Skip to main content
CNS Oncology logoLink to CNS Oncology
. 2017 Dec 15;7(1):51–65. doi: 10.2217/cns-2017-0026

Contemporary management of high-grade gliomas

Hao-Wen Sim 1,1,*,, Erin R Morgan 1,1,, Warren P Mason 1,1,**
PMCID: PMC6001673  PMID: 29241354

Abstract

High-grade gliomas, including glioblastoma, are the most common malignant brain tumors in adults. Despite intensive efforts to develop new therapies for these diseases, treatment options remain limited and prognosis is poor. Recently, there have been important advances in our understanding of the molecular basis of glioma, leading to refinements in our diagnostic and management approach. There is new evidence to guide the treatment of elderly patients. A multitude of new agents have been investigated, including targeted therapies, immunotherapeutics and tumor-treating fields. This review summarizes the key findings from this research, and presents a perspective on future opportunities to advance the field.

Keywords: : bevacizumab, drug development, elderly, high-grade glioma, immunotherapy, molecular biology, tumor-treating fields


Practice points.

  • Molecular markers now have an established role in the diagnosis and management of high-grade glioma, and have been incorporated into the updated WHO 5th Edition Classification of Tumors of the Central Nervous System.

  • There is new evidence to guide management of patients over 65 years of age with high-grade glioma. Most of these patients will benefit from short-course radiotherapy with concurrent and adjuvant temozolomide.

  • For unselected patients with high-grade glioma, the role of bevacizumab is limited.

  • Tumor-treating fields have shown efficacy in patients with newly diagnosed glioblastoma. However, it does not meet established benchmarks for cost–effectiveness and has not been widely adopted as a therapy for patients with this disease.

  • Immunotherapeutics are under evaluation for high-grade glioma. We await the forthcoming results from randomized Phase III trials.

  • Many targeted therapies have been evaluated for high-grade glioma, but so far have lacked clinical utility. Efforts incorporating novel strategies are ongoing.

High-grade gliomas, including glioblastoma (GBM), anaplastic astrocytoma (AA) and anaplastic oligodendroglioma (AO), originate from the supporting neuroglial cells of the CNS. GBM, the most common and most aggressive of the primary brain tumors, typically presents in late adulthood. AA and AO affect a younger age group and generally have a more protracted clinical course. High-grade gliomas can be debilitating, owing to physical disability, cognitive impairment, personality change, depression and seizure disorder, and require complex multidisciplinary care.

For patients with newly diagnosed GBM, maximal safe resection followed by radiotherapy with concomitant and adjuvant temozolomide chemotherapy is the current standard of care. Median overall survival for patients with GBM remains poor and was 14.6 months in the landmark EORTC 26981/22981-NCIC CTG CE3 Phase III trial [1,2]. This same approach is used for patients with newly diagnosed AA, based on preliminary findings of the ‘Concurrent and Adjuvant Temozolomide in Non-1p/19q Deleted Anaplastic Glioma’ (CATNON) trial [3]. For patients with newly diagnosed AO, chemoradiotherapy with procarbazine, lomustine and vincristine (PCV) has demonstrable efficacy in Phase III trials [4,5]. However, due to the increased toxicity and treatment burden of PCV versus monthly temozolomide, many clinicians substitute temozolomide in this setting. Accordingly, a comparison of PCV against temozolomide is underway in the ‘Concomitant and Adjuvant Temozolomide Versus PCV Chemotherapy in Patients With Anaplastic Glioma or Low Grade Glioma’ (CODEL) trial (NCT00887146).

Tumor recurrence occurs in almost all patients. Options include repeat surgical debulking, radiotherapy, lomustine (CCNU), bevacizumab, etoposide and procarbazine. However, there is no global standard and prognosis is limited. Patients are encouraged to enroll in clinical trials to access novel therapies.

Clearly, there exists an unmet need for innovative approaches. In this review, we highlight important changes to the evidence base for high-grade glioma. We begin with a discussion of major updates, namely molecular neuropathology, the management of elderly patients, bevacizumab and tumor-treating fields (TTF). This is followed by an overview of clinical trials of immunotherapeutics and targeted therapies for high-grade gliomas. Finally, we offer our perspective on research priorities and speculate on the future directions of the field.

Major updates

Molecular biology of high-grade gliomas

There have been substantial advances in our understanding of the molecular aberrations found in malignant gliomas. Key discoveries include the isocitrate dehydrogenase (IDH) mutation, codeletion of the short arm of chromosome 1 and long arm of chromosome 19 (1p19q) and O6-methylguanine-DNA methyltransferase (MGMT) gene promoter methylation. These have emerged as being important determinants of treatment response and survival. Consequently, they are now routinely tested and have become fundamental to glioma classification.

IDH catalyzes the oxidative decarboxylation of isocitrate to α-ketoglutarate, and subsequently to the oncometabolite 2-hydroxyglutarate [6]. In turn, 2-hydroxyglutarate acts via a family of dioxygenases to impair epigenetic regulation and increase hypoxia-inducible factor 1-α. The prevalent IDH1 R132H mutation is detectable with immunohistochemistry in over 90% of cases [7]. IDH mutations can also be identified by sequencing IDH1 codon 132 and IDH2 codon 172. These mutations are common in low-grade gliomas and secondary GBMs, and confer significantly improved prognosis [8].

The 1p19q codeletion is an unbalanced reciprocal translocation that is a characteristic of oligodendrogliomas. Multiple studies have demonstrated the favorable prognostic and predictive utility of the 1p/19q codeletion, although the biologic basis remains unclear. Specifically, in randomized Phase III trials evaluating chemoradiotherapy with PCV for AO, patients harboring the 1p19q codeletion derived greater benefit from PCV and lived substantially longer [4,5]. In contrast, partial 1p or 19q loss did not confer this significance.

MGMT gene promotor methylation causes epigenetic silencing of MGMT, which is necessary for DNA repair. Notably, based on review of randomized Phase III trials evaluating temozolomide in patients with GBM, those containing the MGMT gene promoter methylation obtained meaningful survival benefit from temozolomide, whereas those without the methylation did not [9]. Initially, MGMT status was assessed with immunohistochemistry and MGMT methylation-specific PCR; however, widespread clinical use was limited by numerous technical issues including poor reliability, reproducibility and the labor-intensive work [10,11]. Newer methods include bisulfite sequencing, pyrosequencing, high-resolution melt analysis and infinium methylation BeadChip, which have improved standardization and accuracy of MGMT testing [12,13].

In 2016, the WHO published the 5th Edition Classification of Tumors of the Central Nervous System [14]. This represents a seminal update, with the introduction of integrated diagnoses combining histology and molecular parameters for many entities. This incorporates the recently established prognostic and predictive information from IDH and 1p19q.

GBM is now subdivided into IDH wild-type (predominantly primary GBM, patients over 55 years of age, poor prognosis) and IDH-mutant entities (predominantly secondary GBM, younger patients, favorable prognosis). The diagnosis of AO requires IDH-mutant and 1p19q-codeleted status, whereas AA requires IDH-mutant and noncodeleted status. Importantly, both entities are IDH mutant; a glioma that is IDH wild-type with or without 1p19q codeletion instead represents a genomically unstable GBM. In addition, 1p19q codeletion is mutually exclusive with TP53 mutation and ATRX inactivation [15]. Accordingly, a glioma that is IDH-mutant, TP53-mutant and ATRX-inactivated is considered AA. Finally, the use of molecular parameters handles the problematic and indeterminate entity called anaplastic oligoastrocytoma, which was previously defined by a mixed histological pattern and was subject to poor interobserver agreement [16,17]. The combination of histology and molecular parameters effectively differentiates nearly all cases as either AO or AA. To facilitate clinical decision making, the current standard is to incorporate all the tissue-based information (histology, grade, molecular findings) into an integrated diagnosis, which is then reported to clinicians.

Molecular markers have significantly contributed to diagnostic precision in high-grade glioma, and yield important therapeutic implications. The next steps will be to improve understanding of clinical and molecular heterogeneity within glioma subtypes. Ongoing efforts include assessment of additional molecular markers, methylation profiling and a coordinated approach to histologic–molecular correlation as part of clinical trials.

Treatment of high-grade gliomas in the elderly

Although GBM is predominantly a disease of older adults, with a median age of diagnosis of 64 and an increasing number of patients diagnosed over the age of 70 [18], management of this disease in the elderly remains a particular challenge. Compared with younger patients, those over the age of 65 have shorter overall survival [19–21]. The EORTC 26981/22981-NCIC CE3 study excluded patients over 70 [1,2], and although a subgroup analysis showed a trend toward benefit in patients over 60 years old, the degree of benefit diminished with increasing age [19]. Elderly patients are also more susceptible to toxicities of treatment, including radiation-induced cognitive deficits [22] and chemotherapy side effects [23]. GBM in elderly patients also seems to be biologically more aggressive, with a very low incidence of favorable prognostic markers such as IDH mutations and the glioma cytosine–phosphate–guanine island methylator phenotype, although MGMT status does not seem to vary with age [24].

