Skip to main content
Cancer Reports logoLink to Cancer Reports
. 2019 Sep 4;2(6):e1216. doi: 10.1002/cnr2.1216

Current clinical management of patients with glioblastoma

Stephen Lowe 1, Krishna P Bhat 2,, Adriana Olar 1,3,
PMCID: PMC7941570  PMID: 32721125

Abstract

Background

Glioblastoma (GB) is the most aggressive primary brain tumor, historically resistant to treatment, and with overall fatal outcome.

Recent findings

Recently, several molecular subgroups and rare genetic alterations have been described in GB. In this review article, we will describe the current clinical management of patients with GB in the United States, discuss selected next‐generation molecular‐targeted therapies in GB, and present ongoing clinical trials for patients with GB. This review is intended for clinical and preclinical researchers who conduct work on GB and would like to understand more about the current standard of treatment of GB patients, historical perspectives, current challenges, and ongoing and upcoming clinical trials.

Conclusions

GB is an extremely complex disease, and despite recent progress and advanced therapeutic strategies, the overall patient's prognosis remains dismal. Innovative strategies and integrative ways of approach to disease are urgently needed.

Keywords: chemotherapy, glioblastoma, radiation therapy, surgical therapy

1. INTRODUCTION

Every year, about 15 000 Americans will be diagnosed with a World Health Organization (WHO) grade IV diffuse glioma (or glioblastoma [GB]), the most common adult primary brain tumor. Although the incidence of newly diagnosed GB is low, the proportion of deaths due to GB far exceeds other cancers with a 5‐year survival rate of only 5.6%.1, 2 The current standard of care for GB is comprised of maximal safe surgical resection followed by chemo‐radiation, which prolongs the lifespan of patients by about 12 to 14 months.3 Lower‐grade diffuse gliomas are less aggressive, but most patients eventually progress to GB.2 This standard GB treatment regimen has been in place since 2005, when Stupp et al published the seminal paper on concomitant radiotherapy and temozolomide in GB.4 However, while further work on GB treatment has proceeded in earnest, the overall results have been disappointing.

While the concept of targeting genetic alterations that underlie cancer cells or personalized medicine has met with tremendous success in many cancer types, initial clinical trials of targeted therapy against epidermal growth factor receptor (EGFR), phosphatidylinositol 3‐kinases (PI3K), protease kinase B (AKT), mammalian target of rapamycin (mTOR), and vascular endothelial growth factor (VEGF) have all been equally disappointing showing poor efficacy in multiple studies involving GB.5, 6 Immunotherapy using nivolumab failed to improve overall survival (OS) in a recent phase III clinical trial on recurrent GB.7 However, new therapies are constantly under development and offer hope to patients and clinicians.

In this review, we will sequentially describe the current clinical management of patients with GB in the United States. Following this, we will discuss a selected group of the next generation of molecular‐targeted therapy in GB, and we will describe a selected group of ongoing clinical trials for GB.

2. STANDARD OF CARE THERAPY FOR PATIENTS WITH GB

2.1. Surgery

Surgical therapy has long been the backbone of therapy for GB. The first known surgical operation to resect a glioma was performed by Dr Rickman J. Godlee in London, England in 1884.8 While his patient did not survive (he died nearly 2 months postoperatively from postsurgical meningitis), surgical therapy had come to stay. In addition to the immediate life‐saving effects of removal of intracranial mass, multiple studies show a survival benefit associated with resection of GB, with some showing benefit with as little as 78% extent of resection (EOR) of contrast‐enhancing tumor9; however, other studies note that more is better, and that the best outcomes are achieved with 95% to 98% (or greater) EOR.10 Recent evidence even suggests that supratotal resection (ie, resection of abnormal magnetic resonance imaging (MRI) T2‐fluid attenuated inversion recovery (FLAIR) signal past the traditional contrast‐enhancing tumor border) may be beneficial.11, 12 Gross total resection (GTR) has also been shown to improve outcomes in recurrent GB,13 even in those who had initial subtotal resection.14 In either case, the example is clear—radical surgery with a goal to achieve maximal safe resection improves outcomes.15, 16 Importantly, the desire for maximally aggressive resection must be balanced by the knowledge that iatrogenic language or motor deficit after surgery is associated with decreased survival.17 Many lesions are deep‐seated or within eloquent regions of the brain, making aggressive resection impossible.18 In such cases, technology such as laser interstitial thermal therapy (LITT) offers a minimally invasive method of targeting deep‐seated or eloquent lesions.19 This novel therapy allows for stereotactic placement of a fiber‐optic laser‐emitting catheter into an area of tumor via one of several commercially available systems. Laser ablation is then carried out with the assistance of MR‐thermography in conjunction with either standard MRI, or in some cases, diffusion tensor imaging to ensure ablation of a defined area of tumor without damage to the surrounding deep white matter tracts.20, 21 This technique has shown promise, particularly in patients with eloquent or deep‐seated lesions,19, 22, 23 and offers promise in terms of expanding the number of patients who may yet benefit from cytoreductive surgical therapies.

Surgery by itself is not—and likely cannot be—curative. GB is an infiltrative disease and is known to spread well outside of the contrast‐enhancing portions of the tumor seen on MRI24, 25; therefore, adjunct therapies such as chemotherapy and radiation therapy are paramount.

