open-label, phase 3 trial
.
Lancet Oncol
2014
;
15
:
1100
8
.