This has led to efforts to tailor treatment to this patient population through the use of modified radiation schedules and selective use of temozolomide chemotherapy. The French ANOCEF study demonstrated a median survival benefit of 12 weeks with radiotherapy versus supportive care alone in elderly patients [25]. Subsequently, Roa et al. [26] demonstrated similar survival in patients over age 60 who received short-course radiotherapy (40 Gy in 15 fractions) versus the conventional regimen (60 Gy in 30 fractions). Patients treated with short-course radiotherapy were less likely to require increased corticosteroid doses after completing treatment. The Nordic study [27] randomized patients over age 60 with newly diagnosed GBM to standard radiotherapy (60 Gy in 30 fractions), hypofractionated radiotherapy (34 Gy in 10 fractions) or temozolomide monotherapy. This trial demonstrated longer overall survival (OS) in patients over the age of 70 treated with hypofractionated versus standard radiotherapy. In addition, this study demonstrated improved survival in MGMT-methylated patients treated with temozolomide, whereas MGMT status did not influence response to radiotherapy. The NOA-08 study [28] demonstrated that temozolomide monotherapy was noninferior to radiotherapy alone in elderly patients with MGMT-methylated GBM, while patients with unmethylated tumors showed improved survival with radiotherapy alone. In addition, for the frail elderly with Karnofsky performance score <70, Pérez-Larraya et al. [29] showed that temozolomide monotherapy was a well-tolerated regimen, and for the subgroup with MGMT-methylated GBM, survival outcomes were superior to historical control.

The role of combined chemoradiotherapy in elderly patients remained undefined until the recent publication of a Phase III trial, which randomized 562 elderly patients with GBM to either short-course radiotherapy alone (40 Gy in 15 fractions) or radiotherapy with concurrent temozolomide [30]. This trial demonstrated improved OS in patients treated with combined therapy (median 9.3 vs 7.6 months). Of note, survival benefit was greater in patients with MGMT-methylated tumors, although benefit was still observed in the unmethylated patient group.

Based on these studies, we can now propose a basic evidence-based approach for the first-line treatment of patients with GBM 65 years or older. For a minority of very fit, ‘physiologically young’ patients over 65, it remains reasonable to propose standard chemoradiotherapy, with a discussion of the possibility for increased toxicity in this age group. The majority of patients with adequate performance status and without comorbidities precluding combined therapy should receive short-course radiotherapy (40 Gy in 15 fractions) with concurrent and adjuvant temozolomide. Patients with poor performance status and the frail elderly may be offered best supportive care, or treatment based on MGMT status: temozolomide monotherapy for patients with MGMT-methylated GBM or short-course radiotherapy alone for unmethylated GBM.

Role of bevacizumab

GBM is a highly vascular neoplasm, with abnormal vasculature characterized by tortuous blood vessels, vascular permeability and resulting hypoxia leading to the histological finding of pseudopalisading necrosis [31]. Tumor growth and invasion are intrinsically linked to hypoxia, which results in upregulation of hypoxia-inducible factor 1-α, and downstream upregulation of VEGF, which is associated with glioma cell stemness, mesenchymal phenotype and an immunosuppressive cellular milieu [32]. Thus, there is a strong biologic rationale for the use of antiangiogenic agents in GBM, and these drugs have thus been extensively studied as therapeutic targets in both newly diagnosed and recurrent GBM.

Bevacizumab, a humanized monoclonal antibody which binds VEGF-A, is the most extensively studied of the antiangiogenic agents for GBM. Bevacizumab was approved by the US FDA for use in recurrent GBM in 2009 [33]. The ‘Bevacizumab Alone and in Combination with Irinotecan in Recurrent GBM’ (BRAIN) study [34] was a randomized Phase II trial that assigned 167 patients with recurrent GBM to receive bevacizumab 10 mg/kg with or without irinotecan. This trial demonstrated objective response rates of 38 and 28% in patients treated with bevacizumab with and without irinotecan, respectively. Progression-free survival at 6 months (PFS-6) was 42% in patients treated with bevacizumab alone and 50% in the combination arm. In a single-arm study, 48 patients with recurrent GBM were treated with bevacizumab 10 mg/kg with irinotecan added upon disease progression, demonstrating an objective response rate of 35% and PFS-6 of 29% [35]. While these findings led to FDA approval for recurrent GBM in the USA, its use has not been approved in Europe due to concerns regarding the lack of a bevacizumab-free control arm, the modest improvement in OS and difficulties with interpreting MRI-based disease progression in patients treated with bevacizumab [36].

In the USA, widespread use of bevacizumab for recurrent GBM has limited the opportunity for further evaluation in this setting. In Europe, the randomized Phase II ‘Single-Agent Bevacizumab or Lomustine Versus a Combination of Bevacizumab Plus Lomustine in Patients with Recurrent GBM’ (BELOB) trial [37] showed promising results for the combination of bevacizumab and lomustine versus either agent alone. Unfortunately, these findings were not borne out in the subsequent Phase III trial which compared the combination of lomustine and bevacizumab with lomustine alone [38]. This trial showed no difference in OS, although there was a significant increase in PFS from 1.5 to 4.2 months in the combination arm. Several other Phase II trials have evaluated the combination of bevacizumab with a variety of other cytotoxic and targeted agents, including temozolomide, temsirolimus and erlotinib, but none have shown significant activity [31].

Similarly, bevacizumab has been tested in the setting of newly diagnosed GBM, with a series of Phase II trials using bevacizumab in combination with radiotherapy and temozolomide [39–41]. As seen in the recurrent setting, PFS was prolonged in comparison to historical controls (13–14 months), while the effect on OS was modest (10–21 months). Subsequently, two randomized Phase III trials were conducted, ‘A Study of Avastin in Combination With Temozolomide and Radiotherapy in Patients With Newly Diagnosed GBM’ (AVAGlio) [42] and RTOG-0825 [43]. These studies showed longer PFS in patients treated with bevacizumab, but failed to show OS benefit. Thus, despite encouraging preclinical results with in vivo activity and reduction of vasogenic edema, there is abundant high-quality evidence that bevacizumab is not indicated in unselected patients with newly diagnosed GBM.

Role of alternating electric fields

The locoregional use of alternating electric fields to treat tumors, called TTF, represents a novel treatment modality [44]. Preclinical data showed that low-intensity (1–3 V/cm), intermediate-frequency (200 kHz) fields selectively disrupted microtubule assembly, thereby interfering with mitotic spindle formation and segregation of the two daughter cells during mitosis. This caused preferential apoptosis of rapidly dividing malignant cells. In contrast, lower frequencies caused undesirable depolarization of neurons and myocytes, and higher frequencies caused excess heat generation.

TTF is administered via transducer arrays, which are applied to the patient's shaved scalp for 18 h per day. The arrays are oriented to target the tumor volume based on contemporaneous imaging. The device is powered by a portable generator carried by the patient.

Two pivotal randomized Phase III trials have examined the efficacy of TTF in patients with GBM. The EF-11 trial [45] examined 237 patients with progressive GBM treated with either TTF or physician's choice of chemotherapy and failed to demonstrate improvement in PFS (median 2.2 vs 2.1 months; HR: 0.81; 95% CI: 0.60–1.09; p = 0.16) or OS (median 6.6 vs 6.0 months; HR: 0.86; 95% CI: 0.66–1.12; p = 0.27).

The second study, the EF-14 trial [46], investigated 695 patients with newly diagnosed GBM who had already completed temozolomide chemoradiotherapy. The patients were recruited from 83 institutions internationally and their characteristics were comparable to contemporary Phase III trials of newly diagnosed GBM. They received either TTF and temozolomide, or temozolomide alone for 6–12 cycles. The primary endpoint was PFS, which improved from a median of 4.0–7.1 months (HR: 0.62; 98.7% CI: 0.43–0.89; p = 0.001). In addition, OS in the intention-to-treat population improved from a median of 16.6–19.6 months (HR: 0.74; 95% CI 0.56–0.98; p = 0.03). This survival improvement was confirmed in preliminary reporting of the updated EF-14 results [47].

The predominant toxicities from TTF were local skin irritation and headache. Skin irritation was grade 1 or 2 in 43% of patients, and grade 3 in 2% of patients. Grade 1 or 2 headache was more frequently reported (TTF 21% vs control 14%). No additional systemic toxicity was identified with TTF. It was reported that quality of life was not adversely affected, measured by EORTC QLQ C-30 and BN20 questionnaires, performance status and mini-mental examination scores [48]. However, in our opinion, these measures may not fully encompass the decisions involved in patient decision making, such as the perceived stigmatization and inconvenience of wearing the TTF device. Compliance was only 75% in the EF-14 trial, which involved highly motivated patients.

