Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Review ArticleReviewsR

Advances in Experimental Targeted Therapy and Immunotherapy for Patients with Glioblastoma Multiforme

JIRI POLIVKA, JIRI POLIVKA, LUBOS HOLUBEC, TEREZA KUBIKOVA, VLADIMIR PRIBAN, ONDREJ HES, KRISTYNA PIVOVARCIKOVA and INKA TRESKOVA
Anticancer Research January 2017, 37 (1) 21-33;
JIRI POLIVKA JR.
1Biomedical Center, Faculty of Medicine in Plzen, Charles University, Plzen, Czech Republic
2Department of Histology and Embryology, Faculty of Medicine in Plzen, Charles University, Plzen, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JIRI POLIVKA
3Department of Neurology, Faculty of Medicine in Plzen, Charles University, Faculty Hospital Plzen, Plzen, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: polivka@fnplzen.cz
LUBOS HOLUBEC
1Biomedical Center, Faculty of Medicine in Plzen, Charles University, Plzen, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
TEREZA KUBIKOVA
1Biomedical Center, Faculty of Medicine in Plzen, Charles University, Plzen, Czech Republic
2Department of Histology and Embryology, Faculty of Medicine in Plzen, Charles University, Plzen, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
VLADIMIR PRIBAN
4Department of Neurosurgery, Faculty of Medicine in Plzen, Charles University, Czech Republic and Faculty Hospital Plzen, Plzen, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ONDREJ HES
5Department of Pathology, Faculty of Medicine in Plzen, Charles University, and Faculty Hospital Plzen, Plzen, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KRISTYNA PIVOVARCIKOVA
5Department of Pathology, Faculty of Medicine in Plzen, Charles University, and Faculty Hospital Plzen, Plzen, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
INKA TRESKOVA
6Department of Surgery, Faculty of Medicine in Plzen, Charles University, and Faculty Hospital Plzen, Plzen, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Glioblastoma multiforme (GBM) represents the most malignant primary brain tumor in adults with generally dismal prognosis, early clinical deterioration and high mortality. GBM is extremely invasive, characterized by intense and aberrant vascularization and high resistance to multimodal treatment. Standard therapy (surgery, radiotherapy and chemotherapy with temozolomide) has very limited effectiveness, with median overall survival of patients no longer than 15 months. Progress in genetics and epigenetics of GBM over the past decade has revealed various aberrations in cellular signaling pathways, the tumor microenvironment, and pathological angiogenesis. A number of targeted anticancer drugs, such as small-molecule kinase inhibitors and monoclonal antibodies, have been evaluated in clinical trials with newly-diagnosed, as well as recurrent GBM. Unfortunately, to date, only a single anti-angiogenic agent, bevacizumab, has been approved for the treatment of recurrent GBM in the USA and Canada. The novel possibilities of cancer immunotherapy, especially immune checkpoint inhibitors, are being evaluated in clinical trials of patients with GBM. The most recent clinical experiences with targeted therapy as well as immunotherapy of GBM are given in this review. The relative lack of success of some of these approaches recently revealed in well-designed randomized clinical trials is also discussed.

  • Glioblastoma multiforme
  • GBM
  • targeted therapy
  • immunotherapy
  • immune checkpoint inhibitors
  • PD1 inhibition
  • CTLA4 inhibition
  • clinical trials
  • personalized medicine
  • review

Glioblastoma multiforme (GBM) belongs to the largest group of primary central nervous system (CNS) tumors, so-called gliomas, which are formed from supporting glial cells in the brain parenchyma (1, 2). GBM represents the most common and most malignant tumor in this class, with an incidence of 3-4/100,000/year (3, 4). GBM is an extremely invasive and difficult to treat tumor, characterized by intense and aberrant vascularization and high resistance to radiotherapy (RT) and chemotherapy (CHT). The current standard of care for patients with newly-diagnosed GBM comprises of neurosurgery and subsequent concomitant chemoradiotherapy by fractionated external-beam RT and systemic temozolomide followed by systemic temozolomide in the adjuvant setting (5). There are only very limited possibilities for the treatment of subsequent recurrences, generally with minimal clinical efficacy (6). Despite intensive multimodal treatment strategies, the median survival of patients with GBM is still 12.1-14.6 months and only 3-5% of patients survive longer than 3 years (7).

Enormous progress has been made in the genetics and epigenetics of GBM during the past decade. The Cancer Genome Atlas Research Network consortium carried out multiplatform analysis (DNA mutations, mRNA, microRNA expressions) of 500 tumor tissue samples from patients with untreated primary GBM (8). This initiative, together with the other large multiplatform studies, revealed several abnormalities in a diversity of mutated genes and cellular signaling pathways involved in high-grade glioma development and progression (8-12). The most important genetic and epigenetic aberrations were found in following cellular signaling pathways: i) Kirsten rat sarcoma viral oncogene homolog (KRAS) and phosphoinositide 3-kinase (PI3K) oncogenic pathways (88% of GBM), ii) the p53 pathway (87% of GBM), iii) cell-cycle regulatory pathway (78% of GBM), and iv) the newly-discovered alterations in metabolic pathways including isocitrate dehydrogenases 1 and 2 (IDH1/2) mutations (10% of GBM). The mutations in IDH1/2 serve also as an independent and important GBM prognostic factor (13-15). Their routine assessment is now highly recommended in the clinical management of patients with glioma (including GBM) according to the recently updated World Health Organization (WHO) 2016 classification of CNS tumors (16-18).

The GBM microenvironment and its involvement in cancer development and progression was also extensively studied, especially tumor angiogenesis and aberrations in anticancer immune responses (19, 20). Together these findings allow the possibility of developing innovative and better personalized treatment strategies for clinical patient management in the future. The most recent advances in targeted therapy as well as immunotherapy for GBM are given in this review. The relative lack of success of some of these approaches, as recently revealed from well-designed randomized clinical trials, is also discussed.

Perspectives on Novel Therapies for GBM

The standard therapeutic protocols for the treatment of GBM have only limited benefits and provide a median survival of patients of no longer than 15 months (7). The novel so-called targeted therapies can affect and inhibit various tumor-specific features, such as altered cellular signaling pathways, aberrant vascularization, or the tumor microenvironment and impaired anticancer immune response (21, 22). Genetic and epigenetic studies in GBM have revealed potential new targets in cancer cells and the surrounding tumor microenvironment that can be therapeutically influenced by the small molecules and monoclonal antibodies (4, 23). New approaches of GBM immunotherapy could lead to a fundamental breakthrough into the treatment of high-grade gliomas (20).

However, the vast molecular heterogeneity associated with aberrant GBM signaling pathways contributes to the relative lack of success of these therapeutic strategies. A recent study identified a distinct mutation profile of recurrent glioma that varied from the initial mutation analyses in the same patient (24). The exomes of 23 initially low-grade gliomas and recurrent tumors resected from the same patients were sequenced and the mutation profiles were mutually compared. In 10 (43%) of these cases, 50% or fewer of the mutations present in the initial tumor were found at recurrence. Moreover, the mutational profile of GBM is also affected by CHT as recurrent tumors exhibit temozolomide-induced damage to the DNA mismatch repair system resulting in a hypermutated phenotype (25). Another study revealed the possibility of transition proneural gene expression pattern in GBM to a mesenchymal pattern at recurrence that also negatively influenced the effectiveness of new drugs applied at the beginning of the treatment in newly-diagnosed disease (10).

These and other mechanisms, such as the lack of tumor dependence on the proposed target, failure of drug penetration into the CNS, or clonal evolution and antigen escape of tumor after effective therapeutic intervention, could be reasons for the relative lack of success of targeted approaches in the treatment of gliomas. Only a small clinical benefit has been demonstrated with these agents so far, which is discussed in more detail in the following text.

Overcoming these barriers will probably require the use of individualized molecular profiling of each GBM tumor at initial diagnosis and also at recurrence, and application of personalized medicine principles for combinations of targeted therapies with other types of treatment for high-grade gliomas, including GBM.

Inhibitors of Growth Factors and Their Receptors, Inhibitors of Intracellular Signaling Pathways

These are relatively new molecules are able to specifically inhibit various aberrantly activated cell signaling pathways which lead to the formation and progression of cancer (4, 22, 26). Such effects can be achieved by inhibiting specific growth factors and their receptors, including the epidermal growth factor family (EGF) and their receptors (EGFR), platelet-derived growth factors (PDGF) and their receptors (PDGFR), insulin-like growth factors (IGF), fibroblast growth factor (FGF) and their receptors and others that are overexpressed or mutated in a high proportion of GBMs (9, 27).

Molecular aberrations in EGFR signalling, comprising mutations and gene overexpression, are described in approximately 50% of GBMs (8). Therefore aberrantly activated EGFR could be a possible therapeutic target, similar to the situation common in other tumor types (28-30). One of the approved drugs directed against EGFR is the small-molecule kinase inhibitor gefitinib. In an early phase II clinical trial of recurrent GBM treated with gefitinib, the progression-free survival (PFS) at 6 months was 13% and the median overall survival (OS) was 10 months (31). There were more recent studies with gefitinib as monotherapy or in combinations for GBM treatment with results of only very limited efficacy compared to standard treatment (32-34). Another EGFR inhibitor also examined in the GBM setting is erlotinib. A number of phase II trials of erlotinib as a single agent showed minimal benefit for GBM treatment and modest survival benefit in combination with other agents (35-37). Another potentially promising EGFR inhibitor is lapatinib. This agent was tested in multiple clinical trials in patients with GBM but again with very limited antitumor effect (38-40). The newer irreversible EGFR inhibitor afatinib was recently evaluated as a monotherapy and in combination with temozolomide in phase I/II study of patients with recurrent GBM (41). Afatinib had a manageable safety profile but only limited activity. Cetuximab is a chimeric monoclonal antibody with activity against EGFR. Cetuximab was tested in the small group of patients with GBM but also with poor results (42). Some improvement was observed in a phase II study evaluating the combination of cetuximab, irinotecan, and bevacizumab. However, the efficacy data were not superior compared to results with bevacizumab and irinotecan alone (43). The observed effects of EGFR inhibitors in the treatment of patients with GBM are generally weak. Better results could possibly be achieved by stratification of patients eligible for treatment by presence of overexpression or specific mutations of EGFR in their tumor tissue (44-46).

PDGFR is another cell surface receptor frequently overexpressed and activated in GBM, especially in its proneural subtype (8, 47). The aberrant activation of PDGFR assists in the transition from lower-grade glioma to GBM and PDGF ligand stimulates tumor growth and angiogenesis (48, 49). Imatinib is a kinase inhibitor of PDGRF, mast/stem cell growth factor receptor (c-KIT), and oncogene fusion protein BCR–ABL that was also extensively examined in patients with GBM. PFS of 16% at 6 months was observed in one phase II trial of patients with recurrent disease (50). Further multicenter phase II studies confirmed that imatinib as a monotherapy or in combination therapy failed to improve PFS or OS in patients with GBM (51, 52). Multikinase inhibitors influencing tumor angiogenesis, namely sunitinib, sorafenib, and vandetanib, also have inhibitory effect on PDGFR. These substances were also evaluated in the treatment of GBM (53, 54). However, more recent multicentric randomized phase II clinical trial of RT and temozolomide with and without vandetanib in patients with newly diagnosed GBM showed no significant OS benefit of combination compared with the parallel control arm, which led to an early termination of the study (55). Newer multikinase inhibitors affecting PDGFR such as dasatinib or nintedanib also failed to improve OS in patients with recurrent GBM (56-58). Based on the results from these and other clinical trials with various targeted drugs inhibiting overexpressed PDGFR, this approach unfortunately does not seem to be an effective therapeutic strategy for patients with GBM at the moment.