2.2. Radiotherapy

Radiotherapy (RT) for GB first became popularized in the 1960s, 1970s, and 1980s, when several studies showed that addition of RT to chemotherapy improved survival outcomes compared with chemotherapy alone.26, 27, 28 While, initially, radiation was whole‐brain29, 30; today, radiation volumes are obviously more focused. It was again Stupp's seminal phase III trial in 2005 that solidified the role of combined RT and chemotherapy in the postoperative management of GB.4 The current standard‐of‐care for initial RT after surgical diagnosis is the fractionated delivery of external beam radiation to a dose of 60 Gy in 2 Gy fractions over 6 weeks, typically with an initial radiation plan to 46 Gy in 2 Gy/fraction followed by a boost plan of 14 Gy in 2 Gy/fraction.3, 31, 32

In elderly patients (generally defined as >65‐70 years of age), hypofractionated RT is considered a viable option. The Nordic trial showed poorer outcomes with standard RT as compared with hypofractionated therapy (defined as 34 Gy delivered over 10 fractions).3, 33 A systematic review by Zarnett et al also supported hypofractionated therapy as a first line for elderly patients, with the additional insight that for patients with O 6 ‐methylguanine‐DNA methyltransferase (MGMT) promoter methylation that temozolomide (TMZ) alone may provide more benefit.34 The physiology and significance of this particular genetic marker will be further explained in the Chemotherapy section below.

The role of RT in salvage treatment of GB is less defined but is in use. While the ability to administer further radiation will very much depend on the individual patient's prior radiation dosing and history, as well as tumor location and dosing to critical structures such as the brain stem and optic apparatus, in certain circumstances, it has been shown to be of value. Given that most recurrences are local, salvage stereotactic radiosurgery is in use and has been shown to provide acceptable results in certain instances and has been shown to have an acceptable safety profile.35, 36 A more recent meta‐analysis demonstrated that a combination of stereotactic radiosurgery and TMZ provided a superior survival benefit in locally recurrent GB.37 The use of salvage RT must be tempered by the real possibility of inciting radiation necrosis; however, the clinical outcomes of patients suffering radionecrosis are unclear.38, 39, 40

2.3. Chemotherapy and immunotherapy

2.3.1. Temozolomide

TMZ, an alkylating agent, is the most efficacious chemotherapy for GB to date. Cytosolic conversion of TMZ into 3‐methyl‐(triazen‐1‐yl)imidazole‐4‐carboxamide (MTIC) enables it to translocate to the nucleus and deposit methyl groups on DNA guanine bases at position O6, leading to strand breaks and causing tumor cell apoptosis.41 TMZ is effective in GB given its ability to cross the blood‐brain barrier and manageable side effects such as lymphopenia and gastrointestinal upsetting. Early studies showed that in combination with radiation therapy, TMZ significantly improved median OS as well as the percentage of patients alive at 2 and 5 years in comparison with radiation therapy alone42, 43; however, the median OS was improved marginally from 12.1 to only 14.6 months.42, 43 This regimen has been adopted as the standard of care for newly diagnosed GB. It was later discovered that resistance to TMZ may be partially mediated by MGMT, an enzyme that converts O6‐methyl‐guanine to guanine thus repairing DNA damage mediated by TMZ. Patients whose tumors have MGMT promoter methylation may benefit the most from TMZ.42 Although prolonged adjuvant TMZ with up to 24 cycles has been shown to positively impact survival in retrospective studies,44, 45 a dose‐intensified regimen did not improve OS in another randomized clinical trial.46 The current standard of care in newly diagnosed GB includes administration of TMZ at a dose of 75 mg/m2 per day during the duration of radiation therapy (ie, for 6 weeks) followed by a maintenance schedule of six cycles of TMZ at a dose of 150 to 200 mg/m2 for 5 days of each 28‐day cycle. This, however, is decided based on patient's age, performance status (as the Karnofsky performance score), and the methylation status of the MGMT promoter.3, 4

Invariably, however, the disease recurs. While no established standard of care exists for recurrent GB (lomustine and bevacizumab are the only drugs with FDA approval for recurrent GB), many chemotherapy‐based treatment regimens for recurrent GB use TMZ as part of a therapeutic regimen. Patients with MGMT promoter methylation are most likely to benefit with TMZ retreatment.47 However, it is important to mention that the MGMT promoter methylation status is not routinely assessed for all GB patients (at initial diagnosis or at recurrence), and if assessed, the result might not be taken into account for TMZ treatment decision making in some practices. This could be explained by decreased availability of treatment agents for individual patients, the presence of adverse drug reactions, the presence of comorbidities, and/or patient's preference for treatment. Further study on TMZ generally involves studying the effect of the drug in combination with other chemotherapeutic agents, and multiple clinical trials are ongoing (www.clinicaltrials.gov).