There are other considerations when interpreting the EF-14 trial. First, patients and investigators were not blinded. Patients randomized to TTF received additional training and support necessary for use of the TTF device. Together, these may have conferred a placebo benefit. Another qualification was that randomization was performed after successful completion of chemoradiotherapy, such that the benefit of TTF may not be applicable to patients with primary refractory disease.

Of note, the financial cost of TTF relative to its efficacy poses a major impediment to implementation. A cost–effectiveness analysis of TTF using a decision-analysis Markov model was reported based on the EF-14 results and unit cost of €21,000 per month [49]. The incremental cost–effectiveness ratio was €549,909 per life-year gained (95% CI: 447,017–745,805). In Canada and many other countries, an intervention with incremental cost–effectiveness ratio >CAD$100,000 per life-year gained is typically viewed as poor resource utilization, although thresholds are frequently debated.

In summary, for newly diagnosed GBM, the EF-14 trial is supportive of TTF as being an efficacious treatment modality. However, the absolute benefit is modest and it does not meet established benchmarks for cost–effectiveness. For progressive GBM, there is insufficient evidence of benefit. The widespread adoption of TTF will therefore be contingent on identifying subgroups of patients with greatest benefit, and reduction in the market price of TTF to a commensurate level. Further studies are evaluating the utility of TTF for GBM, such as in combination with bevacizumab or stereotactic radiotherapy (NCT02663271, NCT02743078, NCT01894061, NCT01925573, NCT02343549).

Novel therapies

Immunotherapy in high-grade gliomas

The success of immune-based therapies in cancers such as melanoma and non-small-cell lung cancer (NSCLC), as well as other solid and hematologic malignancies, has led to a revolution in systemic cancer therapy. Clinical trials using these agents, which include immune checkpoint inhibitors, antitumor vaccines and autologous cell-based therapies, are ongoing for a wide variety of tumor types, including GBM.

While there is preclinical evidence to support activity of these agents in primary CNS malignancies, their use is complicated by both the unique immunologic milieu of the CNS and by the wide array of mechanisms used by high-grade gliomas to promote peritumoral immunosuppression and avoid immunologic surveillance. Within the CNS, access by activated T cells is limited by the glia limitans, a component of the blood–brain barrier [50], as well as the expression of Fas ligand, leading to apoptosis of Fas-expressing cytotoxic T-lymphocytes [51]. The tumor microenvironment of GBM is characterized by expression of immunosuppressive cytokines, such as TGF-β, IL-10, signal transducer and activator of transcription 3 and programmed-death ligand 1 [50–52]. In addition, systemic immune suppression is inherent in many patients with GBM, as evidenced by significant lymphopenia, and is likely exacerbated by lymphotoxic treatments such as radiation, temozolomide and corticosteroids [50,53]. For instance, treatment-induced lymphopenia has been associated with shorter OS in patients treated with standard chemoradiotherapy, and may also influence response to immune-based therapies [53]. It remains to be seen to what degree these factors will influence the efficacy of immunotherapy in primary CNS malignancies.

Antitumor vaccine therapy is one form of immunotherapy that has been extensively investigated in GBM. Broadly, these vaccines can be characterized as peptide- and cell-based vaccines, although there are many variations of each.

In peptide-based vaccination, patients are directly inoculated with one or more tumor-associated antigens in order to elicit an immune response against tumor cells. The peptide epitopes are selected based on immunogenicity, tumor specificity and homogeneity of expression. These proteins may be linked with carrier proteins to enhance immunogenicity, such as keyhole limpet hemocyanin, and are frequently administered with immune-stimulating adjuvants including granulocytic-macrophage colony-stimulating factor.

The most extensively evaluated vaccine-based therapy for glioma to date is rindopepimut, which consists of the amino acid sequence of EGFR variant III (EGFRvIII) conjugated to keyhole limpet hemocyanin. This primes dendritic cells against EGFRvIII, which then generates a specific immune response against GBM cells harboring the EGFRvIII mutation. Notably, this represents a combination of targeted and immunotherapeutic strategies. For patients with newly diagnosed GBM harboring the EGFRvIII mutation, trials evaluated the addition of rindopepimut to standard temozolomide chemoradiotherapy. In the Phase II ACT-III trial, median PFS and OS were 9.2 and 21.8 months, respectively, which were significantly longer than matched controls [54]. However, there was no survival benefit in the randomized Phase III ACT-IV trial when preliminary results were recently presented [55]. For patients with recurrent GBM harboring the EGFRvIII mutation, preliminary findings from the randomized Phase II ReACT trial showed significant OS improvement from 8.8 to 12.0 months with rindopepimut plus bevacizumab versus bevacizumab alone [56]. Of note, upon progression after treatment with rindopepimut, EGFRvIII expression was lost, although it is unknown whether this represents the natural evolution of disease or the results of immunologic selection.

Other vaccines targeting EGFRvIII are in early clinical testing, including ADU-623, which uses a live attenuated listeria vaccine targeting EGFRvIII in recurrent high-grade glioma (NCT01967758). Another target for peptide vaccines in glioma is IDH1, given that the IDH1 R132H mutation is expressed in the majority of secondary GBM and is thought to represent a driver mutation in glioma development. Phase I studies are underway (NCT02454634, NCT02193347, NCT02771301).

In cell-based vaccination, autologous antigen-presenting cells (APCs) such as dendritic cells are harvested from the patient and primed with one or more tumor-associated antigens. Antigen loading may be performed with one or more peptide epitopes, or harvested APCs may be exposed to a lysate of the patient's own tumor cells in order to generate a personalized vaccine. The primed APCs are then reintroduced to the patient, resulting in antigen-specific T-cell activation. Other targets for cell-based vaccine therapy include tumor-derived mRNA, glioma stem cells and virus-derived antigens. While cell-based vaccines offer a unique opportunity for highly personalized therapy, and potentially limited toxicity due to the specificity of the targeted antigens, these agents are expensive and labor-intensive to produce. The process entails harvesting APCs through apheresis, culturing and differentiating the cells in vitro, priming through exposure to the desired antigen and preparation of the personalized vaccine. This presents challenges in terms of potentially unacceptable delays in treatment as well as cost–effectiveness concerns. To date, there are no reported Phase III trials of cell-based therapies, so clinical utility of these agents remains to be determined.

Immune checkpoint inhibition is another promising form of immunotherapy. There is preclinical evidence of their activity in murine models of glioma, as well as clinical responses in brain metastases of melanoma and NSCLC. Multiple clinical trials are underway using these agents both in newly diagnosed and recurrent GBM, and as monotherapy, dual checkpoint inhibition and combinations with other agents. Preliminary results of CheckMate-143, a randomized Phase III trial using nivolumab in combination with bevacizumab in patients with recurrent GBM, were recently reported and showed no benefit of the nivolumab with bevacizumab combination over bevacizumab alone [57]. So far, responses in early phase studies suggest that the activity of these agents in GBM may be modest at best, in comparison to the dramatic responses observed in melanoma and NSCLC [58]. This may be related in part to the relatively low mutational burden of malignant glioma and the underlying immunosuppressive cellular milieu. Nonetheless, we await the forthcoming trial results to inform our decision making.

Targeted therapies

Targeted therapies represent a paradigm shift toward rational drug design. They selectively target signaling pathways, which are believed to be important for glioma growth and survival, based on our understanding of the fundamental processes driving malignancy [59]. The targeted therapy trials, which will be discussed, are summarized in Table 1.

Table 1. . Targeted therapies in high-grade gliomas.