Intracellular signaling pathway components mediate the response of cells to such growth factors and their interactions with cell surface receptors. Inhibition of such aberrant signaling components can be a promising targeted therapeutic strategy in GBM (4, 22). Protein kinase C (PKC) is an important driver of the signal propagation from several growth factors, such as EGF and PDGF, stimulating glioma cell proliferation. Examples of drugs that inhibit PKC and were evaluated in patients with GBM are tamoxifen (59, 60) and enzastaurin (61, 62). Again, minimal or no benefit was observed. Mammalian target of rapamycin (mTOR) is a crucial intracellular protein kinase involved in cell growth signaling. It transduces the signals from PI3K as well as the KRAS pathways (22). Mutations in the tumor suppressor phosphatase and tensin homolog (PTEN), which normally inhibits PI3K signaling, often increases mTOR activity in GBM (27, 63). Therefore selective mTOR inhibition was extensively examined in GBM settings. The small-molecule mTOR inhibitor sirolimus was not effective as a single agent and had limited efficacy in a phase II trial in combination with erlotinib (37, 64). Another mTOR inhibitor, everolimus, showed no clear clinical benefit in combination with gefitinib for recurrent GBM (33). A recent phase II study evaluating everolimus, temozolomide, and RT in patients with newly diagnosed glioblastoma showed no appreciable survival benefit of the combination compared to historical controls treated with conventional therapy (65). The newer selective PI3K inhibitor PX-866 had a low overall response rate, with median PFS at 6 months of only 17% in a phase II study of patients with recurrent GBM (66). However, 21% of participants had durable stable disease even if no association between stable disease and molecular biomarkers was seen. There are many other targeted therapeutics affecting various aberrantly activated intracellular signaling pathways of cancer cells that are being examined in the GBM setting, such as inhibitors of poly (ADP-ribose) polymerase (PARP), signal transducer and activator of transcription 3 (STAT3) and others (4, 23, 67-69).

However, despite enormous advances in the research of targeted oncological therapy during the past two decades, none of these therapeutics have had proven significant PFS or OS benefit in well-designed phase III clinical trial for patients with newly diagnosed or recurrent GBM as a monotherapy or in combination with standard treatment regimes, which remains truly disappointing.

Inhibition of Angiogenesis in GBM

Cancer research has increasingly highlighted the fundamental role of the tumor microenvironment together with pathological angiogenesis and tumor neovascularization for the development and progression of malignant diseases (21, 70). The processes of pathological angiogenesis and possible mechanisms for their therapeutic inhibition have been extensively studied in GBM (19, 71, 72). The major role in tumor angiogenesis is played by vascular growth factors, especially vascular endothelial growth factor (VEGF) and its variant VEGF-A, primarily through its interactions with the VEGFR1 and VEGFR2 receptors found on endothelial as well as cancer cells. Excessive microvascular proliferation and VEGF overexpression were identified in tumor tissues from patients with GBM. Higher intra-tumoral as well as plasma VEGF concentrations were associated with rapid disease progression and presence of early recurrence of GBM (72-77). Therefore, the evaluation of antiangiogenic and anti-VEGF agents in GBM is highly needed.

The most widely used inhibitor of angiogenesis in advanced cancer treatment at the moment is bevacizumab, a humanized IgG1 monoclonal antibody against VEGF-A. Bevacizumab was extensively examined in clinical trials for treatment of recurrent as well as newly-diagnosed GBM, as a single agent and in various combinations with CHT and other targeted therapeutics (71, 78-83). Bevacizumab gained accelerated approval by the US Food and Drug Administration (FDA) for the treatment of recurrent GBM in 2009 based on a high radiographic response rates and prolonged PFS (84). The multicenter phase II BELOB clinical trial undertaken in 14 hospitals in the Netherlands suggested the possible OS benefit for patients with recurrent GBM treated with the combination of bevacizumab plus lomustine versus bevacizumab or lomustine alone (85). However, recently published results from the well-designed phase III European Organization for Research and Treatment of Cancer (EORTC) 26101 clinical trial failed to confirm the OS benefit of bevacizumab plus lomustine by comparison with lomustine alone [9.1 vs. 8.6 months, hazard ratio (HR)=0.95, 95% confidence interval (CI)=0.74-1.21; p=0.65] in patients with first progression of GBM (86). On the other hand, PFS was longer in the combination arm by comparison with lomustine alone arm (4.2 vs. 1.5 months, HR=0.49, 95% CI=0.39-0.61; p<0.0001). Combinations of bevacizumab with standard treatment for newly-diagnosed GBM were also examined with encouraging results in initial phase II studies (87-89). Based on the previous results, two large phase III clinical trials were designed, AVAglio (NCT00943826) and RTOG-0825 (NCT00884741), evaluating bevacizumab-containing regimes for patients with newly-diagnosed GBM. Unfortunately, neither trial demonstrated a benefit in OS for the combination of bevacizumab with standard RT plus temozolomide treatment compared to standard regimen alone (90, 91). Both studies demonstrated PFS survival benefit of combination, but it reached the pre-specified statistical significance only in the AVAglio trial (10.6 vs. 6.2 months, p<0.001). Moreover, the baseline health-related quality of life and performance status were maintained longer and glucocorticoid use was lower in the bevacizumab arm in the AVAglio trial but with more grade 3 and 4 adverse events (66.8% vs. 51.3%). The retrospective analysis of molecular biomarkers in the AVAglio trial showed that patients with both isocitrate dehydrogenase 1 (IDH1) wild-type tumors and proneural pattern of gene expression may have derived 4.3 months of OS benefit with the addition of bevacizumab to a standard regimen (92). Because of the post-hoc nature of this analysis, the predictive effect in relation to bevacizumab treatment must be interpreted with caution. A recent meta-analysis examined clinical trials that compared bevacizumab plus RT/temozolomide with RT/temozolomide alone in patients with newly diagnosed GBM (79). The meta-analysis included 1,738 patients from three well-designed clinical trials. The result failed to demonstrate OS benefit (HR=1.04, 95% CI=0.84-1.29; p=0.71) but identified increased PFS (HR=0.74, 95% CI=0.62-0.88; p=0.0009) for combined treatment with bevacizumab. Moreover, there was no increase in the 6-month survival (p=0.13) and the rate of serious adverse events was higher in the bevacizumab-treated compared to the placebo group. Based on the results from the AVAglio and RTOG-0825 trials, bevacizumab was not approved for the treatment of patients with newly diagnosed GBM and remains a treatment alternative only in the recurrent setting in the USA and in Canada.

There is a substantial number of studies evaluating other inhibitors of angiogenesis in the treatment of newly-diagnosed as well as recurrent GBM. The VEGFR tyrosine kinase inhibitor cediranib showed activity in an early phase II clinical trial as a monotherapy in patients with recurrent GBM (93). Despite the promising results, cediranib demonstrated no PFS benefit as a monotherapy (HR=1.05, 95% CI=0.74-1.50, p=0.9) or in combination with lomustine (HR=0.76, 95% CI=0.53-1.08; p=0.16) versus lomustine alone in patients with recurrent GBM in a phase III clinical trial (94). Cilengitide is an inhibitor of αvβ3 and αvβ5 integrin receptors that also blocks pathological tumor angiogenesis. Cilengitide was evaluated with promising results in phase II study as a monotherapy in patients with recurrent GBM (95). However, the phase III clinical trial evaluating cilengitide combined with standard treatment compared to standard regime alone failed to show significant OS benefit of the combination in patients with newly diagnosed GBM with O-6-methylguanine DNA methyltransferase (MGMT) methylation (26.3 vs. 26.3 months; HR=1.02, 95% CI=0.81-1.29, p=0.86) (96). Another angiogenesis inhibitor aflibercept, a recombinant produced fusion protein that scavenges both VEGF and PDGF, was studied in the recurrent setting, but demonstrated minimal evidence of single-agent activity in patients with GBM with PFS at 6 months of only 7.7% (97). Unfortunately, apart from bevacizumab, no other inhibitor of angiogenesis has been approved for the treatment of patients with newly-diagnosed or recurrent GBM.

Immunotherapy of GBM

Immunotherapy represents a very promising area of multimodal treatment for many types of cancer (98-101). There has also been great progress in immunotherapy research in GBM over the past few years. There are many different approaches currently being evaluated in GBM clinical trials, including passive immunotherapy with antibodies, utilization of autologously stimulated lymphocytes and cytokines, oncolytic virotherapy, and active immunotherapy with vaccine strategies based on tumor cells, peptides, or dendritic cells (DCs) (20, 102, 103).

More than 40% of GBMs carry the unique deletion mutant variant of EGFR called EGFRvIII that is characterized by a deletion of 267 amino acids in the receptor extracellular domain (104, 105). This mutation causes constitutive ligand-independent receptor activation and signal propagation leading to cancer cell proliferation. The enhanced proliferation of EGFRvIII-positive cancer cells together with the lack of EGFRvIII expression in normal non-cancerous cells makes it an ideal candidate for targeted therapy and the use of personalized medicine in GBM treatment. Rindopepimut is a peptide-based vaccine (containing 13 EGFRvIII-specific amino acid sequences) targeted against EGFRvIII surface antigens. The phase I/II multicenter study evaluating rindopepimut in patients with newly diagnosed GBM showed very promising results, with a median PFS of 15.2 months and an OS of 23.6 months (106). Subsequent phase II clinical trial (ACT III) examined rindopepimut in combination with standard RT and temozolomide in 65 patients with newly diagnosed GBM overexpressing EGFRvIII (107). The median OS was 21.8 months. Patients with unmethylated MGMT promoter had an OS of 20.9 months, whereas those with methylated MGMT had longer OS of 40 months. Based on the very promising results from these early clinical trials, the double-blinded randomized multicenter phase III ACT IV clinical trial (NCT01480479) of rindopepimut in patients with newly diagnosed GBM was designed and started enrollment of patients (108). Unfortunately, rindopepimut combined with temozolomide failed to improve OS by comparison with the standard treatment (20.1 vs. 20 months; HR=1.01, 95% CI=0.79-1.30, p=0.93). The study was discontinued in March 2016 after the second interim analysis, which was a true disappointment. At the same time, this example shows the importance of verification of promising preliminary results from early drug development in well-designed randomized placebo controlled phase III clinical trials. Rindopepimut is still being evaluated in the phase II ReACT clinical trial in combination with bevacizumab in patients with recurrent EGFRvIII-positive GBM (NCT01498328).

There are other peptide vaccines targeting tumor antigens, such as the HLA-restricted Wilms tumor 1 (WT1) 9-mer peptide vaccine, that was examined in patients with recurrent GBM in a phase II clinical trial (109). The median PFS was 20 weeks and the PFS at 6 months was 33.3%. A more recent phase II clinical trial with an autologous heat-shock protein-peptide vaccine HSPPC-96 (vitespen) showed promising results in patients with recurrent GBM, with a median OS of 42.6 weeks (110).

DC vaccines use autologous tumor lysates or common tumor antigens to induce an immune response against cancer. These strategies were evaluated in early-phase clinical trials in newly-diagnosed GBM (111-114). DC vaccine loaded with autologous tumor lysate was examined in a phase I clinical trial with 56 patients with relapsed GBM, resulting in a median PFS of 3 months and median OS of 9.6 months, with 2-year OS of 14.8% (115). The same group investigated the integration of DC vaccine into the standard treatment of patients with newly-diagnosed GBM and achieved an unexpected median OS of 24 months (116). Another large double-blinded randomized phase II trial of DC vaccine (DCVax-L) in patients with newly diagnosed GBM also showed encouraging results, with a median OS of 3 years, with 4-year survival reaching 33%, and 27% of patients exceeding 6-year survival from initial surgery (117, 118). However, the clarification of these promising results with DC vaccine strategies are essential in well-designed randomized phase III clinical trials, such as the DCVax-L phase III study (NCT00045968) which is now ongoing and whose final results are eagerly awaited.

The Role of Immune Checkpoint Inhibitors in Glioblastoma Immunotherapy

There has been dramatic success in the treatment of various advanced solid tumor types such as melanoma, renal cancer, lung cancer, head and neck cancer with the novel class of immunomodulatory anticancer agents called immune checkpoint inhibitors (98-101, 119-121). These therapeutics are able to block inhibitory molecules and their receptors on effector immune cells, which leads to a robust T-cell response against the tumor. At the moment there are FDA-approved monoclonal antibodies directed against distinct inhibitory molecules such as ipilimumab targeting cytotoxic T-lymphocyte antigen 4 (CTLA-4), nivolumab and pembrolizumab targeting programmed cell death 1 (PD-1) and atezolizumab targeting programmed cell death ligand 1 (PD-L1), and many others are in development. The great success of immune checkpoint inhibitors in a therapy of a number of advanced solid tumor type1 leading to the evaluation of these compounds in CNS gliomas, including GBM (122, 123).

There is comprehensive pre-clinical research supporting a role for immune checkpoint inhibitors in the treatment of GBM. In a preclinical study with a murine glioma model, treatment with CTLA-4 blockade effectively reversed glioma-induced changes to the CD4+ T-cell compartment and enhanced the antitumor immunity (124). Another study with a mouse model of GBM showed the synergistic effect of combined treatment of systemic CTLA-4 blockade together with intratumoral interleukin 12 (IL12) application leading to tumor eradication even at advanced disease stages (125). The effectiveness of combined CTLA-4 with PD-L1 and indoleamine-2,3-dioxygenase 1 blockade was studied in mouse model of GBM. It was shown that 100% of mice (n=5) survived on the triple combination therapy for a long time (126). A more recent study with a murine model of GBM showed that anti-CTLA-4 plus anti-PD-1 therapy was able to cure 75% of the animals (12 out of 16), even with advanced and late-stage tumors (127).