2.3.2. Bevacizumab

High levels of neoangiogenesis are observed in GB.2 Bevacizumab, an antivascular endothelial growth factor (VEGF) monoclonal antibody, was regarded as a promising agent for GB. Although multiple clinical trials in recurrent GB demonstrated impressive radiographic responses, several recent clinical trials in patients with newly diagnosed GB showed modest improvements in progression‐free survival (PFS) but failed to show improvement in OS.48, 49, 50

The RTOG 0825 trial investigated the role of bevacizumab as a first‐line treatment in newly diagnosed GB. The trial reported a median OS of 15.7 months for the treatment arm and a median OS of 16.1 months for the control arm.50 A meta‐analysis reported no difference in bevacizumab's therapeutic effects amongst different dose regimens, and that using the smallest therapeutically efficient dose could possibly reduce any potential adverse effects.51

A second phase III trial for newly diagnosed GB, AVAGlio (NCT00943826), reported nonsignificant slightly larger OS rates with bevacizumab compared with placebo.52, 53 Some reports suggest that patients with proneural GB may benefit most from bevacizumab in combination with standard chemoradiation and adjuvant temozolomide.54 This post hoc analysis would need to be validated in a prospective clinical trial before this approach could be used in clinical practice. Since bevacizumab has not convincingly demonstrated an improvement in OS as a monotherapy, there is an interest in combining bevacizumab with agents known to be effective in high‐grade glioma. One recent combination that is showing promise is the addition of lomustine (CCNU) to bevacizumab in recurrent GB, which in one study yielded 5.1 months of life expectancy.55 Lomustine is a bifunctional alkylating nitrosourea initially used as one part of the PCV regimen (procarbazine, CCNU, and Vincristine).56 Lomustine monotherapy has demonstrated efficacy in recurrent GB.57, 58A phase II clinical trial (EORTC 26101) demonstrated an OS benefit of the combination of bevacizumab with lomustine compared with either lomustine or bevacizumab alone59 and was the first clinical trial to demonstrate a survival benefit of a bevacizumab‐containing regimen; however, this phase II data was not replicated in the phase III version of EORTC 26101.60

Bevacizumab has also been combined with irinotecan, a topoisomerase type I inhibitor that impedes DNA replication by preventing DNA strand cleavage and unwinding.61 A 2016 study showed improved PFS but not OS in patients taking bevacizumab plus irinotecan as compared with temozolomide as up‐front treatment for newly diagnosed MGMT promoter unmethylated GB.62 The phase II RTOG 6205 compared bevacizumab + TMZ with bevacizumab + irinotecan in recurrent GB and found no difference in 6‐month PFS.63

Although adverse side effects are associated with bevacizumab,64 the drug is usually well‐tolerated and may provide a benefit to patients through its ability to reduce cerebral edema and allow for reductions in corticosteroid use.65, 66, 67 This benefit may improve patient quality of life as well as reduce the adverse effects of long‐term steroid use,65, 68 which are known to include hypertension, steroid‐induced myopathy, bone marrow toxicity, and weight gain.69

2.3.3. Carmustine wafers

The use of intratumoral carmustine wafers (1,3‐bis(2‐chloroethyl)‐1‐nitrosourea (BCNU) wafers—biodegradable polymers) as an adjunct to surgical resection was first approved in 1995 in recurrent GB after Brem et al demonstrated a modest improvement in OS from 7.2 months in the carmustine group against 5.4 months in the placebo group.70 It was subsequently approved in primary GB in 2003, 2 years before the advent of the now standard chemoradiotherapy regimen. Subsequent studies have shown that concomitant use of carmustine wafers and temozolomide (TMZ) are safe, while others suggest that concomitant use of carmustine wafers with TMZ are associated with an increased adverse event profile.71, 72 Despite many retrospective studies, no randomized controlled trials exist to support (or refute) the safety and efficacy of concomitant carmustine wafer use with TMZ; their use remains controversial. Moreover, carmustine wafer use may represent exclusion criteria from some clinical trials3, 73 (www.clinicaltrials.gov).

2.4. Alternating electric field therapy

Tumor‐treating fields (TTFs) are alternating currents of low‐intensity electric fields aimed at disrupting growth and initiating apoptosis in mitotically active cells.74 TTFs are delivered by a portable device that delivers the low‐intensity alternating electric fields of intermediate frequency through surface electrodes. This device requires the users to shave their heads for proper lead application and is to be worn at least 18 continuous hours per day. TTFs inhibit tumor growth by interfering with microtubules and cell proliferation75, 76 and have also been shown to inhibit migration and invasion, inhibit tumor‐mediated angiogenesis via down‐regulation of VEGF, and decrease NF‐κβ activity in certain GB cell lines.77 A landmark trial showed superiority of TTF in addition to TMZ compared with TMZ alone for GB maintenance therapy. Authors reported a statistically significant improvement in median PFS in the intent‐to‐treat population of 7.1 months in the TTF plus TMZ group versus 4.0 months (95% CI, 3.3‐5.2 months) in the TMZ alone group, and a statistically significant improvement in median OS in the TMZ plus TTF group of 20.5 months versus 15.6 months in the TMZ alone group. The group also found a low rate of significant adverse events relating to the TTF device itself (2% of patients developing severe skin reactions beneath the transducer arrays) and found a relatively high (75%) adherence to therapy.78 A more recent phase III trial (EF‐14) confirmed these findings by showing PFS and OS benefits for patients treated with TTF and TMZ in newly diagnosed GB.79 While TTFs were not found to be superior to chemotherapy in recurrent GB by one group,80 later on, the PRiDe study (a registry of reported recurrent GB cases) showed improved survival in recurrent GB.81 Current treatment guidelines incorporate TTF both for newly diagnosed and recurrent GB.3

However, TTF therapy is not without drawbacks. Some authors have noted that TTF therapy may not be cost effective82 and the necessity to shave one's head and wear the device for at least 18 hours per day may render compliance an issue; however, initial observations suggest overall high rates of compliance with the device.83