Study Year Setting N Design Findings Ref.
EGF inhibition

Rich et al. 2004 Recurrent glioblastoma 57 Phase II trial
Gefitinib 500–1000-mg daily
ORR: 0%
PFS-6: 13%
[60]

Neyns et al. 2009 Recurrent glioblastoma or anaplastic glioma 55 Phase II trial
Cetuximab 250 mg/m2 weekly
ORR: 6%
PFS-6: 9%
[61]

van den Bent et al. 2009 Recurrent glioblastoma 110 Phase II trial
1. Erlotinib 150–200-mg daily
2. Temozolomide 150–200 mg/m2 D1–5 q4w or carmustine 60–80 mg/m2 D1–3 q8w
ORR: 4 vs 10%
PFS-6: 11 vs 24%
[62]

Li et al. 2010 Newly diagnosed glioblastoma 192 Phase II trial
Temozolomide chemoradiotherapy + mAb 425
mOS: 15.7 m [63]

Peereboom et al. 2013 Recurrent glioblastoma 56 Phase II trial
Erlotinib 150 mg daily + sorafenib 400 mg twice-daily
ORR: 5%
PFS-6: 14%
[64]

Reardon et al. 2015 Recurrent glioblastoma 119 Phase II trial
1. Afatinib 40 mg daily + temozolomide 75 mg/m2 D1–21 q4w
2. Afatinib 40 mg daily
3. Temozolomide 75 mg/m2 D1–21 q4w
ORR: 14 vs 14 vs 21%
PFS-6: 10 vs 3 vs 23%
[65]

Westphal et al. 2015 Newly diagnosed glioblastoma 149 Phase III trial
1. Temozolomide chemoradiotherapy + nimotuzumab
2. Temozolomide chemoradiotherapy
mPFS: 7.7 vs 5.8 m
mOS: 22.3 vs 19.6 m
[66]

EGF antibody drug conjugate

Gan et al. (abstract) 2014 Recurrent glioblastoma 12 Phase I trial
ABT-414
ORR: 33% [67]

Reardon et al. 2016 Newly diagnosed glioblastoma 45 Phase I trial
Temozolomide chemoradiotherapy + ABT-414
mPFS: 6.1 m
mOS: not reached
[68]

EGF peptide vaccine

Schuster et al. 2015 Newly diagnosed glioblastoma expressing EGFRvIII 65 Phase II trial
Temozolomide chemoradiotherapy + rindopepimut
mPFS: 9.2 m
mOS: 21.8 m
[54]

Reardon et al. (abstract) 2015 Recurrent glioblastoma expressing EGFRvIII 72 Phase II trial
1. Rindopepimut + bevacizumab
2. Bevacizumab
ORR: 24 vs 17%
PFS-6: 27 vs 11%
mOS: 12.0 vs 8.8 m
[56]

Weller et al. (abstract) 2016 Newly diagnosed glioblastoma expressing EGFRvIII 745 Phase III trial
1. Temozolomide chemoradiotherapy + rindopepimut
2. Temozolomide chemoradiotherapy
Minimal residual disease
mOS: 20.1 vs 20.0 m
Nonminimal residual disease
mOS: 14.8 vs 14.1 m
[55]

VEGF inhibition

Batchelor et al. 2010 Recurrent glioblastoma 31 Phase II trial
Cediranib 45-mg daily
ORR: 27%
PFS-6: 26%
[69]

Iwamoto et al. 2010 Recurrent glioblastoma 35 Phase II trial
Pazopanib 800-mg daily
ORR: 6%
PFS-6: 3%
[70]

de Groot et al. 2011 Recurrent glioblastoma or anaplastic glioma 58 Phase II trial
Aflibercept 4 mg/kg q2w
Glioblastoma
ORR: 18%
PFS-6: 8%
Anaplastic glioma
ORR: 44%
PFS-6: 25%
[71]

Pan et al. 2012 Recurrent glioblastoma or anaplastic glioma 30 Phase II trial
Sunitinib 50-mg daily D1–28 q6w
Glioblastoma
ORR: 0%
PFS-6: 17%
Anaplastic glioma
ORR: 0%
PFS-6: 22%
[72]

Kreisl et al. 2012 Recurrent glioblastoma or anaplastic glioma 64 Phase II trial
Vandetanib 300-mg daily
Glioblastoma
ORR: 13%
PFS-6: 7%
Anaplastic glioma
ORR: 7%
PFS-6: 7%
[73]

Muhic et al. 2013 Recurrent glioblastoma 25 Phase II trial
Nintedanib 200 mg twice-daily
ORR: 0%
PFS-6: 4%
[74]

Reardon et al. 2013 Recurrent glioblastoma 41 Phase II trial
Pazopanib 400-mg daily + lapatinib 1000-mg daily
PTEN/EGFRvIII positive
PFS-6: 0%
PTEN/EGFRvIII negative
PFS-6: 15%
[75]

Kreisl et al. 2013 Recurrent glioblastoma 63 Phase II trial
Sunitinib 37.5-mg daily
Bevacizumab naive
PFS-6: 10%
Bevacizumab resistant
PFS-6: 0%
[76]

Batchelor et al. 2013 Recurrent glioblastoma 325 Phase III trial
1. Cediranib 20-mg daily + lomustine 110 mg/m2 q6w
2. Cediranib 30-mg daily
3. Placebo + lomustine 110 mg/m2 q6w
ORR: 17 vs 15 vs 9%
PFS-6: 35 vs 25 vs 16
mPFS: 4.1 vs 3.0 vs 2.7 m
mOS: 9.4 vs 8.0 vs 9.8 m
[77]

PDGF inhibition

Lassman et al. 2011 Recurrent glioblastoma with overexpression of dasatinib molecular targets 50 Phase II trial
Dasatinib 100 mg twice-daily
ORR: 0%
PFS-6: 6%
[78]

mTOR inhibition

Chang et al. 2005 Recurrent glioblastoma 43 Phase II trial
Temsirolimus 250-mg intravenously weekly
ORR: 5%
PFS-6: 2%
[79]

Galanis et al. 2005 Recurrent glioblastoma 65 Phase II trial
Temsirolimus 250-mg intravenously weekly
ORR: 36%
PFS-6: 8%
[80]

Kreisl et al. 2009 Recurrent glioblastoma 22 Phase II trial
Everolimus 70-mg weekly + gefitinib 250-mg daily
ORR: 14%
PFS-6: 0%
[81]

PI3K inhibition

Pitz et al. 2015 Recurrent glioblastoma 33 Phase II trial
Sonolisib 8-mg daily
ORR: 3%
PFS-6: 17%
[82]

PKC inhibition

Kreisl et al. 2010 Recurrent glioblastoma or anaplastic glioma 118 Phase II trial
Enzastaurin 500–525-mg daily
Glioblastoma
ORR: 30%
PFS-6: 7%
Anaplastic glioma
ORR: 15%
PFS-6: 16%
[83]

Wick et al. 2010 Recurrent glioblastoma 266 Phase III trial
1. Enzastaurin 500-mg daily q6w
2. Lomustine 100–130 mg/m2 q6w
ORR: 3 vs 4%
PFS-6: 11 vs 19%
mPFS: 1.5 vs 1.6 m
mOS: 6.6 vs 7.1 m
[84]

c-Met inhibition

Wen et al. (abstract) 2010 Recurrent glioblastoma 124 Phase II trial
Cabozantinib 125–175-mg daily
PFS-6: 21% [85]

Wen et al. 2011 Recurrent glioblastoma 61 Phase II trial
Rilotumumab 10–20 mg/kg q2w
ORR: 0% [86]

Integrin inhibition

Reardon et al. 2008 Recurrent glioblastoma 81 Phase II trial
1. Cilengitide 500 mg twice-weekly
2. Cilengitide 2000 mg twice-weekly
ORR: 5 vs 13%
PFS-6: 10 vs 15%
[87]

Gilbert et al. 2012 Recurrent glioblastoma undergoing surgery 30 Phase II trial
Cilengitide 2000 mg twice-weekly
PFS-6: 12% [88]

Stupp et al. 2014 Newly diagnosed glioblastoma with MGMT promoter methylation 545 Randomized Phase III trial
1. Temozolomide chemoradiotherapy + cilengitide 2000 mg twice-weekly
2. Temozolomide chemoradiotherapy
mPFS: 13.5 vs 10.7 m
mOS: 26.3 vs 26.3 m
[89]

Microtubule inhibition

Stupp et al. 2011 Recurrent glioblastoma 38 Phase II trial
Sagopilone 16 mg/m2 q3w
ORR: 0%
PFS-6: 7%
[90]

Chamberlain et al. 2014 Recurrent glioblastoma 56 Phase II trial
Verubulin 3.3 mg/m2 D1, 8, 15 q4w
Bevacizumab naive
PFS-6: 14%
Bevacizumab resistant
PFS-6: 8%
[91]

Histone deacetylase inhibition

Galanis et al. 2009 Recurrent glioblastoma 66 Phase II trial
Vorinostat 200 mg twice-daily D1–14 q3w
ORR: 3%
PFS-6: 15%
[92]

Friday et al. 2012 Recurrent glioblastoma 37 Phase II trial
Vorinostat 400-mg daily D1–14 q3w + bortezomib 1.3 mg/m2 D1, 4, 8, 11 q3w
ORR: 3%
PFS-6: 0%
[93]

Lee et al. 2015 Recurrent glioblastoma or anaplastic glioma 39 Phase II trial
Panobinostat 30 mg D1, 3, 5 q2w + bevacizumab 10 mg/kg q2w
Glioblastoma
ORR: 29%
PFS-6: 30%
Anaplastic glioma
ORR: 27%
PFS-6: 47%
[94]