Support for the rationale of using immune checkpoint inhibitors in the GBM setting has also come from substantial clinical findings. Firstly, these drugs effectively overcome the blood–brain barrier and are active in the CNS. The CTLA-4 inhibitor ipilimumab showed activity in patients with brain metastases of melanoma without significant CNS toxicity (128, 129). The PD-1 inhibitor pembrolizumab was active in the treatment of brain metastases in patients with melanoma or non-small-cell lung cancer with an acceptable safety profile (130, 131). Furthermore, PD-L1 expression level in the tumor tissue was positively associated with the likelihood of clinical benefit with PD-1 inhibitor in non-small-cell lung cancer as well as other tumor types (132, 133). A recent study showed robust and diffuse expression of PD-L1 assessed by immunohistochemistry in newly-diagnosed as well as recurrent GBM (88% vs. 72.2%, respectively), which is a relatively high percentage compared to other cancer types such as melanoma (134). Higher expression of PD-L1 in tumor tissue correlated with worse outcome in another study with 94 patients with GBM (135). Because of the promising pre-clinical experiments, proven activity in the CNS and the presence of targets in GBM tumor tissue, clinical trials with specific immune checkpoint inhibitors are warranted in patients with GBM in newly diagnosed as well as recurrent settings.

Clinical trials evaluating immune checkpoint inhibitors, including ipilimumab, nivolumab, pembrolizumab and others, in patients with GBM are currently being conducted (Table I). The phase III CheckMate 143 study (NCT02017717) is evaluating nivolumab alone and nivolumab plus ipilimumab versus bevacizumab as an active comparator in patients with recurrent GBM. The latest results included the observation of high activity of the nivolumab arm, with 12-month OS of 40% (136). Nivolumab alone was also the best tolerated arm with no new safety signals. Another currently running phase II clinical trial (NCT02337491) is validating the combination of pembrolizumab and bevacizumab in patients with recurrent GBM. Very preliminary results from six patients treated with the combination showed a median OS of 6.8 months and two patients remained alive long-term (327 and 328 days) (137). Pembrolizumab is also being examined in combination with bevacizumab and hypofractionated stereotactic RT in a phase I/II study (NCT02313272) in patients with recurrent GBM. Preliminary results from six patients who were treated with the combination showed durable disease control in all three patients evaluable for response (138). The phase II CheckMate 548 (NCT02667587) study is examining the combination of standard RT plus temozolomide treatment with nivolumab or placebo in patients with newly-diagnosed GBM. Moreover, the phase III CheckMate 498 (NCT02617589) clinical trial is evaluating the head to head comparison of nivolumab to temozolomide both in combination with standard RT in patients with newly diagnosed GBM with unmethylated MGMT promoter in tumor tissue. The preliminary results from both these studies are eagerly awaited. Pembrolizumab is also being examined in the newly diagnosed setting in combination with standard RT plus temozolomide regime in a phase I/II (NCT02530502) clinical trial. The results have not been published yet. Newer checkpoint inhibitors are also being evaluated for the treatment of GBM patients, such as the phase II study of PD-L1 inhibitor durvalumab in newly diagnosed as well as recurrent settings (NCT02336165). On the other hand, there are also conflicting results. A recently published retrospective analysis of 22 patients with heavily pretreated refractory progressive primary brain tumors (including 10 with GBM) treated with pembrolizumab as a salvage agent in four major Israeli brain tumor centers showed only limited OS of no more than 3.2 months (139). The authors do not recommend further use of pembrolizumab for the recurrent primary brain tumors until convincing positive prospective clinical trial data is published.

Immune checkpoint inhibitors represent a significant breakthrough in the treatment of various advanced tumor types (such as melanoma, renal cancer, lung cancer, head and neck cancer, and urinary bladder cancer) in recent years that has dramatically changed the prognosis of patients with cancer (98-101, 119-121). In some cases, therapy with these drugs means long-term disease control or hopefully even cure. Although the preliminary results for these drugs are promising and generally also encouraging in patients with GBM, it is necessary to wait for the mature results from multiple phase III clinical trials that are expected to finish in 2018 and beyond.

Summary

GBM remains a devastating disease for patients and their families, with very poor prognosis and early clinical deterioration, despite aggressive multimodal treatment with surgery, RT and CHT. On the other hand, there have been many novel discoveries in basic and translational research in recent years. The genetic and epigenetic basis of GBM formation and progression has been explored well, together with the mutational changes occurring during the standard-treatment course. Possible molecular and genetic targets in tumor cells that can be affected and inhibit by novel therapeutics are known. A number of targeted anticancer drugs are being evaluated as single agents or in combinations for newly diagnosed as well as recurrent GBM. However, despite excessive research in this area, to date only a single antiangiogenic monoclonal antibody, bevacizumab, has been approved for the treatment of recurrent GBM in the USA and Canada.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

The major ongoing clinical trials incorporating immune checkpoint inhibitors into glioblastoma multiforme (GBM) treatment.

There has been significant progress in the research of immunotherapy strategies for GBM. The very successful anticancer therapeutics, immune checkpoints inhibitors, are now also being extensively evaluated in clinical trials with GBM and the results are eagerly awaited. These and other immunotherapeutic strategies together with a better understanding of the role of targeted anticancer drugs in this context hopefully will shed new light onto the future treatment of this serious and almost exclusively fatal disease.

Acknowledgements

This work was supported by MH CZ - DRO (Faculty Hospital Plzen - FNPl, 00669806) and by the National Sustainability Program I (NPU I) Nr. LO1503 provided by the Ministry of Education Youth and Sports of the Czech Republic.

Footnotes

  • This article is freely accessible online.

  • Conflicts of Interest

    The Authors declare that they have no conflict of interests regarding the publication of this article.

  • Received November 15, 2016.
  • Revision received December 3, 2016.
  • Accepted December 6, 2016.
  • Copyright© 2017 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved

References

  1. ↵
    1. Dolecek TA,
    2. Propp JM,
    3. Stroup NE,
    4. Kruchko C
    : CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro-Oncol 14(Suppl 5): v1-49, 2012.
    OpenUrlFREE Full Text
  2. ↵
    1. Louis DN,
    2. Ohgaki H,
    3. Wiestler OD,
    4. Cavenee WK,
    5. Burger PC,
    6. Jouvet A,
    7. Scheithauer BW,
    8. Kleihues P
    : The 2007 WHO Classification of Tumours of the Central Nervous System. Acta Neuropathol 114: 97-109, 2007.
    OpenUrlCrossRefPubMed
  3. ↵
    1. Ostrom QT,
    2. Gittleman H,
    3. Liao P,
    4. Rouse C,
    5. Chen Y,
    6. Dowling J,
    7. Wolinsky Y,
    8. Kruchko C,
    9. Barnholtz-Sloan J
    : CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2007-2011. Neuro-Oncol 16(Suppl 4): iv1-63, 2014.
    OpenUrlFREE Full Text
  4. ↵
    1. Polivka J Jr.,
    2. Polivka J,
    3. Rohan V,
    4. Topolcan O,
    5. Ferda J
    : New molecularly targeted therapies for glioblastoma multiforme. Anticancer Res 32: 2935-2946, 2012.
    OpenUrlAbstract/FREE Full Text
  5. ↵
    1. Stupp R,
    2. Mason WP,
    3. van den Bent MJ,
    4. Weller M,
    5. Fisher B,
    6. Taphoorn MJB,
    7. Belanger K,
    8. Brandes AA,
    9. Marosi C,
    10. Bogdahn U,
    11. Curschmann J,
    12. Janzer RC,
    13. Ludwin SK,
    14. Gorlia T,
    15. Allgeier A,
    16. Lacombe D,
    17. Cairncross JG,
    18. Eisenhauer E,
    19. Mirimanoff RO,
    20. European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups,
    21. National Cancer Institute of Canada Clinical Trials Group
    : Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352: 987-996, 2005.
    OpenUrlCrossRefPubMed
  6. ↵
    1. Tosoni A,
    2. Franceschi E,
    3. Poggi R,
    4. Brandes AA
    : Relapsed glioblastoma: treatment strategies for initial and subsequent recurrences. Curr Treat Options Oncol 17: 49, 2016.
    OpenUrl
  7. ↵
    1. Krex D,
    2. Klink B,
    3. Hartmann C,
    4. von Deimling A,
    5. Pietsch T,
    6. Simon M,
    7. Sabel M,
    8. Steinbach JP,
    9. Heese O,
    10. Reifenberger G,
    11. Weller M,
    12. Schackert G,
    13. German Glioma Network
    : Long-term survival with glioblastoma multiforme. Brain J Neurol 130: 2596-2606, 2007.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Cancer Genome Atlas Research Network
    : Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455: 1061-1068, 2008.
    OpenUrlCrossRefPubMed
  9. ↵
    1. Parsons DW,
    2. Jones S,
    3. Zhang X,
    4. Lin JC-H,
    5. Leary RJ,
    6. Angenendt P,
    7. Mankoo P,
    8. Carter H,
    9. Siu I-M,
    10. Gallia GL,
    11. Olivi A,
    12. McLendon R,
    13. Rasheed BA,
    14. Keir S,
    15. Nikolskaya T,
    16. Nikolsky Y,
    17. Busam DA,
    18. Tekleab H,
    19. Diaz LA Jr.,
    20. Hartigan J,
    21. Smith DR,
    22. Strausberg RL,
    23. Marie SKN,
    24. Shinjo SMO,
    25. Yan H,
    26. Riggins GJ,
    27. Bigner DD,
    28. Karchin R,
    29. Papadopoulos N,
    30. Parmigiani G,
    31. Vogelstein B,
    32. Velculescu VE,
    33. Kinzler KW
    : An integrated genomic analysis of human glioblastoma multiforme. Science 321: 1807-1812, 2008.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    1. Phillips HS,
    2. Kharbanda S,
    3. Chen R,
    4. Forrest WF,
    5. Soriano RH,
    6. Wu TD,
    7. Misra A,
    8. Nigro JM,
    9. Colman H,
    10. Soroceanu L,
    11. Williams PM,
    12. Modrusan Z,
    13. Feuerstein BG,
    14. Aldape K
    : Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9: 157-173, 2006.
    OpenUrlCrossRefPubMed
    1. Ceccarelli M,
    2. Barthel FP,
    3. Malta TM,
    4. Sabedot TS,
    5. Salama SR,
    6. Murray BA,
    7. Morozova O,
    8. Newton Y,
    9. Radenbaugh A,
    10. Pagnotta SM,
    11. Anjum S,
    12. Wang J,
    13. Manyam G,
    14. Zoppoli P,
    15. Ling S,
    16. Rao AA,
    17. Grifford M,
    18. Cherniack AD,
    19. Zhang H,
    20. Poisson L,
    21. Carlotti CG,
    22. Tirapelli DP da C,
    23. Rao A,
    24. Mikkelsen T,
    25. Lau CC,
    26. Yung WKA,
    27. Rabadan R,
    28. Huse J,
    29. Brat DJ,
    30. Lehman NL,
    31. Barnholtz-Sloan JS,
    32. Zheng S,
    33. Hess K,
    34. Rao G,
    35. Meyerson M,
    36. Beroukhim R,
    37. Cooper L,
    38. Akbani R,
    39. Wrensch M,
    40. Haussler D,
    41. Aldape KD,
    42. Laird PW,
    43. Gutmann DH,
    44. TCGA Research Network,
    45. Noushmehr H,
    46. Iavarone A,
    47. Verhaak RGW
    : Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164: 550-563, 2016.
    OpenUrlCrossRefPubMed
  11. ↵
    1. Polivka J,
    2. Polivka J,
    3. Repik T,
    4. Rohan V,
    5. Hes O,
    6. Topolcan O
    : Co-deletion of 1p/19q as prognostic and predictive biomarker for patients in West Bohemia with anaplastic oligodendroglioma. Anticancer Res 36: 471-476, 2016.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Xia L,
    2. Wu B,
    3. Fu Z,
    4. Feng F,
    5. Qiao E,
    6. Li Q,
    7. Sun C,
    8. Ge M
    : Prognostic role of IDH mutations in gliomas: a meta-analysis of 55 observational studies. Oncotarget 6: 17354-17365, 2015.
    OpenUrl
    1. Polivka J,
    2. Polivka J Jr.,
    3. Rohan V,
    4. Pesta M,
    5. Repik T,
    6. Pitule P,
    7. Topolcan O
    : Isocitrate dehydrogenase-1 mutations as prognostic biomarker in glioblastoma multiforme patients in west bohemia. BioMed Res Int 2014: 735659, 2014.
    OpenUrl
  13. ↵
    1. Polivka J,
    2. Polivka J Jr.,
    3. Rohan V,
    4. Topolcan O
    : Glioblastoma multiforme – a review of pathogenesis, biomarkers and therapeutic perspectives. Cesk Slov Neurol N 76/109: 575-583, 2013.
    OpenUrl
  14. ↵
    1. Louis DN,
    2. Perry A,
    3. Reifenberger G,
    4. von Deimling A,
    5. Figarella-Branger D,
    6. Cavenee WK,
    7. Ohgaki H,
    8. Wiestler OD,
    9. Kleihues P,
    10. Ellison DW
    : The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol (Berl) 131: 803-820, 2016.
    OpenUrlCrossRefPubMed
    1. Polivka J,
    2. Polivka J,
    3. Rohan V,
    4. Topolcan O
    : New treatment paradigm for patients with anaplastic oligodendroglial tumors. Anticancer Res 34: 1587-1594, 2014.
    OpenUrlAbstract/FREE Full Text
  15. ↵
    1. Polivka J,
    2. Krakorova K,
    3. Peterka M,
    4. Topolcan O
    : Current status of biomarker research in neurology. EPMA J 7: 14, 2016.
    OpenUrl
  16. ↵
    1. Popescu AM,
    2. Purcaru SO,
    3. Alexandru O,
    4. Dricu A
    : New perspectives in glioblastoma antiangiogenic therapy. Contemp Oncol Pozn Pol 20: 109-118, 2016.
    OpenUrl
  17. ↵
    1. Kamran N,
    2. Calinescu A,
    3. Candolfi M,
    4. Chandran M,
    5. Mineharu Y,
    6. Asad AS,
    7. Koschmann C,
    8. Nunez FJ,
    9. Lowenstein PR,
    10. Castro MG
    : Recent advances and future of immunotherapy for glioblastoma. Expert Opin Biol Ther: 1-20, 2016.
  18. ↵
    1. Hanahan D,
    2. Weinberg RA
    : Hallmarks of cancer: the next generation. Cell 144: 646-674, 2011.
    OpenUrlCrossRefPubMed
  19. ↵
    1. Polivka J,
    2. Janku F
    : Molecular targets for cancer therapy in the PI3K/AKT/mTOR pathway. Pharmacol Ther 142: 164-175, 2014.
    OpenUrlCrossRefPubMed
  20. ↵
    1. Chen R,
    2. Cohen AL,
    3. Colman H
    : Targeted therapeutics in patients with high-grade gliomas: past, present, and future. Curr Treat Options Oncol 17: 42, 2016.
    OpenUrl
  21. ↵
    1. Johnson BE,
    2. Mazor T,
    3. Hong C,
    4. Barnes M,
    5. Aihara K,
    6. McLean CY,
    7. Fouse SD,
    8. Yamamoto S,
    9. Ueda H,
    10. Tatsuno K,
    11. Asthana S,
    12. Jalbert LE,
    13. Nelson SJ,
    14. Bollen AW,
    15. Gustafson WC,
    16. Charron E,
    17. Weiss WA,
    18. Smirnov IV,
    19. Song JS,
    20. Olshen AB,
    21. Cha S,
    22. Zhao Y,
    23. Moore RA,
    24. Mungall AJ,
    25. Jones SJM,
    26. Hirst M,
    27. Marra MA,
    28. Saito N,
    29. Aburatani H,
    30. Mukasa A,
    31. Berger MS,
    32. Chang SM,
    33. Taylor BS,
    34. Costello JF
    : Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 343: 189-193, 2014.
    OpenUrlAbstract/FREE Full Text
  22. ↵
    1. Cahill DP,
    2. Levine KK,
    3. Betensky RA,
    4. Codd PJ,
    5. Romany CA,
    6. Reavie LB,
    7. Batchelor TT,
    8. Futreal PA,
    9. Stratton MR,
    10. Curry WT,
    11. Iafrate AJ,
    12. Louis DN
    : Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during temozolomide treatment. Clin Cancer Res Off J Am Assoc Cancer Res 13: 2038-2045, 2007.
    OpenUrl
  23. ↵
    1. Polivka J,
    2. Pesta M,
    3. Janku F
    : Testing for oncogenic molecular aberrations in cell-free DNA-based liquid biopsies in the clinic: Are we there yet? Expert Rev Mol Diagn 15: 1631-1644, 2015.
    OpenUrlCrossRefPubMed
  24. ↵
    1. Rao SK,
    2. Edwards J,
    3. Joshi AD,
    4. Siu I-M,
    5. Riggins GJ
    : A survey of glioblastoma genomic amplifications and deletions. J Neurooncol 96: 169-179, 2010.
    OpenUrlCrossRefPubMed
  25. ↵
    1. Ke E-E,
    2. Wu Y-L
    : EGFR as a pharmacological target in EGFR-mutant non-small-cell lung cancer: Where do we stand now? Trends Pharmacol Sci 37: 887-903, 2016.
    OpenUrl
    1. Sacco AG,
    2. Worden FP
    : Molecularly targeted therapy for the treatment of head and neck cancer: a review of the ERBB family inhibitors. OncoTargets Ther 9: 1927-1943, 2016.
    OpenUrl
  26. ↵
    1. Zaniboni A,
    2. Formica V
    : The Best. First. Anti-EGFR before anti-VEGF, in the first-line treatment of RAS wild-type metastatic colorectal cancer: from bench to bedside. Cancer Chemother Pharmacol, 2016.
  27. ↵
    1. Rich JN,
    2. Reardon DA,
    3. Peery T,
    4. Dowell JM,
    5. Quinn JA,
    6. Penne KL,
    7. Wikstrand CJ,
    8. Van Duyn LB,
    9. Dancey JE,
    10. McLendon RE,
    11. Kao JC,
    12. Stenzel TT,
    13. Ahmed Rasheed BK,
    14. Tourt-Uhlig SE,
    15. Herndon JE,
    16. Vredenburgh JJ,
    17. Sampson JH,
    18. Friedman AH,
    19. Bigner DD,
    20. Friedman HS
    : Phase II trial of gefitinib in recurrent glioblastoma. J Clin Oncol Off J Am Soc Clin Oncol 22: 133-142, 2004.
    OpenUrl
  28. ↵
    1. Franceschi E,
    2. Cavallo G,
    3. Lonardi S,
    4. Magrini E,
    5. Tosoni A,
    6. Grosso D,
    7. Scopece L,
    8. Blatt V,
    9. Urbini B,
    10. Pession A,
    11. Tallini G,
    12. Crinò L,
    13. Brandes AA
    : Gefitinib in patients with progressive high-grade gliomas: a multicentre phase II study by Gruppo Italiano Cooperativo di Neuro-Oncologia (GICNO). Br J Cancer 96: 1047-1051, 2007.
    OpenUrlCrossRefPubMed
  29. ↵
    1. Kreisl TN,
    2. Lassman AB,
    3. Mischel PS,
    4. Rosen N,
    5. Scher HI,
    6. Teruya-Feldstein J,
    7. Shaffer D,
    8. Lis E,
    9. Abrey LE
    : A pilot study of everolimus and gefitinib in the treatment of recurrent glioblastoma (GBM). J Neurooncol 92: 99-105, 2009.
    OpenUrlCrossRefPubMed
  30. ↵
    1. Brown N,
    2. McBain C,
    3. Nash S,
    4. Hopkins K,
    5. Sanghera P,
    6. Saran F,
    7. Phillips M,
    8. Dungey F,
    9. Clifton-Hadley L,
    10. Wanek K,
    11. Krell D,
    12. Jeffries S,
    13. Khan I,
    14. Smith P,
    15. Mulholland P
    : Multi-center randomized phase II study comparing cediranib plus gefitinib with cediranib plus placebo in subjects with recurrent/progressive glioblastoma. PloS One 11: e0156369, 2016.
    OpenUrl
  31. ↵
    1. Peereboom DM,
    2. Ahluwalia MS,
    3. Ye X,
    4. Supko JG,
    5. Hilderbrand SL,
    6. Phuphanich S,
    7. Nabors LB,
    8. Rosenfeld MR,
    9. Mikkelsen T,
    10. Grossman SA,
    11. New Approaches to Brain Tumor Therapy Consortium
    : NABTT 0502: a phase II and pharmacokinetic study of erlotinib and sorafenib for patients with progressive or recurrent glioblastoma multiforme. Neuro-Oncol 15: 490-496, 2013.
    OpenUrlAbstract/FREE Full Text
    1. van den Bent MJ,
    2. Brandes AA,
    3. Rampling R,
    4. Kouwenhoven MCM,
    5. Kros JM,
    6. Carpentier AF,
    7. Clement PM,
    8. Frenay M,
    9. Campone M,
    10. Baurain J-F,
    11. Armand J-P,
    12. Taphoorn MJB,
    13. Tosoni A,
    14. Kletzl H,
    15. Klughammer B,
    16. Lacombe D,
    17. Gorlia T
    : Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC Brain Tumor Group study 26034. J Clin Oncol Off J Am Soc Clin Oncol 27: 1268-1274, 2009.
    OpenUrl
  32. ↵
    1. Reardon DA,
    2. Desjardins A,
    3. Vredenburgh JJ,
    4. Gururangan S,
    5. Friedman AH,
    6. Herndon JE,
    7. Marcello J,
    8. Norfleet JA,
    9. McLendon RE,
    10. Sampson JH,
    11. Friedman HS
    : Phase 2 trial of erlotinib plus sirolimus in adults with recurrent glioblastoma. J Neurooncol 96: 219-230, 2010.
    OpenUrlCrossRefPubMed
  33. ↵
    1. Thiessen B,