3. NEW FRONTIERS IN GB THERAPY

The next frontier of GB therapy is molecular targeted therapy and immunotherapy. An example of an early attempt at this therapy involved targeting the receptor tyrosine kinase (RTK) pathways. RTKs are cell membrane proteins that bind growth factors, cytokines, or hormones and regulate cell growth through phosphorylation (the transfer of the phosphate of ATP to tyrosine residues on protein substrates) that leads to the initiation of cellular signal transduction pathways. Mutations in RTKs commonly cause receptor autophosphorylation and activation of a series of signaling pathways that lead to uncontrolled cell proliferation.84 RTK pathways are activated in approximately 90% of all GB including EGFR, fibroblast growth factor receptor (FGFR), and platelet‐derived growth factor receptor alpha (PDGFRA),85 but multiple attempts to improve patient outcomes by targeting these RTKs have not been effective. Despite increased EGFR alterations, EGFR tyrosine kinase inhibitors (erlotinib, geftinib) have proven ineffective in newly diagnosed and recurrent GB86, 87, 88, 89 and the EGFR pathway seems to remain active despite efficient receptor blockade.90, 91, 92 Despite these early failures, other molecular targets offer promise. In this section, we will discuss selected molecular targeted therapies that are currently under investigation as adjuvant therapies in GB. Table 1 illustrates the active phase III trials in the United States at the time of the writing of this manuscript.

Table 1.

Active phase III trials

Trial Name (Acronym) Trial ID Phase Design Inclusion Criteria Arms Primary Outcome Number of Patients Results Expected Status Drug Name (Mechanism)
VAL‐083 Phase 3 Study in Temozolomide‐Avastin (Bevacizumab) Recurrent GB (STAR‐3) NCT03149575 III Randomized, open‐label, parallel assignment Recurrence of GB on bevacizumab after SOC VAL‐083 (Dianhydrogalactitol) vs Physician's Choice Salvage Therapy (TMZ, Lomustine, or Carboplatin) OS 180 2019 Active, not recruiting VAL‐083 (Alkylating Agent)
Intraoperative Radiotherapy in Newly Diagnosed GB (INTRAGO‐II) NCT02685605 III Prospective, randomized, 2‐arm open‐label Newly‐suspected GB, age 18‐80 y, felt to be amenable to GTR, KPS ≥60 Surgery+interoperative radiotherapy (20‐30 gy) + Stupp vs Sugery+Stupp Median PFS at 24 mo 314 2021 Recruiting
Safety and Efficacy Study of Trans Sodium Crocetinate (TSC) in Newly Diagnosed GB Biopsy‐Only Subjects (INTACT) NCT03393000 III Open‐label, randomized‐controlled Age 18‐70 y, KPS ≥60, biopsy‐only GB, no previous chemo/RT SOC vs SOC + Trans‐Sodium Crocetinate OS 22 2022 Active, not recruiting Radiosensitizing Agent
A Study of ABT‐414 in Subjects With Newly Diagnosed GB With Epidermal Growth Factor Receptor (EGFR) Amplification (INTELLANCE1) NCT02573324 III Randomized, parallel‐assignment, quadruple‐blinded Age 18‐99 y, KPS ≥70, Confirmed EGFR amplification TMZ + Radiation+Placebo vs TMZ + Radiation+ABT‐414 OS 640 2021 Recruiting ABT‐414 (EGFR inhibitor)
Temozolomide With or Without Veliparib in Treating Patients With Newly Diagnosed GB NCT02152982 II/III Randomized, parallel‐assignment, double‐blinded Age ≥18 y, MGMT promoter Methylation present, has completed SOC, but no other adjuvant treatment TMZ + Placebo vs TMZ + Veliparib OS 440 2022 Active, not recruiting Veliparib (PARP inhibitor)
An Investigational Immuno‐therapy Study of Temozolomide Plus Radiation Therapy With Nivolumab or Placebo, for Newly Diagnosed Patients With GB NCT02667587 III Randomized, parallel‐assignment, triple‐blind Age ≥18 y, KPS ≥70, MGMT promoter Methylation, Newly Diagnosed GB Nivolumab+TMZ + RT vs Placebo + TMZ + RT OS, PFS 693 2023 Active, not recruiting Nivolumab (PD‐1 inhibitor)
An Investigational Immuno‐therapy Study of Nivolumab Compared to Temozolomide, Each Given With Radiation Therapy, for Newly‐diagnosed Patients With GB (CheckMate 498) NCT02617589 III Randomized, parallel‐assignment, open‐label Age ≥18 y, Newly‐Diagnosed GB, MGMT promoter Unmethylated, KPS ≥70 Nivolumab + RT vs TMZ + RT OS 550 2019 Recruiting Nivolumab (PD‐1 inhibitor)
The Toca 5 Trial: Toca 511 & Toca FC Versus Standard of Care in Patients With Recurrent High Grade Glioma (Toca5) NCT02414165 II/III Randomized, parallel‐assignment, open‐label Age 18‐75 y, recurrent GB or AA after first‐line therapy Surgery + Toca 511 + Toca FC vs Surgery + TMZ OR Lomustine OR Bevacizumab OS 403 2023 Active, not recruiting Flucytosine (The Toca 511 retroviral vector converts 5 fluorocytosine to 5‐FU)
A Study of the Effectiveness and Safety of Nivolumab Compared to Bevacizumab and of Nivolumab With or Without Ipilimumab in GB Patients (CheckMate 143) NCT02017717 III Randomized, parallel assignment, open label Newly diagnosed MGMT promoter unmethylated or recurrent GB, KPS ≥70 Nivolumab vs Nivolumab + Ipilimumab vs Bevacizumab OS 626 2019 Active, not recruiting Nivolumab (PD‐1 inhibitor)
Standard Chemotherapy vs Chemotherapy Guided by Cancer Stem Cell Test in Recurrent GB (CSCRGBM) NCT03632135 III Randomized, parallel‐assignment, quadruple‐blinded Age >18 y, recurrent GB Physician Choice Treatment vs Chemotherapy per drug assay ChemoID test OS 300 2022 Recruiting The ChemoID drug response assay reports a prioritized list of effective and ineffective chemotherapies. The test is designed to target cancer stem cells to mitigate tumor relapse.