TGF-β inhibition

Brandes et al. 2016 Recurrent glioblastoma 158 Phase II trial
1. Galunisertib 150 mg twice-daily D1–14 q4w + lomustine 100–130 mg/m2 q6w
2. Galunisertib 150 mg twice-daily D1–14 q4w
3. Placebo + lomustine 100–130 mg/m2 q6w
ORR: 1 vs 5 vs 0%
PFS-6: 6 vs 15 vs 6%
mPFS: 2 vs 2 vs 2 m
mOS: 6.7 vs 8.0 vs 7.5 m
[95]

XPO inhibition

Mau-Sørensen (abstract) 2016 Recurrent glioblastoma 35 Phase II trial
1. Selinexor 50 mg/m2 twice-daily ×3, then surgery
2. Selinexor 50 mg/m2 twice-daily
3. Selinexor 60 mg twice-daily
4. Selinexor 80-mg daily
Pooled results
ORR: 11%
PFS-6: 15%
[96]

D#: Day #; EGFRvIII: EGFR variant III; m: Month; mAb: Monoclonal antibody; MGMT: O6-methylguanine-DNA methyltransferase; mOS: Median overall survival; mPFS: Median progression-free survival; N: Number; ORR: Objective response rate; PFS-6: Progression-free survival rate at 6 months; PTEN: Phosphate and tensin homolog; q8w: Every # weeks; XPO: Exportin-1.

The EGF receptor is overexpressed in many GBMs [97]. Early studies suggested that GBMs harboring a specific EGF receptor mutation (EGFRvIII) and intact phosphate and tensin homolog suppressor (PTEN) gene might respond favorably to EGF inhibition [98]. Numerous EGF receptor inhibitors were therefore evaluated in GBM trials, including the small molecule inhibitors gefitinib [60], erlotinib [62,64] and afatinib [65], and the monoclonal antibodies cetuximab [61], mAb 425 [63] and nimotuzumab [66]. Unfortunately, despite being a proven strategy in NSCLC and other malignancies, results have consistently showed limited activity in GBM, even among those with EGF receptor amplification.

A novel approach to EGF inhibition is the use of antibody–drug conjugates such as ABT-414. The antibody component of ABT-414 effectively binds to mutant EGF receptors despite the blood–brain barrier, then delivers the cytotoxic drug component directly to the tumor cell. This obviates the pharmacokinetic challenges with conventional EGF receptor inhibition. There have been encouraging results from preliminary studies of ABT-414 [67,68], and Phase III trials are now underway (NCT02573324, NCT02343406).

Another approach is the use of EGF peptide vaccines such as rindopepimut, and studies evaluating this vaccine in patients with GBM have been presented elsewhere in this review.

Since GBM is a highly vascular tumor, another group of targeted therapies have been developed against tumor angiogenesis. Important mediators of angiogenesis include VEGF, PDGF, mTOR, PI3K, PKC and hepatocyte growth factor (c-Met). Bevacizumab, a monoclonal antibody against VEGF, was discussed previously. Unfortunately, the vast armamentarium of other antiangiogenic agents targeting VEGF, PDGF, mTOR, PI3K, PKC and c-Met have uniformly failed to show an efficacy signal, and no reliable biomarkers have been identified to improve treatment selection [69–86].

Regarding novel drug targets, cilengitide is an integrin inhibitor, which mediates transmembrane interactions between cells and the surrounding stroma. Despite early suggestion of cilengitide activity in GBM with MGMT promoter methylation [87,88], there was no survival benefit in the larger randomized Phase III ‘Cilengitide Combined With Standard Treatment for Patients With Newly Diagnosed GBM With Methylated MGMT Promoter’ (CENTRIC) trial, so development was discontinued [89].

Sagopilone and verubulin target the microtubules, which are needed for cell division [90,91]. Vorinostat and panobinostat inhibit histone deacetylase, with the aim of reversing epigenetic changes associated with malignancy [92–94]. Galunisertib inhibits TGF-β receptor-1, which has been associated with several growth signaling pathways [95]. Selinexor is a selective inhibitor of exportin-1 (XPO), which causes accumulation of tumor suppressor proteins within the nucleus [96]. Discouragingly, all of these drugs lacked relevant activity when tested in patients with recurrent GBM.

Overall, targeted therapies have yet to demonstrate efficacy beyond standard cytotoxic chemotherapy. A consideration is whether adequate intratumoral drug concentrations are reached due to the blood–brain barrier, given presence of efflux transporters on the endothelial cells and limited passage of large-molecular-weight antibodies. The presence of intratumoral heterogeneity is another complicating factor, since only malignant cells bearing the relevant target will be affected. In addition, there are likely varying degrees of pathway redundancy, so that blockade of one pathway alone may be insufficient for cytotoxicity.

To further the development of effective targeted therapies, it is essential that the pharmacokinetic and pharmacodynamic properties of prospective drugs are fully characterized in GBM patients at an early stage. An example would be use of window-of-opportunity studies, whereby test doses of drug are administered preoperatively, then drug levels and proof of drug action can be evaluated in the resection specimen. It may be necessary for certain drugs to be administered via alternate routes such as direct intratumoral injection. Novel approaches will be required to circumvent intratumoral heterogeneity. Possibilities include drug conjugates or combinations with radiopharmaceuticals and immunotherapeutics, whereby we leverage bystander effect and the immune response to deal with adjacent tumor cells not bearing the relevant target. Finally, there is critical need for predictive biomarkers to select subgroups of patients to benefit from particular treatments. We anticipate the increased adoption of molecular sequencing of tumors, novel imaging such as magnetic resonance spectroscopy, and the development of radiomics-based imaging analysis will provide new opportunities for biomarker discovery.

Conclusion & future perspective

Despite exhaustive efforts to investigate novel therapies, treatment options for high-grade glioma have not changed significantly in recent times. A limiting factor is that trials are often small and underpowered, or require lengthy periods of patient accrual and follow-up before meaningful results become available. Moving forward, it is essential that we take maximum advantage of our finite patient pool. First, we must renew our support of international, multicenter, collaborative trial efforts which offer the greatest opportunity for robust and generalizable information to inform clinical decision making. Second, we need to embrace innovative trial methodologies that will allow us to more rapidly and efficiently test novel therapies. An exemplar is the ‘GBM Adaptive Global Innovative Learning Environment’ (GBM AGILE), which will use an adaptive trial design founded on Bayesian statistics. Patients will be preferentially recruited to treatment arms showing signs of efficacy, whereas ineffective treatment arms will be rapidly discontinued and replaced by newer options. Biomarker testing will be integrated into the selection process. In so doing, we optimize our expenditure of time and resources to support novel therapies with the greatest potential.

To achieve the promise of highly personalized medicine, we need to build on our advances in molecular neuropathology. A commitment to biobank collection of surgical specimens will afford an essential and rich resource for this. We envisage that concerted efforts in genomic sequencing, proteomic analysis and methylation profiling of high-grade gliomas, coupled with advances in bioinformatics and machine learning to interpret this data, will provide the next important discoveries. Novel research should be encouraged, such as investigations into miRNAs, the tumor microenvironment, and the glymphatic and immune response. Beyond pathology, we anticipate integration of novel imaging biomarkers into the repertoire, driven by development of techniques such as magnetic resonance spectroscopy and radiomics-based imaging analysis.

Finally, we need to adapt new methods to counter the blood–brain barrier, tumor heterogeneity and treatment resistance. Innovative combinations of treatment modalities will present such opportunities. Neurosurgery provides valuable access to the peritumoral space. Targeted drug conjugates can selectively deliver cytotoxic chemotherapy, immunotherapeutics and radiopharmaceuticals directly to tumor cells. A vast suite of different immunotherapy approaches remain to be investigated. Sequencing of radiotherapy with immunotherapeutics can produce an abscopal effect, a concept which is being trialed in several solid tumor sites. All in all, treatments are limited only by our creative potential.

The future promises to be an era characterized by robust and efficient trial design, a resolute commitment to deciphering the key molecular neuropathology and an engagement with innovative combination therapy. Together, they offer a realistic prospect of ameliorating this devastating disease.