    2. Stewart C,
    3. Tsao M,
    4. Kamel-Reid S,
    5. Schaiquevich P,
    6. Mason W,
    7. Easaw J,
    8. Belanger K,
    9. Forsyth P,
    10. McIntosh L,
    11. Eisenhauer E
    : A phase I/II trial of GW572016 (lapatinib) in recurrent glioblastoma multiforme: clinical outcomes, pharmacokinetics and molecular correlation. Cancer Chemother Pharmacol 65: 353-361, 2010.
    OpenUrlCrossRefPubMed
    1. Karavasilis V,
    2. Kotoula V,
    3. Pentheroudakis G,
    4. Televantou D,
    5. Lambaki S,
    6. Chrisafi S,
    7. Bobos M,
    8. Fountzilas G
    : A phase I study of temozolomide and lapatinib combination in patients with recurrent high-grade gliomas. J Neurol 260: 1469-1480, 2013.
    OpenUrl
  34. ↵
    1. Reardon DA,
    2. Groves MD,
    3. Wen PY,
    4. Nabors L,
    5. Mikkelsen T,
    6. Rosenfeld S,
    7. Raizer J,
    8. Barriuso J,
    9. McLendon RE,
    10. Suttle AB,
    11. Ma B,
    12. Curtis CM,
    13. Dar MM,
    14. de Bono J
    : A phase I/II trial of pazopanib in combination with lapatinib in adult patients with relapsed malignant glioma. Clin Cancer Res Off J Am Assoc Cancer Res 19: 900-908, 2013.
    OpenUrl
  35. ↵
    1. Reardon DA,
    2. Nabors LB,
    3. Mason WP,
    4. Perry JR,
    5. Shapiro W,
    6. Kavan P,
    7. Mathieu D,
    8. Phuphanich S,
    9. Cseh A,
    10. Fu Y,
    11. Cong J,
    12. Wind S,
    13. Eisenstat DD,
    14. BI 1200 36 Trial Group and the Canadian Brain Tumour Consortium
    : 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 17: 430-439, 2015.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Belda-Iniesta C,
    2. Carpeño J de C,
    3. Saenz EC,
    4. Gutiérrez M,
    5. Perona R,
    6. Barón MG
    : Long-term responses with cetuximab therapy in glioblastoma multiforme. Cancer Biol Ther 5: 912-914, 2006.
    OpenUrlCrossRefPubMed
  37. ↵
    1. Hasselbalch B,
    2. Lassen U,
    3. Hansen S,
    4. Holmberg M,
    5. Sørensen M,
    6. Kosteljanetz M,
    7. Broholm H,
    8. Stockhausen M-T,
    9. Poulsen HS
    : Cetuximab, bevacizumab, and irinotecan for patients with primary glioblastoma and progression after radiation therapy and temozolomide: a phase II trial. Neuro-Oncol 12: 508-516, 2010.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    1. Mellinghoff IK,
    2. Wang MY,
    3. Vivanco I,
    4. Haas-Kogan DA,
    5. Zhu S,
    6. Dia EQ,
    7. Lu KV,
    8. Yoshimoto K,
    9. Huang JHY,
    10. Chute DJ,
    11. Riggs BL,
    12. Horvath S,
    13. Liau LM,
    14. Cavenee WK,
    15. Rao PN,
    16. Beroukhim R,
    17. Peck TC,
    18. Lee JC,
    19. Sellers WR,
    20. Stokoe D,
    21. Prados M,
    22. Cloughesy TF,
    23. Sawyers CL,
    24. Mischel PS
    : Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med 353: 2012-2024, 2005.
    OpenUrlCrossRefPubMed
    1. Wachsberger PR,
    2. Lawrence RY,
    3. Liu Y,
    4. Rice B,
    5. Daskalakis C,
    6. Dicker AP
    : Epidermal growth factor receptor mutation status and RAD51 determine the response of glioblastoma to multimodality therapy with cetuximab, temozolomide, and radiation. Front Oncol 3: 13, 2013.
    OpenUrl
  39. ↵
    1. Gajadhar AS,
    2. Bogdanovic E,
    3. Muñoz DM,
    4. Guha A
    : In situ analysis of mutant EGFRs prevalent in glioblastoma multiforme reveals aberrant dimerization, activation, and differential response to anti-EGFR targeted therapy. Mol Cancer Res MCR 10: 428-440, 2012.
    OpenUrl
  40. ↵
    1. Verhaak RGW,
    2. Hoadley KA,
    3. Purdom E,
    4. Wang V,
    5. Qi Y,
    6. Wilkerson MD,
    7. Miller CR,
    8. Ding L,
    9. Golub T,
    10. Mesirov JP,
    11. Alexe G,
    12. Lawrence M,
    13. O'Kelly M,
    14. Tamayo P,
    15. Weir BA,
    16. Gabriel S,
    17. Winckler W,
    18. Gupta S,
    19. Jakkula L,
    20. Feiler HS,
    21. Hodgson JG,
    22. James CD,
    23. Sarkaria JN,
    24. Brennan C,
    25. Kahn A,
    26. Spellman PT,
    27. Wilson RK,
    28. Speed TP,
    29. Gray JW,
    30. Meyerson M,
    31. Getz G,
    32. Perou CM,
    33. Hayes DN,
    34. Cancer Genome Atlas Research Network
    : Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17: 98-110, 2010.
    OpenUrlCrossRefPubMed
  41. ↵
    1. Hermanson M,
    2. Funa K,
    3. Koopmann J,
    4. Maintz D,
    5. Waha A,
    6. Westermark B,
    7. Heldin CH,
    8. Wiestler OD,
    9. Louis DN,
    10. von Deimling A,
    11. Nistér M
    : Association of loss of heterozygosity on chromosome 17p with high platelet-derived growth factor alpha receptor expression in human malignant gliomas. Cancer Res 56: 164-171, 1996.
    OpenUrlAbstract/FREE Full Text
  42. ↵
    1. Ostman A
    : PDGF receptors – mediators of autocrine tumor growth and regulators of tumor vasculature and stroma. Cytokine Growth Factor Rev 15: 275-286, 2004.
    OpenUrlCrossRefPubMed
  43. ↵
    1. Raymond E,
    2. Brandes AA,
    3. Dittrich C,
    4. Fumoleau P,
    5. Coudert B,
    6. Clement PMJ,
    7. Frenay M,
    8. Rampling R,
    9. Stupp R,
    10. Kros JM,
    11. Heinrich MC,
    12. Gorlia T,
    13. Lacombe D,
    14. van den Bent MJ,
    15. European Organisation for Research and Treatment of Cancer Brain Tumor Group Study
    : Phase II study of imatinib in patients with recurrent gliomas of various histologies: a European Organisation for Research and Treatment of Cancer Brain Tumor Group Study. J Clin Oncol Off J Am Soc Clin Oncol 26: 4659-4665, 2008.
    OpenUrl
  44. ↵
    1. Reardon DA,
    2. Dresemann G,
    3. Taillibert S,
    4. Campone M,
    5. van den Bent M,
    6. Clement P,
    7. Blomquist E,
    8. Gordower L,
    9. Schultz H,
    10. Raizer J,
    11. Hau P,
    12. Easaw J,
    13. Gil M,
    14. Tonn J,
    15. Gijtenbeek A,
    16. Schlegel U,
    17. Bergstrom P,
    18. Green S,
    19. Weir A,
    20. Nikolova Z
    : Multicentre phase II studies evaluating imatinib plus hydroxyurea in patients with progressive glioblastoma. Br J Cancer 101: 1995-2004, 2009.
    OpenUrlCrossRefPubMed
  45. ↵
    1. Dresemann G,
    2. Weller M,
    3. Rosenthal MA,
    4. Wedding U,
    5. Wagner W,
    6. Engel E,
    7. Heinrich B,
    8. Mayer-Steinacker R,
    9. Karup-Hansen A,
    10. Fluge O,
    11. Nowak A,
    12. Mehdorn M,
    13. Schleyer E,
    14. Krex D,
    15. Olver IN,
    16. Steinbach JP,
    17. Hosius C,
    18. Sieder C,
    19. Sorenson G,
    20. Parker R,
    21. Nikolova Z
    : Imatinib in combination with hydroxyurea versus hydroxyurea alone as oral therapy in patients with progressive pretreated glioblastoma resistant to standard dose temozolomide. J Neurooncol 96: 393-402, 2010.
    OpenUrlCrossRefPubMed
  46. ↵
    1. Hainsworth JD,
    2. Ervin T,
    3. Friedman E,
    4. Priego V,
    5. Murphy PB,
    6. Clark BL,
    7. Lamar RE
    : Concurrent radiotherapy and temozolomide followed by temozolomide and sorafenib in the first-line treatment of patients with glioblastoma multiforme. Cancer 116: 3663-3669, 2010.
    OpenUrlCrossRefPubMed
  47. ↵
    1. Drappatz J,
    2. Norden AD,
    3. Wong ET,
    4. Doherty LM,
    5. Lafrankie DC,
    6. Ciampa A,
    7. Kesari S,
    8. Sceppa C,
    9. Gerard M,
    10. Phan P,
    11. Schiff D,
    12. Batchelor TT,
    13. Ligon KL,
    14. Young G,
    15. Muzikansky A,
    16. Weiss SE,
    17. Wen PY
    : Phase I study of vandetanib with radiotherapy and temozolomide for newly diagnosed glioblastoma. Int J Radiat Oncol Biol Phys 78: 85-90, 2010.
    OpenUrlCrossRefPubMed
  48. ↵
    1. Lee EQ,
    2. Kaley TJ,
    3. Duda DG,
    4. Schiff D,
    5. Lassman AB,
    6. Wong ET,
    7. Mikkelsen T,
    8. Purow BW,
    9. Muzikansky A,
    10. Ancukiewicz M,
    11. Huse JT,
    12. Ramkissoon S,
    13. Drappatz J,
    14. Norden AD,
    15. Beroukhim R,
    16. Weiss SE,
    17. Alexander BM,
    18. McCluskey CS,
    19. Gerard M,
    20. Smith KH,
    21. Jain RK,
    22. Batchelor TT,
    23. Ligon KL,
    24. Wen PY
    : A Multicenter, phase II, randomized, noncomparative clinical trial of radiation and temozolomide with or without vandetanib in newly diagnosed glioblastoma patients. Clin Cancer Res Off J Am Assoc Cancer Res 21: 3610-3618, 2015.
    OpenUrl
  49. ↵
    1. Muhic A,
    2. Poulsen HS,
    3. Sorensen M,
    4. Grunnet K,
    5. Lassen U
    : Phase II open-label study of nintedanib in patients with recurrent glioblastoma multiforme. J Neurooncol 111: 205-212, 2013.
    OpenUrlCrossRefPubMed
    1. Norden AD,
    2. Schiff D,
    3. Ahluwalia MS,
    4. Lesser GJ,
    5. Nayak L,
    6. Lee EQ,
    7. Rinne ML,
    8. Muzikansky A,
    9. Dietrich J,
    10. Purow B,
    11. Doherty LM,
    12. LaFrankie DC,
    13. Pulverenti JR,
    14. Rifenburg JA,
    15. Ruland SF,
    16. Smith KH,
    17. Gaffey SC,
    18. McCluskey C,
    19. Ligon KL,
    20. Reardon DA,
    21. Wen PY
    : Phase II trial of triple tyrosine kinase receptor inhibitor nintedanib in recurrent high-grade gliomas. J Neurooncol 121: 297-302, 2015.
    OpenUrlCrossRefPubMed
  50. ↵
    1. Lassman AB,
    2. Pugh SL,
    3. Gilbert MR,
    4. Aldape KD,
    5. Geinoz S,
    6. Beumer JH,
    7. Christner SM,
    8. Komaki R,
    9. DeAngelis LM,
    10. Gaur R,
    11. Youssef E,
    12. Wagner H,
    13. Won M,
    14. Mehta MP
    : Phase 2 trial of dasatinib in target-selected patients with recurrent glioblastoma (RTOG 0627). Neuro-Oncol 17: 992-998, 2015.
    OpenUrlAbstract/FREE Full Text
  51. ↵
    1. Brandes AA,
    2. Ermani M,
    3. Turazzi S,
    4. Scelzi E,
    5. Berti F,
    6. Amistà P,
    7. Rotilio A,
    8. Licata C,
    9. Fiorentino MV
    : Procarbazine and high-dose tamoxifen as a second-line regimen in recurrent high-grade gliomas: a phase II study. J Clin Oncol Off J Am Soc Clin Oncol 17: 645-650, 1999.
    OpenUrl
  52. ↵
    1. Spence AM,
    2. Peterson RA,
    3. Scharnhorst JD,
    4. Silbergeld DL,
    5. Rostomily RC
    : Phase II study of concurrent continuous temozolomide (TMZ) and tamoxifen (TMX) for recurrent malignant astrocytic gliomas. J Neurooncol 70: 91-95, 2004.
    OpenUrlCrossRefPubMed
  53. ↵
    1. Wick W,
    2. Puduvalli VK,
    3. Chamberlain MC,
    4. van den Bent MJ,
    5. Carpentier AF,
    6. Cher LM,
    7. Mason W,
    8. Weller M,
    9. Hong S,
    10. Musib L,
    11. Liepa AM,
    12. Thornton DE,
    13. Fine HA
    : Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J Clin Oncol Off J Am Soc Clin Oncol 28: 1168-1174, 2010.
    OpenUrl
  54. ↵
    1. Kreisl TN,
    2. Kotliarova S,
    3. Butman JA,
    4. Albert PS,
    5. Kim L,
    6. Musib L,
    7. Thornton D,
    8. Fine HA
    : A phase I/II trial of enzastaurin in patients with recurrent high-grade gliomas. Neuro-Oncol 12: 181-189, 2010.
    OpenUrlAbstract/FREE Full Text
  55. ↵
    1. Sathornsumetee S,
    2. Reardon DA,
    3. Desjardins A,
    4. Quinn JA,
    5. Vredenburgh JJ,
    6. Rich JN
    : Molecularly targeted therapy for malignant glioma. Cancer 110: 13-24, 2007.
    OpenUrlCrossRefPubMed
  56. ↵
    1. Akhavan D,
    2. Cloughesy TF,
    3. Mischel PS
    : mTOR signaling in glioblastoma: lessons learned from bench to bedside. Neuro-Oncol 12: 882-889, 2010.
    OpenUrlAbstract/FREE Full Text
  57. ↵
    1. Ma DJ,
    2. Galanis E,
    3. Anderson SK,
    4. Schiff D,
    5. Kaufmann TJ,
    6. Peller PJ,
    7. Giannini C,
    8. Brown PD,
    9. Uhm JH,
    10. McGraw S,
    11. Jaeckle KA,
    12. Flynn PJ,
    13. Ligon KL,
    14. Buckner JC,
    15. Sarkaria JN
    : A phase II trial of everolimus, temozolomide, and radiotherapy in patients with newly diagnosed glioblastoma: NCCTG N057K. Neuro-Oncol 17: 1261-1269, 2015.
    OpenUrlAbstract/FREE Full Text
  58. ↵
    1. Pitz MW,
    2. Eisenhauer EA,
    3. MacNeil MV,
    4. Thiessen B,
    5. Easaw JC,
    6. Macdonald DR,
    7. Eisenstat DD,
    8. Kakumanu AS,
    9. Salim M,
    10. Chalchal H,
    11. Squire J,
    12. Tsao MS,
    13. Kamel-Reid S,
    14. Banerji S,
    15. Tu D,
    16. Powers J,
    17. Hausman DF,
    18. Mason WP
    : Phase II study of PX-866 in recurrent glioblastoma. Neuro-Oncol 17: 1270-1274, 2015.
    OpenUrlAbstract/FREE Full Text
  59. ↵
    1. Bai R-Y,
    2. Staedtke V,
    3. Riggins GJ
    : Molecular targeting of glioblastoma: Drug discovery and therapies. Trends Mol Med 17: 301-312, 2011.
    OpenUrlCrossRefPubMed
    1. Majuelos-Melguizo J,
    2. Rodríguez MI,
    3. López-Jiménez L,
    4. Rodríguez-Vargas JM,
    5. Martí Martín-Consuegra JM,
    6. Serrano-Sáenz S,
    7. Gavard J,
    8. de Almodóvar JMR,
    9. Oliver FJ
    : PARP targeting counteracts gliomagenesis through induction of mitotic catastrophe and aggravation of deficiency in homologous recombination in PTEN-mutant glioma. Oncotarget 6: 4790-4803, 2015.
    OpenUrl
  60. ↵
    1. Gray GK,