Abbreviations: AA, anaplastic astrocytoma; AO, anaplastic oligodendroglioma; EGFR, epidermal growth factor receptor; GB, glioblastoma; GTR, gross total resection; KPS, Karnofsky Performance Status; LE, life expectancy; MGMT, O6‐methylguanine‐DNA methyltransferase; OS=overall survival; PARP, poly‐ADP ribose polymerase; PD‐1, programmed‐death 1; PFS=progression‐free survival; RT, radiotherapy; SOC=standard of care; TMZ, temozolomide; 5‐FU, 5‐fluorouracil.

FGFR‐TACC fusions have oncogenic properties,93 and in GB, they are known to portend poorer prognosis, shorter time to progression, and radioresistance.94 These alterations are seen in isocitrate dehydrogenase (IDH)–wild–type gliomas, already known for their more aggressive behavior compared with their IDH‐mutated cousins.95 An FGFR inhibitor phase II trial for recurrent GB (BGJ398) (NCT01975701) is currently awaiting results. BGJ398 is an oral drug, with selective, ATP‐competitive pan‐FGFR kinase inhibitor activity in FGFR‐altered tumors.96 Despite these encouraging advances, this alteration is seen in only approximately 3% of GB, making the application of FGFR inhibitors likely narrow in scope; however, it does offer promise for patients with recurrent IDH‐wild–type GB with few treatment options.93, 95

IDH is a family of mitochondrial enzymes involved in the tricarboxylic acid cycle with roles in converting isocitrate to alpha‐ketoglutarate. IDH1 or IDH2 mutations have been described in diffuse glioma and have been associated with an improved prognosis. IDH mutations cause an accumulation of 2‐hydroxyglutarate which is a competitive inhibitor of a set of enzymes (alpha‐ketoglutarate‐dependent dioxygenases), inhibition that causes DNA and histone methylation and ultimately triggers tumorigenesis.97 IDH‐mutant GB is less common than IDH‐wild–type GB and is seen in younger patients, commonly in those who had a previous lower‐grade diffuse glioma that progressed to GB.2 The most common mutation is IDH1 p.R132H 2. An IDH1–R132H mutant‐specific inhibitor (AGI‐5198) has been developed and tested on cell lines and glioma xenografts.98 IDH1 inhibitors AG‐120 and AG‐881 and the IDH2 inhibitor AG‐221, developed by Agios Pharmaceuticals,99 are now in phase 1 clinical trials for IDH1‐mutated solid tumors (AG‐120) including glioma and results are expected soon.

Poly‐ADP ribose polymerases (PARP) are a family of enzymes involved in genomic stability, DNA repair, and antiapoptosis. PARP are often overexpressed in GB and may represent a mechanism of secondary treatment resistance.100 PARP inhibition is thought to inhibit the antiapoptotic action of PARP and in in vitro models has been shown to both cross the blood‐brain barrier and reactivate apoptotic signaling in GB.100 Furthermore, PARP inhibitors have been shown in in vitro models to act synergistically with TMZ in IDH‐mutated tumors, opening promise for new treatment paradigms in GB with this genetic profile.101 Veliparib (ABT‐888) is a PARP inhibitor that interferes with DNA repair that has been shown to potentiate the effects of other treatments including TMZ on syngeneic and xenograft models.102 It has also been shown to decrease PARP levels synergistically with radiation in patient‐derived cell lines and xenografts.103 Phase I/II trial data is available for Veliparib,104 but the phase II/III trial is still ongoing (NCT02152982), and is expected to be completed in 2022. Another PARP inhibitor currently under clinical trial investigation is Olaparib (Table 2, www.clinicaltrials.gov).

Table 2.