Footnotes

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

Open access

This work is licensed under the Creative Commons Attribution 4.0 License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

References

Papers of special note have been highlighted as: • of interest; •• of considerable interest

  • 1.Stupp R, Mason WP, Van Den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 2005;352(10):987–996. doi: 10.1056/NEJMoa043330. [DOI] [PubMed] [Google Scholar]
  • 2.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;10(5):459–466. doi: 10.1016/S1470-2045(09)70025-7. [DOI] [PubMed] [Google Scholar]
  • 3.Van Den Bent MJ, Erridge S, Vogelbaum MA, et al. Results of the interim analysis of the EORTC randomized Phase III CATNON trial on concurrent and adjuvant temozolomide in anaplastic glioma without 1p/19q co-deletion: an intergroup trial. J. Clin. Oncol. 2016;34(18_Suppl.) LBA2000–LBA2000. [Google Scholar]
  • 4.Cairncross G, Wang M, Shaw E, et al. Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402. J. Clin. Oncol. 2013;31(3):337–343. doi: 10.1200/JCO.2012.43.2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Van Den Bent MJ, Brandes AA, Taphoorn MJ, 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(3):344–350. doi: 10.1200/JCO.2012.43.2229. [DOI] [PubMed] [Google Scholar]
  • 6.Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462(7274):739–744. doi: 10.1038/nature08617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Preusser M, Wohrer A, Stary S, Hoftberger R, Streubel B, Hainfellner JA. Value and limitations of immunohistochemistry and gene sequencing for detection of the IDH1-R132H mutation in diffuse glioma biopsy specimens. J. Neuropathol. Exp. Neurol. 2011;70(8):715–723. doi: 10.1097/NEN.0b013e31822713f0. [DOI] [PubMed] [Google Scholar]
  • 8.Parsons DW, Jones S, Zhang X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science (New York, NY) 2008;321(5897):1807–1812. doi: 10.1126/science.1164382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N. Engl. J. Med. 2005;352(10):997–1003. doi: 10.1056/NEJMoa043331. [DOI] [PubMed] [Google Scholar]
  • 10.Preusser M, Elezi L, Hainfellner JA. Reliability and reproducibility of PCR-based testing of O6-methylguanine-DNA methyltransferase gene (MGMT) promoter methylation status in formalin-fixed and paraffin-embedded neurosurgical biopsy specimens. Clin. Neuropathol. 2008;27(6):388–390. doi: 10.5414/npp27388. [DOI] [PubMed] [Google Scholar]
  • 11.Lalezari S, Chou AP, Tran A, et al. Combined analysis of O6-methylguanine-DNA methyltransferase protein expression and promoter methylation provides optimized prognostication of glioblastoma outcome. Neuro Oncol. 2013;15(3):370–381. doi: 10.1093/neuonc/nos308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Switzeny OJ, Christmann M, Renovanz M, Giese A, Sommer C, Kaina B. MGMT promoter methylation determined by HRM in comparison to MSP and pyrosequencing for predicting high-grade glioma response. Clin. Epigenetics. 2016;8:49. doi: 10.1186/s13148-016-0204-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bady P, Sciuscio D, Diserens AC, et al. MGMT methylation analysis of glioblastoma on the Infinium methylation BeadChip identifies two distinct CpG regions associated with gene silencing and outcome, yielding a prediction model for comparisons across datasets, tumor grades, and CIMP-status. Acta Neuropathologica. 2012;124(4):547–560. doi: 10.1007/s00401-012-1016-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathologica. 2016;131(6):803–820. doi: 10.1007/s00401-016-1545-1. [DOI] [PubMed] [Google Scholar]; •• A fundamental summary of the principles and key changes adopted by the 5th Edition WHO Classification of Tumors of the Central Nervous System.
  • 15.Sahm F, Reuss D, Koelsche C, et al. Farewell to oligoastrocytoma: in situ molecular genetics favor classification as either oligodendroglioma or astrocytoma. Acta Neuropathol. 2014;128(4):551–559. doi: 10.1007/s00401-014-1326-7. [DOI] [PubMed] [Google Scholar]
  • 16.Coons SW, Johnson PC, Scheithauer BW, Yates AJ, Pearl DK. Improving diagnostic accuracy and interobserver concordance in the classification and grading of primary gliomas. Cancer. 1997;79(7):1381–1393. doi: 10.1002/(sici)1097-0142(19970401)79:7<1381::aid-cncr16>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
  • 17.Kros JM, Gorlia T, Kouwenhoven MC, et al. Panel review of anaplastic oligodendroglioma from European Organization For Research and Treatment of Cancer Trial 26951: assessment of consensus in diagnosis, influence of 1p/19q loss, and correlations with outcome. J. Neuropathol. Exp. Neurol. 2007;66(6):545–551. doi: 10.1097/01.jnen.0000263869.84188.72. [DOI] [PubMed] [Google Scholar]
  • 18.Ostrom QT, Gittleman H, Liao P, et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007–2011. Neuro Oncol. 2014;16(Suppl. 4):iv1–iv63. doi: 10.1093/neuonc/nou223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Laperriere N, Weller M, Stupp R, et al. Optimal management of elderly patients with glioblastoma. Cancer Treat. Rev. 2013;39(4):350–357. doi: 10.1016/j.ctrv.2012.05.008. [DOI] [PubMed] [Google Scholar]
  • 20.Morgan ER, Norman A, Laing K, Seal MD. Treatment and outcomes for glioblastoma in elderly compared with non-elderly patients: a population-based study. Curr. Oncol. (Toronto, Ont.) 2017;24(2):e92–e98. doi: 10.3747/co.24.3424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Paszat L, Laperriere N, Groome P, Schulze K, Mackillop W, Holowaty E. A population-based study of glioblastoma multiforme. Int. J. Radiat. Oncol. Biol. Phys. 2001;51(1):100–107. doi: 10.1016/s0360-3016(01)01572-3. [DOI] [PubMed] [Google Scholar]
  • 22.Brandes AA, Franceschi E, Tosoni A, et al. Temozolomide concomitant and adjuvant to radiotherapy in elderly patients with glioblastoma: correlation with MGMT promoter methylation status. Cancer. 2009;115(15):3512–3518. doi: 10.1002/cncr.24406. [DOI] [PubMed] [Google Scholar]
  • 23.Sijben AE, Mcintyre JB, Roldan GB, et al. Toxicity from chemoradiotherapy in older patients with glioblastoma multiforme. J. Neurooncol. 2008;89(1):97–103. doi: 10.1007/s11060-008-9593-6. [DOI] [PubMed] [Google Scholar]
  • 24.Wiestler B, Claus R, Hartlieb SA, et al. Malignant astrocytomas of elderly patients lack favorable molecular markers: an analysis of the NOA-08 study collective. Neuro Oncol. 2013;15(8):1017–1026. doi: 10.1093/neuonc/not043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Keime-Guibert F, Chinot O, Taillandier L, et al. Radiotherapy for glioblastoma in the elderly. N. Engl. J. Med. 2007;356(15):1527–1535. doi: 10.1056/NEJMoa065901. [DOI] [PubMed] [Google Scholar]
  • 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;22(9):1583–1588. doi: 10.1200/JCO.2004.06.082. [DOI] [PubMed] [Google Scholar]
  • 27.Malmstrom A, Gronberg BH, Marosi C, et al. 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;13(9):916–926. doi: 10.1016/S1470-2045(12)70265-6. [DOI] [PubMed] [Google Scholar]
  • 28.Wick W, Platten M, Meisner C, et al. Temozolomide chemotherapy alone versus radiotherapy alone for malignant astrocytoma in the elderly: the NOA-08 randomised, Phase 3 trial. Lancet Oncol. 2012;13(7):707–715. doi: 10.1016/S1470-2045(12)70164-X. [DOI] [PubMed] [Google Scholar]
  • 29.Gallego Perez-Larraya J, Ducray F, Chinot O, et al. Temozolomide in elderly patients with newly diagnosed glioblastoma and poor performance status: an ANOCEF Phase II trial. J. Clin. Oncol. 2011;29(22):3050–3055. doi: 10.1200/JCO.2011.34.8086. [DOI] [PubMed] [Google Scholar]
  • 30.Perry JR, Laperriere N, O'Callaghan CJ, et al. Short-course radiation plus temozolomide in elderly patients with glioblastoma. N. Engl. J. Med. 2017;376(11):1027–1037. doi: 10.1056/NEJMoa1611977. [DOI] [PubMed] [Google Scholar]; •• EORTC 26062/22061-NCIC CTG CE6: practice-changing randomized Phase III trial of chemoradiotherapy in elderly patients with glioblastoma.