    2. McFarland BC,
    3. Nozell SE,
    4. Benveniste EN
    : NF-ĸB and STAT3 in glioblastoma: therapeutic targets coming of age. Expert Rev Neurother 14: 1293-1306, 2014.
    OpenUrlCrossRefPubMed
  61. ↵
    1. Hui L,
    2. Chen Y
    : Tumor microenvironment: Sanctuary of the devil. Cancer Lett 368: 7-13, 2015.
    OpenUrlPubMed
  62. ↵
    1. Khasraw M,
    2. Ameratunga MS,
    3. Grant R,
    4. Wheeler H,
    5. Pavlakis N
    : Antiangiogenic therapy for high-grade glioma. Cochrane Database Syst Rev: CD008218, 2014.
  63. ↵
    1. Norden AD,
    2. Drappatz J,
    3. Wen PY
    : Novel anti-angiogenic therapies for malignant gliomas. Lancet Neurol 7: 1152-1160, 2008.
    OpenUrlCrossRefPubMed
    1. Salmaggi A,
    2. Eoli M,
    3. Frigerio S,
    4. Silvani A,
    5. Gelati M,
    6. Corsini E,
    7. Broggi G,
    8. Boiardi A
    : Intracavitary VEGF, bFGF, IL-8, IL-12 levels in primary and recurrent malignant glioma. J Neurooncol 62: 297-303, 2003.
    OpenUrlCrossRefPubMed
    1. Nam D-H,
    2. Park K,
    3. Suh YL,
    4. Kim J-H
    : Expression of VEGF and brain specific angiogenesis inhibitor-1 in glioblastoma: prognostic significance. Oncol Rep 11: 863-869, 2004.
    OpenUrlPubMed
    1. Beal K,
    2. Abrey LE,
    3. Gutin PH
    : Antiangiogenic agents in the treatment of recurrent or newly diagnosed glioblastoma: analysis of single-agent and combined modality approaches. Radiat Oncol Lond Engl 6: 2, 2011.
    OpenUrl
    1. Sica G,
    2. Lama G,
    3. Anile C,
    4. Geloso MC,
    5. La Torre G,
    6. De Bonis P,
    7. Maira G,
    8. Lauriola L,
    9. Jhanwar-Uniyal M,
    10. Mangiola A
    : Assessment of angiogenesis by CD105 and nestin expression in peritumor tissue of glioblastoma. Int J Oncol 38: 41-49, 2011.
    OpenUrlPubMed
  64. ↵
    1. Afshar Moghaddam N,
    2. Mahsuni P,
    3. Taheri D
    : Evaluation of Endoglin as an Angiogenesis Marker in Glioblastoma. Iran J Pathol 10: 89-96, 2015.
    OpenUrlPubMed
  65. ↵
    1. Curry RC,
    2. Dahiya S,
    3. Alva Venur V,
    4. Raizer JJ,
    5. Ahluwalia MS
    : Bevacizumab in high-grade gliomas: past, present, and future. Expert Rev Anticancer Ther 15: 387-397, 2015.
    OpenUrl
  66. ↵
    1. Fu P,
    2. He Y-S,
    3. Huang Q,
    4. Ding T,
    5. Cen Y-C,
    6. Zhao H-Y,
    7. Wei X
    : Bevacizumab treatment for newly diagnosed glioblastoma: Systematic review and meta-analysis of clinical trials. Mol Clin Oncol 4: 833-838, 2016.
    OpenUrl
    1. Ghiaseddin A,
    2. Peters KB
    : Use of bevacizumab in recurrent glioblastoma. CNS Oncol 4: 157-169, 2015.
    OpenUrl
    1. Vredenburgh JJ,
    2. Desjardins A,
    3. Herndon JE,
    4. Marcello J,
    5. Reardon DA,
    6. Quinn JA,
    7. Rich JN,
    8. Sathornsumetee S,
    9. Gururangan S,
    10. Sampson J,
    11. Wagner M,
    12. Bailey L,
    13. Bigner DD,
    14. Friedman AH,
    15. Friedman HS
    : Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol Off J Am Soc Clin Oncol 25: 4722-4729, 2007.
    OpenUrl
    1. Poulsen HS,
    2. Grunnet K,
    3. Sorensen M,
    4. Olsen P,
    5. Hasselbalch B,
    6. Nelausen K,
    7. Kosteljanetz M,
    8. Lassen U
    : Bevacizumab plus irinotecan in the treatment patients with progressive recurrent malignant brain tumours. Acta Oncol Stockh Swed 48: 52-58, 2009.
    OpenUrl
  67. ↵
    1. Verhoeff JJC,
    2. Lavini C,
    3. van Linde ME,
    4. Stalpers LJA,
    5. Majoie CBLM,
    6. Reijneveld JC,
    7. van Furth WR,
    8. Richel DJ
    : Bevacizumab and dose-intense temozolomide in recurrent high-grade glioma. Ann Oncol Off J Eur Soc Med Oncol ESMO 21: 1723–1727, 2010.
    OpenUrl
  68. ↵
    1. Friedman HS,
    2. Prados MD,
    3. Wen PY,
    4. Mikkelsen T,
    5. Schiff D,
    6. Abrey LE,
    7. Yung WKA,
    8. Paleologos N,
    9. Nicholas MK,
    10. Jensen R,
    11. Vredenburgh J,
    12. Huang J,
    13. Zheng M,
    14. Cloughesy T
    : Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol Off J Am Soc Clin Oncol 27: 4733-4740, 2009.
    OpenUrl
  69. ↵
    1. Taal W,
    2. Oosterkamp HM,
    3. Walenkamp AME,
    4. Dubbink HJ,
    5. Beerepoot LV,
    6. Hanse MCJ,
    7. Buter J,
    8. Honkoop AH,
    9. Boerman D,
    10. de Vos FYF,
    11. Dinjens WNM,
    12. Enting RH,
    13. Taphoorn MJB,
    14. van den Berkmortel FWPJ,
    15. Jansen RLH,
    16. Brandsma D,
    17. Bromberg JEC,
    18. van Heuvel I,
    19. Vernhout RM,
    20. van der Holt B,
    21. van den Bent MJ
    : 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 15: 943-953, 2014.
    OpenUrlCrossRefPubMed
  70. ↵
    1. Wick W,
    2. Brandes AA,
    3. Gorlia T,
    4. Bendszus M,
    5. Sahm F,
    6. Taal W,
    7. Taphoorn MJB,
    8. Domont J,
    9. Idbaih A,
    10. Campone M,
    11. Clement PM,
    12. Stupp R,
    13. Fabbro M,
    14. Rhun EL,
    15. Dubois F,
    16. Klein M,
    17. Platten M,
    18. Weller M,
    19. Golfinopoulos V,
    20. Bent MJVD
    : EORTC 26101 phase III trial exploring the combination of bevacizumab and lomustine in patients with first progression of a glioblastoma. J Clin Oncol 34, 2016.
  71. ↵
    1. Vredenburgh JJ,
    2. Desjardins A,
    3. Reardon DA,
    4. Peters KB,
    5. Herndon JE,
    6. Marcello J,
    7. Kirkpatrick JP,
    8. Sampson JH,
    9. Bailey L,
    10. Threatt S,
    11. Friedman AH,
    12. Bigner DD,
    13. Friedman HS
    : The addition of bevacizumab to standard radiation therapy and temozolomide followed by bevacizumab, temozolomide, and irinotecan for newly diagnosed glioblastoma. Clin Cancer Res Off J Am Assoc Cancer Res 17: 4119-4124, 2011.
    OpenUrl
    1. Hainsworth JD,
    2. Shih KC,
    3. Shepard GC,
    4. Tillinghast GW,
    5. Brinker BT,
    6. Spigel DR
    : Phase II study of concurrent radiation therapy, temozolomide, and bevacizumab followed by bevacizumab/everolimus as first-line treatment for patients with glioblastoma. Clin Adv Hematol Oncol HO 10: 240-246, 2012.
    OpenUrl
  72. ↵
    1. Lai A,
    2. Tran A,
    3. Nghiemphu PL,
    4. Pope WB,
    5. Solis OE,
    6. Selch M,
    7. Filka E,
    8. Yong WH,
    9. Mischel PS,
    10. Liau LM,
    11. Phuphanich S,
    12. Black K,
    13. Peak S,
    14. Green RM,
    15. Spier CE,
    16. Kolevska T,
    17. Polikoff J,
    18. Fehrenbacher L,
    19. Elashoff R,
    20. Cloughesy T
    : Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. J Clin Oncol Off J Am Soc Clin Oncol 29: 142-148, 2011.
    OpenUrl
  73. ↵
    1. Gilbert MR,
    2. Dignam JJ,
    3. Armstrong TS,
    4. Wefel JS,
    5. Blumenthal DT,
    6. Vogelbaum MA,
    7. Colman H,
    8. Chakravarti A,
    9. Pugh S,
    10. Won M,
    11. Jeraj R,
    12. Brown PD,
    13. Jaeckle KA,
    14. Schiff D,
    15. Stieber VW,
    16. Brachman DG,
    17. Werner-Wasik M,
    18. Tremont-Lukats IW,
    19. Sulman EP,
    20. Aldape KD,
    21. Curran WJ,
    22. Mehta MP
    : A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 370: 699-708, 2014.
    OpenUrlCrossRefPubMed
  74. ↵
    1. Chinot OL,
    2. Wick W,
    3. Mason W,
    4. Henriksson R,
    5. Saran F,
    6. Nishikawa R,
    7. Carpentier AF,
    8. Hoang-Xuan K,
    9. Kavan P,
    10. Cernea D,
    11. Brandes AA,
    12. Hilton M,
    13. Abrey L,
    14. Cloughesy T
    : Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 370: 709-722, 2014.
    OpenUrlCrossRefPubMed
  75. ↵
    1. Sandmann T,
    2. Bourgon R,
    3. Garcia J,
    4. Li C,
    5. Cloughesy T,
    6. Chinot OL,
    7. Wick W,
    8. Nishikawa R,
    9. Mason W,
    10. Henriksson R,
    11. Saran F,
    12. Lai A,
    13. Moore N,
    14. Kharbanda S,
    15. Peale F,
    16. Hegde P,
    17. Abrey LE,
    18. Phillips HS,
    19. Bais C
    : Patients with proneural glioblastoma may derive overall survival benefit from the addition of bevacizumab to first-line radiotherapy and temozolomide: retrospective analysis of the AVAglio trial. J Clin Oncol Off J Am Soc Clin Oncol 33: 2735-2744, 2015.
    OpenUrl
  76. ↵
    1. Batchelor TT,
    2. Duda DG,
    3. di Tomaso E,
    4. Ancukiewicz M,
    5. Plotkin SR,
    6. Gerstner E,
    7. Eichler AF,
    8. Drappatz J,
    9. Hochberg FH,
    10. Benner T,
    11. Louis DN,
    12. Cohen KS,
    13. Chea H,
    14. Exarhopoulos A,
    15. Loeffler JS,
    16. Moses MA,
    17. Ivy P,
    18. Sorensen AG,
    19. Wen PY,
    20. Jain RK
    : Phase II study of cediranib, an oral pan-vascular endothelial growth factor receptor tyrosine kinase inhibitor, in patients with recurrent glioblastoma. J Clin Oncol Off J Am Soc Clin Oncol 28: 2817-2823, 2010.
    OpenUrl
  77. ↵
    1. Batchelor TT,
    2. Mulholland P,
    3. Neyns B,
    4. Nabors LB,
    5. Campone M,
    6. Wick A,
    7. Mason W,
    8. Mikkelsen T,
    9. Phuphanich S,
    10. Ashby LS,
    11. Degroot J,
    12. Gattamaneni R,
    13. Cher L,
    14. Rosenthal M,
    15. Payer F,
    16. Jürgensmeier JM,
    17. Jain RK,
    18. Sorensen AG,
    19. Xu J,
    20. Liu Q,
    21. van den Bent M
    : 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 Off J Am Soc Clin Oncol 31: 3212-3218, 2013.
    OpenUrl
  78. ↵
    1. Reardon DA,
    2. Fink KL,
    3. Mikkelsen T,
    4. Cloughesy TF,
    5. O'Neill A,
    6. Plotkin S,
    7. Glantz M,
    8. Ravin P,
    9. Raizer JJ,
    10. Rich KM,
    11. Schiff D,
    12. Shapiro WR,
    13. Burdette-Radoux S,
    14. Dropcho EJ,
    15. Wittemer SM,
    16. Nippgen J,
    17. Picard M,
    18. Nabors LB
    : Randomized phase II study of cilengitide, an integrin-targeting arginine-glycine-aspartic acid peptide, in recurrent glioblastoma multiforme. J Clin Oncol Off J Am Soc Clin Oncol 26: 5610-5617, 2008.
    OpenUrl
  79. ↵
    1. Stupp R,
    2. Hegi ME,
    3. Gorlia T,
    4. Erridge SC,
    5. Perry J,
    6. Hong Y-K,
    7. Aldape KD,
    8. Lhermitte B,
    9. Pietsch T,
    10. Grujicic D,
    11. Steinbach JP,
    12. Wick W,
    13. Tarnawski R,
    14. Nam D-H,
    15. Hau P,
    16. Weyerbrock A,
    17. Taphoorn MJB,
    18. Shen C-C,
    19. Rao N,
    20. Thurzo L,
    21. Herrlinger U,
    22. Gupta T,
    23. Kortmann R-D,
    24. Adamska K,
    25. McBain C,
    26. Brandes AA,
    27. Tonn JC,
    28. Schnell O,
    29. Wiegel T,
    30. Kim C-Y,
    31. Nabors LB,
    32. Reardon DA,
    33. van den Bent MJ,
    34. Hicking C,
    35. Markivskyy A,
    36. Picard M,
    37. Weller M,
    38. European Organisation for Research and Treatment of Cancer (EORTC),
    39. Canadian Brain Tumor Consortium,
    40. CENTRIC study team
    : 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 15: 1100-1108, 2014.
    OpenUrlCrossRefPubMed
  80. ↵
    1. de Groot JF,
    2. Lamborn KR,
    3. Chang SM,
    4. Gilbert MR,
    5. Cloughesy TF,
    6. Aldape K,
    7. Yao J,
    8. Jackson EF,
    9. Lieberman F,
    10. Robins HI,
    11. Mehta MP,
    12. Lassman AB,
    13. Deangelis LM,
    14. Yung WKA,
    15. Chen A,
    16. Prados MD,
    17. Wen PY
    : Phase II study of aflibercept in recurrent malignant glioma: a North American Brain Tumor Consortium study. J Clin Oncol Off J Am Soc Clin Oncol 29: 2689-2695, 2011.
    OpenUrl
  81. ↵
    1. Curtis SA,
    2. Cohen JV,
    3. Kluger HM
    : Evolving immuno-therapy approaches for renal cell carcinoma. Curr Oncol Rep 18: 57, 2016.
    OpenUrl
    1. Khanna P,
    2. Blais N,
    3. Gaudreau P-O,
    4. Corrales-Rodriguez L
    : Immunotherapy comes of age in lung cancer. Clin Lung Cancer, 2016.
    1. Donin NM,
    2. Lenis AT,
    3. Holden S,
    4. Drakaki A,
    5. Pantuck A,
    6. Belldegrun A,
    7. Chamie K
    : Immunotherapy in the treatment of urothelial carcinoma. J Urol, 2016.
  82. ↵
    1. Margolin K
    : The promise of molecularly targeted and immunotherapy for advanced melanoma. Curr Treat Options Oncol 17: 48, 2016.
    OpenUrl
  83. ↵
    1. Fecci PE,
    2. Heimberger AB,
    3. Sampson JH
    : Immunotherapy for primary brain tumors: no longer a matter of privilege. Clin Cancer Res Off J Am Assoc Cancer Res 20: 5620-5629, 2014.
    OpenUrl
  84. ↵
    1. Weathers S-P,
    2. Gilbert MR
    : Current challenges in designing GBM trials for immunotherapy. J Neurooncol 123: 331-337, 2015.
    OpenUrl
  85. ↵
    1. Pelloski CE,
    2. Ballman KV,
    3. Furth AF,
    4. Zhang L,
    5. Lin E,
    6. Sulman EP,
    7. Bhat K,
    8. McDonald JM,
    9. Yung WKA,
    10. Colman H,
    11. Woo SY,
    12. Heimberger AB,
    13. Suki D,
    14. Prados MD,
    15. Chang SM,
    16. Barker FG,
    17. Buckner JC,
    18. James CD,
    19. Aldape K
    : Epidermal growth factor receptor variant III status defines clinically distinct subtypes of glioblastoma. J Clin Oncol Off J Am Soc Clin Oncol 25: 2288-2294, 2007.
    OpenUrl
  86. ↵
    1. Gan HK,
    2. Kaye AH,
    3. Luwor RB