Selected active PARP/CTLA‐4/PD‐1/PD‐L1 inhibitor trials

Trial Name Trial ID Phase Design Criteria Arms Primary Outcome Number of Patients Results Expected Status Drug Name (Mechanism)
Olaparib in Treating Patients With Advanced Glioma, Cholangiocarcinoma, or Solid Tumors With IDH1 or IDH2 Mutations NCT03212274 II Single group, open label Age ≥18 y, Biopsy‐Confirmed IDH1/2 mutation, Recurrent Glioma, Must have enhancing component, Must have LE >16 wk Olaparib (GB OR Cholangiocarcinoma OR Other Solid Neoplasm) Overall response rates in recurrent IDH 1/2 mutant tumors 145 2019 Recruiting Olaparib (PARP inhibitor)
Temozolomide With or Without Veliparib in Treating Patients With Newly Diagnosed Glioblastoma Multiforme NCT02152982 II/III Shown in Table 1
Tremelimumab and Durvalumab in Combination or Alone in Treating Patients With Recurrent Malignant Glioma NCT02794883 II Randomized, parallel‐assignment, open‐label Age ≥18 y, Grade III or IV with progression after >12 wk from SOC, surgical candidate, Life expectancy >12 wk Durvalumab OR Tremelimumab OR Durvalumab + Tremelimumab T‐cell immunologic changes in peripheral blood 36 2020 Recruiting Durvalumab+Tremelimumab (anti‐PD‐L1, anti‐CTLA‐4)
Radiation Therapy With Temozolomide and Pembrolizumab in Treating Patients With Newly Diagnosed Glioblastoma NCT02530502 I Single group, open label Age ≥18 y, GB, No prior adjuvant treatment, KPS ≥70 RT + TMZ + Pembrolizumab Dose‐limiting toxicity of RT + TMZ + Pembrolizumab 4 2020 Active, not recruiting Pembrolizumab (anti‐PD‐1)
Avelumab With Hypofractionated Radiation Therapy in Adults With Isocitrate Dehydrogenase (IDH) Mutant Glioblastoma NCT02968940 II Single group, open label Age ≥18 y, IDH 1/2 Mutation, Grade II/III tumor prior to TMZ or PCV with GB after TMZ or PCV Avelumab+Hypofractionated RT Safety + PFS 43 2020 Active, not recruiting Avelumab (anti‐PD‐L1)
Cediranib Maleate and Olaparib Compared to Bevacizumab in Treating Patients With Recurrent Glioblastoma NCT02974621 II Randomized, parallel assignment, open label Age ≥18 y, First or second recurrent GB, 12 wk from RT, KPS ≥60, at least 3 mo LE Olaparib + Cediranib maleate vs Bevacizumab PFS at 6 mo 70 2019 Active, not recruiting Olaparib + Cediranib maleate (PARP inhibitor + VEGFR inhibitor)
Trial of Combination TTF (Optune), Nivolumab Plus/Minus Ipilimumab for Bevacizumab‐naive, Recurrent GB NCT03430791 II Nonrandomized, parallel‐assignment, open‐label Age ≥18 y, Recurent supratentorial IDH‐wild type GB, KPS ≥60 TTF + Nivolumab vs TTF + Nivolumab + Ipilimumab Objective response rate according to modified iRANO criteria (at 4 mo) 60 2021 Recruiting Ipilimumab (anti‐CTLA‐4)
A Study of the Effectiveness and Safety of Nivolumab Compared to Bevacizumab and of Nivolumab With or Without Ipilimumab in Glioblastoma Patients NCT02017717 III Shown in Table 1 Active, not recruiting
Nivolumab, Ipilimumab, and Short‐course Radiotherapy in Adults With Newly Diagnosed, MGMT Unmethylated Glioblastoma NCT03367715 II Single group, open label Age 18‐100 y, MGMT promoter unmathylated GB, maximum tumor diameter of 6.6 cm or less, KPS ≥60 Nivolumab + Ipilimumab + Short‐course RT 1‐y OS 24 2020 Recruiting Nivolumab+Ipilimumab (anti‐PD‐1, anti‐CTLA‐4)
Ipilimumab and/or Nivolumab in Combination With Temozolomide in Treating Patients With Newly Diagnosed Glioblastoma or Gliosarcoma NCT02311920 I Randomized, parallel‐assignment, open‐label Age ≥18 y, GB with GTR s/p SOC, KPS ≥70 TMZ + Ipilimumab vs TMZ + Nivolumab vs TMZ + Nivolumab+Ipilimumab Immune‐related dose‐limiting toxicities 32 N/A Active, not recruiting Nivolumab+Ipilimumab (anti‐PD‐1, anti‐CTLA‐4)
Biomarker‐Driven Therapy Using Immune Activators With Nivolumab in Patients With First Recurrence of Glioblastoma NCT03707457 I Nonrandomized, parallel‐assignment, open‐label Age ≥18 y, First Recurent GB, KPS ≥60 Nivolumab + MK‐4166 vs Nivolumab + INCB024360 vs Nivolumab + Ipilimumab Drug toxicity 30 2024 Recruiting Nivolumab, MK‐4166, Ipilimumab, INCB024360 (anti‐PD‐1, anti‐GITR, anti‐CTLA‐4, IDO1 inhibitor)
Neoantigen‐based Personalized Vaccine Combined With Immune Checkpoint Blockade Therapy in Patients With Newly Diagnosed, Unmethylated Glioblastoma NCT03422094 I Nonrandomized, sequencial‐assignment, open‐label Age ≥18 y, Newly diagnosed MGMT promoter unmethylated GB, KPS ≥60 NeoVax + Nivolumab (start at progression) vs NeoVax + Nivolumab (start with Cycle 2) vs NeoVax + Nivolumab (start with Cycle 1) vs NeoVax + Ipilimumab + Nivolumab (start with Cycle 3) vs NeoVax + Ipilimumab + Nivolumab (days 1 and 15, each cycle) Safety and tolerability of regimen 30 2023 Recruiting NeoVax, Nivolumab, Ipilimumab (Synthetic long peptides plus poly‐ICLC, anti‐PD‐1, anti‐CTLA‐4)
Cytokine Microdialysis for Real‐Time Immune Monitoring in Glioblastoma Patients Undergoing Checkpoint Blockade NCT03493932 I Single group, open label Age ≥18 y, Recurent GB amenable to surgical resection Nivolumab+BMS‐986016 % of patients that have a rise in interferon gamma levels within the tumor microenvironment 15 2021 Recruiting Nivolumab+BMS‐986016 (anti‐PD‐1, anti‐Lymphocyte Activation Gene‐3)
Tremelimumab and Durvalumab in Combination or Alone in Treating Patients With Recurrent Malignant Glioma NCT02794883 II Randomized, parallel‐assignment, open‐label Age ≥18 y, Recurent anaplastic glioma or GB, KPS ≥70 Durvalumab vs Tremelimumab+Durvalumab T‐cell (immunologic) changes in blood 36 2020 Active, not recruiting Durvalumab, Tremelimumab (anti‐PD‐L1, anti‐CTLA‐4)