  • 31.Wang N, Jain RK, Batchelor TT. New directions in anti-angiogenic therapy for glioblastoma. Neurotherapeutics. 2017;14(2):321–332. doi: 10.1007/s13311-016-0510-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lu-Emerson C, Duda DG, Emblem KE, et al. Lessons from anti-vascular endothelial growth factor and anti-vascular endothelial growth factor receptor trials in patients with glioblastoma. J. Clin. Oncol. 2015;33(10):1197–1213. doi: 10.1200/JCO.2014.55.9575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Cohen MH, Shen YL, Keegan P, Pazdur R. FDA drug approval summary: bevacizumab (Avastin) as treatment of recurrent glioblastoma multiforme. Oncologist. 2009;14(11):1131–1138. doi: 10.1634/theoncologist.2009-0121. [DOI] [PubMed] [Google Scholar]
  • 34.Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J. Clin. Oncol. 2009;27(28):4733–4740. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
  • 35.Kreisl TN, Kim L, Moore K, 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(5):740–745. doi: 10.1200/JCO.2008.16.3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Wick W, Weller M, Van Den Bent M, Stupp R. Bevacizumab and recurrent malignant gliomas: a European perspective. J. Clin. Oncol. 2010;28(12):e188–e189. doi: 10.1200/JCO.2009.26.9027. author reply e190–e182. [DOI] [PubMed] [Google Scholar]
  • 37.Taal W, Oosterkamp HM, Walenkamp AM, 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(9):943–953. doi: 10.1016/S1470-2045(14)70314-6. [DOI] [PubMed] [Google Scholar]
  • 38.Wick W, Brandes AA, Gorlia T, et al. EORTC 26101 Phase III trial exploring the combination of bevacizumab and lomustine in patients with first progression of a glioblastoma. J. Clin. Oncol. 2016;34(15 Suppl.) 2001–2001. [Google Scholar]
  • 39.Lai A, Tran A, Nghiemphu PL, et al. Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. J. Clin. Oncol. 2011;29(2):142–148. doi: 10.1200/JCO.2010.30.2729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Vredenburgh JJ, Desjardins A, Kirkpatrick JP, et al. Addition of bevacizumab to standard radiation therapy and daily temozolomide is associated with minimal toxicity in newly diagnosed glioblastoma multiforme. Int. J. Radiat. Oncol. Biol. Phys. 2012;82(1):58–66. doi: 10.1016/j.ijrobp.2010.08.058. [DOI] [PubMed] [Google Scholar]
  • 41.Vredenburgh JJ, Desjardins A, Reardon DA, et al. The addition of bevacizumab to standard radiation therapy and temozolomide followed by bevacizumab, temozolomide, and irinotecan for newly diagnosed glioblastoma. Clin. Cancer Res. 2011;17(12):4119–4124. doi: 10.1158/1078-0432.CCR-11-0120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chinot OL, Wick W, Mason W, et al. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N. Engl. J. Med. 2014;370(8):709–722. doi: 10.1056/NEJMoa1308345. [DOI] [PubMed] [Google Scholar]; • AVAGlio: Phase III randomized trial of bevacizumab in newly diagnosed glioblastoma showing lack of efficacy.
  • 43.Gilbert MR, Dignam JJ, Armstrong TS, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 2014;370(8):699–708. doi: 10.1056/NEJMoa1308573. [DOI] [PMC free article] [PubMed] [Google Scholar]; • RTOG-0825: Phase III randomized trial of bevacizumab in newly diagnosed glioblastoma showing lack of efficacy.
  • 44.Hottinger AF, Pacheco P, Stupp R. Tumor treating fields: a novel treatment modality and its use in brain tumors. Neuro Oncol. 2016;18(10):1338–1349. doi: 10.1093/neuonc/now182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Stupp R, Wong ET, Kanner AA, et al. NovoTTF-100A versus physician's choice chemotherapy in recurrent glioblastoma: a randomised Phase III trial of a novel treatment modality. Eur. J. Cancer. 2012;48(14):2192–2202. doi: 10.1016/j.ejca.2012.04.011. [DOI] [PubMed] [Google Scholar]
  • 46.Stupp R, Taillibert S, Kanner AA, et al. Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial. JAMA. 2015;314(23):2535–2543. doi: 10.1001/jama.2015.16669. [DOI] [PubMed] [Google Scholar]; • EF14: Phase III randomized trial of tumor-treating fields in newly diagnosed glioblastoma; the first treatment to confer overall survival benefit in over a decade.
  • 47.Stupp R, Hegi ME, Idbaih A, et al. American Association for Cancer Research. Annual Meeting 2017. WA, DC, USA: 1–5 April 2017. Abstract CT007: tumor treating fields added to standard chemotherapy in newly diagnosed glioblastoma (GBM): final results of a randomized, multi-center, Phase III trial. Presented at. [Google Scholar]
  • 48.Zhu J-J, Pannullo S, Mehdorn M, et al. Quality of life, cognitive function and functional status in the EF-14 trial: a prospective, multi-center trial of TTFields wih temozolomide compared with temozolomide alone in patients with newly diagnosed GBM. Neuro Oncol. 2015;17(Suppl. 5) v9–v9. [Google Scholar]
  • 49.Bernard-Arnoux F, Lamure M, Ducray F, Aulagner G, Honnorat J, Armoiry X. The cost–effectiveness of tumor-treating fields therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. 2016;18(8):1129–1136. doi: 10.1093/neuonc/now102. [DOI] [PMC free article] [PubMed] [Google Scholar]; • A key counter argument to the widespread use of tumor-treating fields.
  • 50.Reardon DA, Wen PY, Wucherpfennig KW, Sampson JH. Immunomodulation for glioblastoma. Curr. Opin. Neurol. 2017;30(3):361–369. doi: 10.1097/WCO.0000000000000451. [DOI] [PubMed] [Google Scholar]
  • 51.Engelhardt B, Vajkoczy P, Weller RO. The movers and shapers in immune privilege of the CNS. Nat. Immunol. 2017;18(2):123–131. doi: 10.1038/ni.3666. [DOI] [PubMed] [Google Scholar]
  • 52.Tan AC, Heimberger AB, Khasraw M. Immune checkpoint inhibitors in gliomas. Curr. Oncol. Rep. 2017;19(4):23. doi: 10.1007/s11912-017-0586-5. [DOI] [PubMed] [Google Scholar]
  • 53.Yovino S, Grossman SA. Severity, etiology and possible consequences of treatment-related lymphopenia in patients with newly diagnosed high-grade gliomas. CNS Oncol. 2012;1(2):149–154. doi: 10.2217/cns.12.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Schuster J, Lai RK, Recht LD, et al. A Phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: the ACT III study. Neuro Oncol. 2015;17(6):854–861. doi: 10.1093/neuonc/nou348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Weller M, Butowski N, Tran D, et al. ACT IV: an international, double-blind, Phase 3 trial of rindopepimut in newly diagnosed, EGFRvIII-expressing glioblastoma. Neuro Oncol. 2016;18(Suppl. 6):vi17–vi18. doi: 10.1016/S1470-2045(17)30517-X. [DOI] [PubMed] [Google Scholar]
  • 56.Reardon DA, Schuster J, Tran DD, et al. ReACT: overall survival from a randomized Phase II study of rindopepimut (CDX-110) plus bevacizumab in relapsed glioblastoma. J. Clin. Oncol. 2015;33(15 Suppl.):2009–2009. [Google Scholar]
  • 57.Reardon DA, Omuro A, Brandes AA, et al. Randomized Phase 3 study evaluating the efficacy and safety of nivolumab vs bevacizumab in patients with recurrent glioblastoma: CheckMate 143. Neuro Oncol. 2017;19(Suppl. 3):iii21–iii21. [Google Scholar]
  • 58.Sampson JH, Maus MV, June CH. Immunotherapy for brain tumors. J. Clin. Oncol. 2017;35(21):2450–2456. doi: 10.1200/JCO.2017.72.8089. [DOI] [PubMed] [Google Scholar]
  • 59.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 60.Rich JN, Reardon DA, Peery T, et al. Phase II trial of gefitinib in recurrent glioblastoma. J. Clin. Oncol. 2004;22(1):133–142. doi: 10.1200/JCO.2004.08.110. [DOI] [PubMed] [Google Scholar]
  • 61.Neyns B, Sadones J, Joosens E, et al. Stratified Phase II trial of cetuximab in patients with recurrent high-grade glioma. Ann. Oncol. 2009;20(9):1596–1603. doi: 10.1093/annonc/mdp032. [DOI] [PubMed] [Google Scholar]
  • 62.Van Den Bent MJ, Brandes AA, Rampling R, 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(8):1268–1274. doi: 10.1200/JCO.2008.17.5984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Li L, Quang TS, Gracely EJ, et al. A Phase II study of anti-epidermal growth factor receptor radioimmunotherapy in the treatment of glioblastoma multiforme. J. Neurosurg. 2010;113(2):192–198. doi: 10.3171/2010.2.JNS091211. [DOI] [PubMed] [Google Scholar]