    : The EGFRvIII variant in glioblastoma multiforme. J Clin Neurosci Off J Neurosurg Soc Australas 16: 748-754, 2009.
    OpenUrl
  87. ↵
    1. Sampson JH,
    2. Aldape KD,
    3. Archer GE,
    4. Coan A,
    5. Desjardins A,
    6. Friedman AH,
    7. Friedman HS,
    8. Gilbert MR,
    9. Herndon JE,
    10. McLendon RE,
    11. Mitchell DA,
    12. Reardon DA,
    13. Sawaya R,
    14. Schmittling R,
    15. Shi W,
    16. Vredenburgh JJ,
    17. Bigner DD,
    18. Heimberger AB
    : Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro-Oncol 13: 324-333, 2011.
    OpenUrlAbstract/FREE Full Text
  88. ↵
    1. Schuster J,
    2. Lai RK,
    3. Recht LD,
    4. Reardon DA,
    5. Paleologos NA,
    6. Groves MD,
    7. Mrugala MM,
    8. Jensen R,
    9. Baehring JM,
    10. Sloan A,
    11. Archer GE,
    12. Bigner DD,
    13. Cruickshank S,
    14. Green JA,
    15. Keler T,
    16. Davis TA,
    17. Heimberger AB,
    18. Sampson JH
    : A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: the ACT III study. Neuro-Oncol 17: 854-861, 2015.
    OpenUrlAbstract/FREE Full Text
  89. ↵
    1. Zussman BM,
    2. Engh JA
    : Outcomes of the ACT III study: Rindopepimut (CDX-110) therapy for glioblastoma. Neurosurgery 76: N17, 2015.
    OpenUrl
  90. ↵
    1. Izumoto S,
    2. Tsuboi A,
    3. Oka Y,
    4. Suzuki T,
    5. Hashiba T,
    6. Kagawa N,
    7. Hashimoto N,
    8. Maruno M,
    9. Elisseeva OA,
    10. Shirakata T,
    11. Kawakami M,
    12. Oji Y,
    13. Nishida S,
    14. Ohno S,
    15. Kawase I,
    16. Hatazawa J,
    17. Nakatsuka S-I,
    18. Aozasa K,
    19. Morita S,
    20. Sakamoto J,
    21. Sugiyama H,
    22. Yoshimine T
    : Phase II clinical trial of Wilms tumor 1 peptide vaccination for patients with recurrent glioblastoma multiforme. J Neurosurg 108: 963-971, 2008.
    OpenUrlCrossRefPubMed
  91. ↵
    1. Bloch O,
    2. Crane CA,
    3. Fuks Y,
    4. Kaur R,
    5. Aghi MK,
    6. Berger MS,
    7. Butowski NA,
    8. Chang SM,
    9. Clarke JL,
    10. McDermott MW,
    11. Prados MD,
    12. Sloan AE,
    13. Bruce JN,
    14. Parsa AT
    : Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: a phase II, single-arm trial. Neuro-Oncol 16: 274-279, 2014.
    OpenUrlAbstract/FREE Full Text
  92. ↵
    1. Prins RM,
    2. Soto H,
    3. Konkankit V,
    4. Odesa SK,
    5. Eskin A,
    6. Yong WH,
    7. Nelson SF,
    8. Liau LM
    : Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res Off J Am Assoc Cancer Res 17: 1603-1615, 2011.
    OpenUrl
    1. Phuphanich S,
    2. Wheeler CJ,
    3. Rudnick JD,
    4. Mazer M,
    5. Wang H,
    6. Nuño MA,
    7. Richardson JE,
    8. Fan X,
    9. Ji J,
    10. Chu RM,
    11. Bender JG,
    12. Hawkins ES,
    13. Patil CG,
    14. Black KL,
    15. Yu JS
    : Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol Immunother CII 62: 125-135, 2013.
    OpenUrl
    1. Yang L,
    2. Guo G,
    3. Niu X,
    4. Liu J
    : Dendritic Cell-Based Immunotherapy Treatment for Glioblastoma Multiforme. BioMed Res Int 2015: 717530, 2015.
    OpenUrl
  93. ↵
    1. Fong B,
    2. Jin R,
    3. Wang X,
    4. Safaee M,
    5. Lisiero DN,
    6. Yang I,
    7. Li G,
    8. Liau LM,
    9. Prins RM
    : Monitoring of regulatory T cell frequencies and expression of CTLA-4 on T-cells, before and after DC vaccination, can predict survival in GBM patients. PloS One 7: e32614, 2012.
    OpenUrlCrossRefPubMed
  94. ↵
    1. De Vleeschouwer S,
    2. Fieuws S,
    3. Rutkowski S,
    4. Van Calenbergh F,
    5. Van Loon J,
    6. Goffin J,
    7. Sciot R,
    8. Wilms G,
    9. Demaerel P,
    10. Warmuth-Metz M,
    11. Soerensen N,
    12. Wolff JEA,
    13. Wagner S,
    14. Kaempgen E,
    15. Van Gool SW
    : Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res Off J Am Assoc Cancer Res 14: 3098-3104, 2008.
    OpenUrl
  95. ↵
    1. Ardon H,
    2. Van Gool S,
    3. Lopes IS,
    4. Maes W,
    5. Sciot R,
    6. Wilms G,
    7. Demaerel P,
    8. Bijttebier P,
    9. Claes L,
    10. Goffin J,
    11. Van Calenbergh F,
    12. De Vleeschouwer S
    : Integration of autologous dendritic cell-based immunotherapy in the primary treatment for patients with newly diagnosed glioblastoma multiforme: a pilot study. J Neurooncol 99: 261-272, 2010.
    OpenUrlCrossRefPubMed
  96. ↵
    1. Hdeib A,
    2. Sloan AE
    : Dendritic cell immunotherapy for solid tumors: evaluation of the DCVax® platform in the treatment of glioblastoma multiforme. CNS Oncol 4: 63-69, 2015.
    OpenUrlCrossRefPubMed
  97. ↵
    1. Polyzoidis S,
    2. Ashkan K
    : DCVax®-L--developed by Northwest Biotherapeutics. Hum Vaccines Immunother 10: 3139-3145, 2014.
    OpenUrl
  98. ↵
    1. Schoppy DW,
    2. Sunwoo JB
    : Immunotherapy for head and neck squamous cell carcinoma. Hematol Oncol Clin North Am 29: 1033-1043, 2015.
    OpenUrlPubMed
    1. Modena A,
    2. Ciccarese C,
    3. Iacovelli R,
    4. Brunelli M,
    5. Montironi R,
    6. Fiorentino M,
    7. Tortora G,
    8. Massari F
    : Immune checkpoint inhibitors and prostate cancer: A new frontier? Oncol Rev 10: 293, 2016.
    OpenUrl
  99. ↵
    1. Mittica G,
    2. Genta S,
    3. Aglietta M,
    4. Valabrega G
    : Immune checkpoint inhibitors: A new opportunity in the treatment of ovarian cancer? Int J Mol Sci 17, 2016.
  100. ↵
    1. Preusser M,
    2. Lim M,
    3. Hafler DA,
    4. Reardon DA,
    5. Sampson JH
    : Prospects of immune checkpoint modulators in the treatment of glioblastoma. Nat Rev Neurol 11: 504-514, 2015.
    OpenUrlCrossRefPubMed
  101. ↵
    1. Carter T,
    2. Shaw H,
    3. Cohn-Brown D,
    4. Chester K,
    5. Mulholland P
    : Ipilimumab and bevacizumab in glioblastoma. Clin Oncol R Coll Radiol G B, 2016.
  102. ↵
    1. Fecci PE,
    2. Ochiai H,
    3. Mitchell DA,
    4. Grossi PM,
    5. Sweeney AE,
    6. Archer GE,
    7. Cummings T,
    8. Allison JP,
    9. Bigner DD,
    10. Sampson JH
    : Systemic CTLA-4 blockade ameliorates glioma-induced changes to the CD4+ T-cell compartment without affecting regulatory T-cell function. Clin Cancer Res Off J Am Assoc Cancer Res 13: 2158-2167, 2007.
    OpenUrl
  103. ↵
    1. Vom Berg J,
    2. Vrohlings M,
    3. Haller S,
    4. Haimovici A,
    5. Kulig P,
    6. Sledzinska A,
    7. Weller M,
    8. Becher B
    : Intratumoral IL-12 combined with CTLA-4 blockade elicits T-cell-mediated glioma rejection. J Exp Med 210: 2803-2811, 2013.
    OpenUrlAbstract/FREE Full Text
  104. ↵
    1. Wainwright DA,
    2. Chang AL,
    3. Dey M,
    4. Balyasnikova IV,
    5. Kim CK,
    6. Tobias A,
    7. Cheng Y,
    8. Kim JW,
    9. Qiao J,
    10. Zhang L,
    11. Han Y,
    12. Lesniak MS
    : Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res Off J Am Assoc Cancer Res 20: 5290-5301, 2014.
    OpenUrl
  105. ↵
    1. Reardon DA,
    2. Gokhale PC,
    3. Klein SR,
    4. Ligon KL,
    5. Rodig SJ,
    6. Ramkissoon SH,
    7. Jones KL,
    8. Conway AS,
    9. Liao X,
    10. Zhou J,
    11. Wen PY,
    12. Van Den Abbeele AD,
    13. Hodi FS,
    14. Qin L,
    15. Kohl NE,
    16. Sharpe AH,
    17. Dranoff G,
    18. Freeman GJ
    : Glioblastoma eradication following immune checkpoint blockade in an orthotopic, immunocompetent model. Cancer Immunol Res 4: 124-135, 2016.
    OpenUrlAbstract/FREE Full Text
  106. ↵
    1. Margolin K,
    2. Ernstoff MS,
    3. Hamid O,
    4. Lawrence D,
    5. McDermott D,
    6. Puzanov I,
    7. Wolchok JD,
    8. Clark JI,
    9. Sznol M,
    10. Logan TF,
    11. Richards J,
    12. Michener T,
    13. Balogh A,
    14. Heller KN,
    15. Hodi FS
    : Ipilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trial. Lancet Oncol 13: 459-465, 2012.
    OpenUrlCrossRefPubMed
  107. ↵
    1. Di Giacomo AM,
    2. Ascierto PA,
    3. Queirolo P,
    4. Pilla L,
    5. Ridolfi R,
    6. Santinami M,
    7. Testori A,
    8. Simeone E,
    9. Guidoboni M,
    10. Maurichi A,
    11. Orgiano L,
    12. Spadola G,
    13. Del Vecchio M,
    14. Danielli R,
    15. Calabrò L,
    16. Annesi D,
    17. Giannarelli D,
    18. Maccalli C,
    19. Fonsatti E,
    20. Parmiani G,
    21. Maio M
    : Three-year follow-up of advanced melanoma patients who received ipilimumab plus fotemustine in the Italian Network for Tumor Biotherapy (NIBIT)-M1 phase II study. Ann Oncol Off J Eur Soc Med Oncol ESMO 26: 798-803, 2015.
    OpenUrl
  108. ↵
    1. Goldberg SB,
    2. Gettinger SN,
    3. Mahajan A,
    4. Chiang AC,
    5. Herbst RS,
    6. Sznol M,
    7. Tsiouris AJ,
    8. Cohen J,
    9. Vortmeyer A,
    10. Jilaveanu L,
    11. Yu J,
    12. Hegde U,
    13. Speaker S,
    14. Madura M,
    15. Ralabate A,
    16. Rivera A,
    17. Rowen E,
    18. Gerrish H,
    19. Yao X,
    20. Chiang V,
    21. Kluger HM
    : Pembrolizumab for patients with melanoma or non-small-cell lung cancer and untreated brain metastases: early analysis of a non-randomised, open-label, phase 2 trial. Lancet Oncol 17: 976-983, 2016.
    OpenUrl
  109. ↵
    1. Rosell R,
    2. Karachaliou N
    : Trends in immunotherapy for brain metastases. Lancet Oncol 17: 859-860, 2016.
    OpenUrl
  110. ↵
    1. Garon EB,
    2. Rizvi NA,
    3. Hui R,
    4. Leighl N,
    5. Balmanoukian AS,
    6. Eder JP,
    7. Patnaik A,
    8. Aggarwal C,
    9. Gubens M,
    10. Horn L,
    11. Carcereny E,
    12. Ahn M-J,
    13. Felip E,
    14. Lee J-S,
    15. Hellmann MD,
    16. Hamid O,
    17. Goldman JW,
    18. Soria J-C,
    19. Dolled-Filhart M,
    20. Rutledge RZ,
    21. Zhang J,
    22. Lunceford JK,
    23. Rangwala R,
    24. Lubiniecki GM,
    25. Roach C,
    26. Emancipator K,
    27. Gandhi L,
    28. KEYNOTE-001 Investigators
    : Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372: 2018-2028, 2015.
    OpenUrlCrossRefPubMed
  111. ↵
    1. Herbst RS,
    2. Soria J-C,
    3. Kowanetz M,
    4. Fine GD,
    5. Hamid O,
    6. Gordon MS,
    7. Sosman JA,
    8. McDermott DF,
    9. Powderly JD,
    10. Gettinger SN,
    11. Kohrt HEK,
    12. Horn L,
    13. Lawrence DP,
    14. Rost S,
    15. Leabman M,
    16. Xiao Y,
    17. Mokatrin A,
    18. Koeppen H,
    19. Hegde PS,
    20. Mellman I,
    21. Chen DS,
    22. Hodi FS
    : Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 515: 563-567, 2014.
    OpenUrlCrossRefPubMed
  112. ↵
    1. Berghoff AS,
    2. Kiesel B,
    3. Widhalm G,
    4. Rajky O,
    5. Ricken G,
    6. Wöhrer A,
    7. Dieckmann K,
    8. Filipits M,
    9. Brandstetter A,
    10. Weller M,
    11. Kurscheid S,
    12. Hegi ME,
    13. Zielinski CC,
    14. Marosi C,
    15. Hainfellner JA,
    16. Preusser M,
    17. Wick W
    : Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro-Oncol 17: 1064-1075, 2015.
    OpenUrlAbstract/FREE Full Text
  113. ↵
    1. Nduom EK,
    2. Wei J,
    3. Yaghi NK,
    4. Huang N,
    5. Kong L-Y,
    6. Gabrusiewicz K,
    7. Ling X,
    8. Zhou S,
    9. Ivan C,
    10. Chen JQ,
    11. Burks JK,
    12. Fuller GN,
    13. Calin GA,
    14. Conrad CA,
    15. Creasy C,
    16. Ritthipichai K,
    17. Radvanyi L,
    18. Heimberger AB
    : PD-L1 expression and prognostic impact in glioblastoma. Neuro-Oncol 18: 195-205, 2016.
    OpenUrlAbstract/FREE Full Text
  114. ↵
    1. Reardon DA,
    2. Sampson JH,
    3. Sahebjam S,
    4. Lim M,
    5. Baehring JM,
    6. Vlahovic G,
    7. Cloughesy TF,
    8. Strauss LC,
    9. Latek RR,
    10. Paliwal P,
    11. Harbison CT,
    12. Voloschin AD,
    13. Omuro AMP
    : Safety and activity of nivolumab (nivo) monotherapy and nivo in combination with ipilimumab (ipi) in recurrent glioblastoma (GBM): Updated results from checkmate-143. J Clin Oncol 34, 2016.
  115. ↵
    1. Reardon DA,
    2. Groot JFD,
    3. Colman H,
    4. Jordan JT,
    5. Daras M,
    6. Clarke JL,
    7. Nghiemphu PL,
    8. Gaffey SC,
    9. Peters KB
    : Safety of pembrolizumab in combination with bevacizumab in recurrent glioblastoma (rGBM). J Clin Oncol 34, 2016.
  116. ↵
    1. Sahebjam S,
    2. Johnstone PA,
    3. Forsyth PAJ,
    4. Arrington J,
    5. Vrionis FD,
    6. Etame AB,
    7. Tran ND,
    8. Dalvi PH,
    9. Kim S,
    10. Macaulay RJ,
    11. Chinnaiyan P,
    12. Yu M
    : Safety and antitumor activity of hypofractionated stereotactic irradiation (HFSRT) with pembrolizumab (Pembro) and bevacizumab (Bev) in patients (pts) with recurrent high grade gliomas: Preliminary results from phase I study. J Clin Oncol 34, 2016.
  117. ↵
    1. Blumenthal DT,
    2. Yalon M,
    3. Vainer GW,
    4. Lossos A,
    5. Yust S,
    6. Tzach L,
    7. Cagnano E,
    8. Limon D,
    9. Bokstein F
    : Pembrolizumab: first experience with recurrent primary central nervous system (CNS) tumors. J Neurooncol 129: 453-460, 2016.
    OpenUrl
PreviousNext
Back to top