Abbreviations: AA, anaplastic astrocytoma; AO, anaplastic oligodendroglioma; CTLA‐4, cytotoxic T lymphocyte associated antigen‐4; GITR, glucocorticoid‐induced TNFR‐related protein; GB, glioblastoma; GTR, gross total resection; IDO1, Indoleamine‐pyrrole 2;3‐dioxygenase; KPS, Karnofsky Performance Status; LE, life expectancy; MGMT, O6‐methylguanine‐DNA methyltransferase; N/A, not available; OS, overall survival; PARP, poly‐ADP ribose polymerase; PD‐1, programmed‐death 1; PD‐L1, programmed‐death 1 ligant; PFS, progression‐free survival; RT, radiotherapy; SOC, standard of care; TMZ, temozolomide; TTF, tumor treating fields (Optune portable device); VEGFR, vascular endothelial growth factor receptor; 5‐FU, 5‐fluorouracil.

Immunomodulatory therapies have recently been very successful in several solid tumors. Two important pathways under investigation in GB are inhibition of cytotoxic T lymphocyte associated antigen‐4 (CTLA‐4) and programmed‐death 1 (PD‐1) – programmed‐death 1 ligand (PD‐L1) interaction blockade, and as such, will be discussed here.105

Briefly, the GB tumor cells release antigens that are taken up by the antigen‐presenting cells (APCs) in the GB microenvironment (microglia and macrophages). The APCs present the tumor antigens to T cells in lymph nodes with subsequent T‐cell activation. Two signal interactions are required for T‐cell activation. First, the T‐cell receptor (TCR) on the surface of T cells in the presence of cytokines recognizes antigens bound to the major histocompatibility complex (MHC) molecule on APCs (signal 1). Then, there is a costimulatory interaction between CD28 on the T‐cell surface and CD80/CD86 (B7‐1/B7‐2) on the APC (signal 2). Cytotoxic T lymphocyte‐associated antigen (CTLA‐4) is a negative regulator of the T‐cell activation or a coinhibitory checkpoint. CTLA‐4 translates to the surface of activated T cells and subsequently binds with higher affinity than CD28 to CD80/CD86 on the surface of the APC inhibiting the T‐cell. Anti‐CTLA‐4 antibodies like Ipilimumab are used to block the CTLA‐4 – B7 interaction, prolong the T‐cell response and potentiate the anti‐tumor T‐cell response.106 Ipilimumab has been studied in combination with bevacizumab and was shown to be well‐tolerated,107 and several clinical trials studying ipilimumab in GB are ongoing (Table 2, www.clinicaltrials.gov). In a similar fashion, PD‐L1 on tumor and APCs interacts with PD‐1 on T cells resulting in a coinhibitory signal and T‐cell suppression. Blocking antibodies (ie, nivolumab and pembrolizumab) block the PD‐1–PD‐L1 interaction and activate T cells.105, 106 It is thought that high expression of PD‐L1 on GB cells alters the tumor microenvironment via suppression of the antitumor immune response.108 Anti‐PD‐1/PD‐L1 agents have been successful in solid tumors,109, 110, 111 and several clinical trials studying these agents are ongoing (Table 2, www.clinicaltrials.gov). Interestingly, the expression of PD‐L1 in glioma is associated with PTEN loss and poorer survival.112, 113 Simultaneous blockade of CTLA‐4 and PD‐1‐PD‐L1 may be synergistic and can result in improved response rates.114 NCT02794883 is a phase II trial investigating individual and concurrent use of tremelimumab and durvalumab (anti‐CTLA‐4 and anti‐PD‐1, respectively) in GB (Table 2, www.clinicaltrials.gov).