  • 64.Peereboom DM, Ahluwalia MS, Ye X, et al. NABTT 0502: a Phase II and pharmacokinetic study of erlotinib and sorafenib for patients with progressive or recurrent glioblastoma multiforme. Neuro Oncol. 2013;15(4):490–496. doi: 10.1093/neuonc/nos322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Reardon DA, Nabors LB, Mason WP, et al. Phase I/randomized Phase II study of afatinib, an irreversible ErbB family blocker, with or without protracted temozolomide in adults with recurrent glioblastoma. Neuro Oncol. 2015;17(3):430–439. doi: 10.1093/neuonc/nou160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Westphal M, Heese O, Steinbach JP, et al. A randomised, open label Phase III trial with nimotuzumab, an anti-epidermal growth factor receptor monoclonal antibody in the treatment of newly diagnosed adult glioblastoma. Eur. J. Cancer. 2015;51(4):522–532. doi: 10.1016/j.ejca.2014.12.019. [DOI] [PubMed] [Google Scholar]
  • 67.Gan HK, Fichtel L, Lassman AB, et al. A Phase 1 study evaluating ABT-414 in combination with temozolomide (TMZ) for subjects with recurrent or unresectable glioblastoma (GBM) J. Clin. Oncol. 2014;32(15 Suppl.):2021–2021. [Google Scholar]
  • 68.Reardon DA, Lassman AB, Van Den Bent M, et al. Efficacy and safety results of ABT-414 in combination with radiation and temozolomide in newly diagnosed glioblastoma. Neuro Oncol. 2016;19(7):965–975. doi: 10.1093/neuonc/now257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Batchelor TT, Duda DG, Di Tomaso 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(17):2817–2823. doi: 10.1200/JCO.2009.26.3988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Iwamoto FM, Lamborn KR, Robins HI, 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(8):855–861. doi: 10.1093/neuonc/noq025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.De Groot JF, Lamborn KR, Chang SM, et al. Phase II study of aflibercept in recurrent malignant glioma: a North American Brain Tumor Consortium Study. J. Clin. Oncol. 2011;29(19):2689–2695. doi: 10.1200/JCO.2010.34.1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Pan E, Yu D, Yue B, et al. A prospective Phase II single-institution trial of sunitinib for recurrent malignant glioma. J. Neurooncol. 2012;110(1):111–118. doi: 10.1007/s11060-012-0943-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kreisl TN, Mcneill KA, Sul J, Iwamoto FM, Shih J, Fine HA. A Phase I/II trial of vandetanib for patients with recurrent malignant glioma. Neuro Oncol. 2012;14(12):1519–1526. doi: 10.1093/neuonc/nos265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Muhic A, Poulsen HS, Sorensen M, Grunnet K, Lassen U. Phase II open-label study of nintedanib in patients with recurrent glioblastoma multiforme. J. Neurooncol. 2013;111(2):205–212. doi: 10.1007/s11060-012-1009-y. [DOI] [PubMed] [Google Scholar]
  • 75.Reardon DA, Groves MD, Wen PY, et al. A Phase I/II trial of pazopanib in combination with lapatinib in adult patients with relapsed malignant glioma. Clin. Cancer Res. 2013;19(4):900–908. doi: 10.1158/1078-0432.CCR-12-1707. [DOI] [PubMed] [Google Scholar]
  • 76.Kreisl TN, Smith P, Sul J, et al. Continuous daily sunitinib for recurrent glioblastoma. J. Neuroncol. 2013;111(1):41–48. doi: 10.1007/s11060-012-0988-z. [DOI] [PubMed] [Google Scholar]
  • 77.Batchelor TT, Mulholland P, Neyns B, 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(26):3212–3218. doi: 10.1200/JCO.2012.47.2464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lassman AB, Pugh SL, Gilbert MR, et al. Phase 2 trial of dasatinib in target-selected patients with recurrent glioblastoma (RTOG 0627) Neuro Oncol. 2015;17(7):992–998. doi: 10.1093/neuonc/nov011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chang SM, Wen P, Cloughesy T, et al. Phase II study of CCI-779 in patients with recurrent glioblastoma multiforme. Invest. New Drugs. 2005;23(4):357–361. doi: 10.1007/s10637-005-1444-0. [DOI] [PubMed] [Google Scholar]
  • 80.Galanis E, Buckner JC, Maurer MJ, et al. Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: a North Central Cancer Treatment Group Study. J. Clin. Oncol. 2005;23(23):5294–5304. doi: 10.1200/JCO.2005.23.622. [DOI] [PubMed] [Google Scholar]
  • 81.Kreisl TN, Lassman AB, Mischel PS, et al. A pilot study of everolimus and gefitinib in the treatment of recurrent glioblastoma (GBM) J. Neurooncol. 2009;92(1):99–105. doi: 10.1007/s11060-008-9741-z. [DOI] [PubMed] [Google Scholar]
  • 82.Pitz MW, Eisenhauer EA, Macneil MV, et al. Phase II study of PX-866 in recurrent glioblastoma. Neuro Oncol. 2015;17(9):1270–1274. doi: 10.1093/neuonc/nou365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kreisl TN, Kotliarova S, Butman JA, et al. A Phase I/II trial of enzastaurin in patients with recurrent high-grade gliomas. Neuro Oncol. 2010;12(2):181–189. doi: 10.1093/neuonc/nop042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wick W, Puduvalli VK, Chamberlain MC, et al. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J. Clin. Oncol. 2010;28(7):1168–1174. doi: 10.1200/JCO.2009.23.2595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wen PY, Prados M, Schiff D, 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. 2010;28(15 Suppl.) 2006–2006. [Google Scholar]
  • 86.Wen PY, Schiff D, Cloughesy TF, et al. A Phase II study evaluating the efficacy and safety of AMG 102 (rilotumumab) in patients with recurrent glioblastoma. Neuro Oncol. 2011;13(4):437–446. doi: 10.1093/neuonc/noq198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Reardon DA, Fink KL, Mikkelsen T, 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(34):5610–5617. doi: 10.1200/JCO.2008.16.7510. [DOI] [PubMed] [Google Scholar]
  • 88.Gilbert MR, Kuhn J, Lamborn KR, 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. 2012;106(1):147–153. doi: 10.1007/s11060-011-0650-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Stupp R, Hegi ME, Gorlia T, 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(10):1100–1108. doi: 10.1016/S1470-2045(14)70379-1. [DOI] [PubMed] [Google Scholar]
  • 90.Stupp R, Tosoni A, Bromberg JE, et al. Sagopilone (ZK-EPO, ZK 219477) for recurrent glioblastoma. A Phase II multicenter trial by the European Organisation for Research and Treatment of Cancer (EORTC) Brain Tumor Group. Ann. Oncol. 2011;22(9):2144–2149. doi: 10.1093/annonc/mdq729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Chamberlain MC, Grimm S, Phuphanich S, et al. A Phase 2 trial of verubulin for recurrent glioblastoma: a prospective study by the Brain Tumor Investigational Consortium (BTIC) J. Neurooncol. 2014;118(2):335–343. doi: 10.1007/s11060-014-1437-y. [DOI] [PubMed] [Google Scholar]
  • 92.Galanis E, Jaeckle KA, Maurer MJ, et al. Phase II trial of vorinostat in recurrent glioblastoma multiforme: a north central cancer treatment group study. J. Clin. Oncol. 2009;27(12):2052–2058. doi: 10.1200/JCO.2008.19.0694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Friday BB, Anderson SK, Buckner J, et al. Phase II trial of vorinostat in combination with bortezomib in recurrent glioblastoma: a north central cancer treatment group study. Neuro Oncol. 2012;14(2):215–221. doi: 10.1093/neuonc/nor198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Lee EQ, Reardon DA, Schiff D, et al. Phase II study of panobinostat in combination with bevacizumab for recurrent glioblastoma and anaplastic glioma. Neuro Oncol. 2015;17(6):862–867. doi: 10.1093/neuonc/nou350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Brandes AA, Carpentier AF, Kesari S, et al. A Phase II randomized study of galunisertib monotherapy or galunisertib plus lomustine compared with lomustine monotherapy in patients with recurrent glioblastoma. Neuro Oncol. 2016;18(8):1146–1156. doi: 10.1093/neuonc/now009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mau-Sørensen M, Plotkin SR, Wen PY, et al. A Phase 2 study on efficacy, safety and intratumoral pharmacokinetics of oral selinexor (KPT-330) in patients with recurrent glioblastoma (GBM) J. Clin. Oncol. 2016;34(15 Suppl.):2077–2077. [Google Scholar]
  • 97.Rao SK, Edwards J, Joshi AD, Siu IM, Riggins GJ. A survey of glioblastoma genomic amplifications and deletions. J. Neurooncol. 2010;96(2):169–179. doi: 10.1007/s11060-009-9959-4. [DOI] [PubMed] [Google Scholar]
  • 98.Mellinghoff IK, Wang MY, Vivanco I, et al. Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N. Engl. J. Med. 2005;353(19):2012–2024. doi: 10.1056/NEJMoa051918. [DOI] [PubMed] [Google Scholar]

Articles from CNS Oncology are provided here courtesy of Taylor & Francis

RESOURCES