In this issue

Anticancer Research: 37 (1)
Anticancer Research
Vol. 37, Issue 1
January 2017
  • Table of Contents
  • Table of Contents (PDF)
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Advances in Experimental Targeted Therapy and Immunotherapy for Patients with Glioblastoma Multiforme
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
13 + 1 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Advances in Experimental Targeted Therapy and Immunotherapy for Patients with Glioblastoma Multiforme
JIRI POLIVKA, JIRI POLIVKA, LUBOS HOLUBEC, TEREZA KUBIKOVA, VLADIMIR PRIBAN, ONDREJ HES, KRISTYNA PIVOVARCIKOVA, INKA TRESKOVA
Anticancer Research Jan 2017, 37 (1) 21-33;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Advances in Experimental Targeted Therapy and Immunotherapy for Patients with Glioblastoma Multiforme
JIRI POLIVKA, JIRI POLIVKA, LUBOS HOLUBEC, TEREZA KUBIKOVA, VLADIMIR PRIBAN, ONDREJ HES, KRISTYNA PIVOVARCIKOVA, INKA TRESKOVA
Anticancer Research Jan 2017, 37 (1) 21-33;
Reddit logo Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Perspectives on Novel Therapies for GBM
    • Inhibitors of Growth Factors and Their Receptors, Inhibitors of Intracellular Signaling Pathways
    • Inhibition of Angiogenesis in GBM
    • Immunotherapy of GBM
    • The Role of Immune Checkpoint Inhibitors in Glioblastoma Immunotherapy
    • Summary
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Identification of Tumor Antigens Among the HLA Peptidomes of Glioblastoma Tumors and Plasma
  • Identification of Tumor Antigens Among the HLA Peptidomes of Glioblastoma Tumors and Plasma
  • Investigation of Survivin Gene Polymorphism and Serum Survivin Levels in Patients with Brain Tumors
  • Brainstem Glioblastoma Multiforme in a Patient with NF1
  • Application of Albumin-embedded Magnetic Nanoheaters for Release of Etoposide in Integrated Chemotherapy and Hyperthermia of U87-MG Glioma Cells
  • Expression of FAS-L Differs from Primary to Relapsed Low-grade Gliomas and Predicts Progression-free Survival
  • Systemic Intravenous Adoptive Transfer of Autologous Lymphokine-activated {alpha}{beta} T-Cells Improves Temozolomide-induced Lymphopenia in Patients with Glioma
  • Google Scholar

More in this TOC Section

  • Cytokine-based Cancer Immunotherapy: Challenges and Opportunities for IL-10
  • Proteolytic Enzyme Therapy in Complementary Oncology: A Systematic Review
  • Multimodal Treatment of Primary Advanced Ovarian Cancer
Show more Reviews

Similar Articles

Keywords

  • Glioblastoma multiforme
  • GBM
  • targeted therapy
  • immunotherapy
  • immune checkpoint inhibitors
  • PD1 inhibition
  • CTLA4 inhibition
  • Clinical trials
  • personalized medicine
  • review
Anticancer Research

© 2023 Anticancer Research

Powered by HighWire