Finally, dendritic cell (DC) vaccines are an emerging and exciting frontier in GB therapy. DC vaccines are developed from circulating monocytes sensitized to tumor lysates, preferably from the entire tumor. These DCs are matured via cytokines and then reinjected into the patient.115 The timing of this injection in relation to the current standard of care is debated. In a meta‐analysis of 21 studies and 403 patients, median OS was reported as 71.6 to 138 weeks in recurrent and 65 to 230.4 weeks in newly diagnosed GB compared with the control median OS of 58.4 weeks.116 Moreover, patients with mesenchymal GB on DC vaccine therapy had significantly increased survival compared to control patients and vaccine‐treated mesenchymal GBs had an increased number of tumor infiltrating lymphocytes.117 Many clinical trials of DC vaccines are active, and a recent phase III clinical trial of DCVax‐L in GB (NCT00045968) demonstrated a likely increase in survival in the intention‐to‐treat group, particularly in patients with MGMT promoter methylation.118 This is currently an active area of research and has shown early promise in GB treatment. Active DC vaccine trials are listed in Table 3.

Table 3.

Selected dendritic cell vaccine trials

Trial Name (Acronym) Trial ID Phase Design Criteria Arms Primary Outcome Number of Patients Results Expected Status
Phase I Study of a Dendritic Cell Vaccine for Patients With Either Newly Diagnosed or Recurrent Glioblastoma NCT02010606 I Non‐randomized, open‐label, parallel assignment Age ≥18 y, Newly diagnosed GB, KPS ≥70 DC Vaccine + SOC vs DC Vaccine ± bevacizumab Safety, tolerability, # of adverse events 40 2021 Active, Not Recruiting
Vaccine Therapy for the Treatment of Newly Diagnosed Glioblastoma Multiforme (ATTAC‐II) NCT02465268 II Randomized, parallel assignment, single Age ≥18 y, Newly diagnosed supratentorial GB, KPS ≥70 pp65‐shLAMP DC + GM‐CSF + Td vs pp65‐flLAMP DC + GM‐CSF + Td vs Placebo Change in median OS 120 2024 Recruiting
Dendritic Cell Vaccine for Patients With Brain Tumors NCT01204684 II Randomized, parallel assignment, open‐label Age 18‐70 y, Recurrent AA, AO, GB, KPS > 60 DC + Placebo vs DC + resiquimod vs DC + adjuvant polyICLC Most effective combination of DC vaccine components 60 2021 Active, Not Recruiting
Autologous Dendritic Cells Loaded With Autologous Tumor Associated Antigens for Treatment of Newly Diagnosed Glioblastoma NCT03400917 II Single‐arm, open label Age 18‐70 y, KPS 70‐100, Successful establishment of autologous stem cell line, Plans to begin adjuvant therapy DC Vaccine (AV‐GBM‐1) + GM‐CSF OS 55 2023 Recruiting

Abbreviations: AA, anaplastic astrocytoma; AO, anaplastic oligodendroglioma; DC, dendritic cells; GB, glioblastoma; GM‐CSF, granulocyte‐macrophage colony stimulating factor; GTR, gross total resection; KPS, Karnofsky Performance Status; LE, life expectancy; MGMT, O6‐methylguanine‐DNA methyltransferase; OS, overall survival; PARP, poly‐ADP ribose polymerase; PD‐1, programmed‐death 1; PFS, progression‐free survival; RT, radiotherapy; SOC, standard of care; TMZ, temozolomide.

4. CONCLUSION

GB is an extremely aggressive and complex disease. Despite recent advances, the clinical standard‐of‐care management, including surgery, temozolomide, RT, bevacizumab, and carmustine wafers, is currently limited, and the overall prognosis remains dismal. New therapies, such as DC vaccines, PARP inhibitors, anti‐PD/PD‐L1 agents, and CTLA‐4 inhibitors, are a small sample of the exciting potential new therapies on the horizon. Thankfully, clinical trial activity in this disease remains active and many new ideas and therapies remain to be studied and further developed.

AUTHORS' CONTRIBUTIONS

All authors had full access to the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Conceptualization, K.P.B., A.O.; Methodology, S.L., A.O.; Investigation, S.L.; Formal Analysis, S.L.; Resources, S.L.; Writing ‐ Original Draft, S.L.; Writing ‐ Review & Editing, S.L., K.P.B., A.O.; Visualization, S.L.; Supervision, A.O.; Funding Acquisition, No funding.

CONFLICT OF INTERESTS

The authors have no conflict of interest to report.

Lowe S, Bhat KP, Olar A. Current clinical management of patients with glioblastoma. Cancer Reports. 2019;2:e1216. 10.1002/cnr2.1216

Contributor Information

Krishna P. Bhat, Email: kbhat@mdanderson.org.

Adriana Olar, Email: adriana_olar@yahoo.com.

REFERENCES

  • 1. Ostrom QT, Gittleman H, Truitt G, Boscia A, Kruchko C, Barnholtz‐Sloan JS. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2011‐2015. Neuro Oncol. 2018;20(suppl_4):iv1‐iv86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Louis DN, Ohgaki H, Wiestler OD, et al. WHO classification of tumours of the central nervous system. 4th revised ed. Lyon (France): IARC; 2016. [Google Scholar]
  • 3. NCC N . Clinical practice guidelines in oncology. Central Nervous System Cancers. Version 2. 2018. http://www.nccn.org. Updated November 26,2018.
  • 4. Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987‐996. [DOI] [PubMed] [Google Scholar]
  • 5. Sturm D, Bender S, Jones DT, et al. Paediatric and adult glioblastoma: multiform (epi)genomic culprits emerge. Nat Rev Cancer. 2014;14(2):92‐107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Lau D, Magill ST, Aghi MK. Molecularly targeted therapies for recurrent glioblastoma: current and future targets. Neurosurg Focus. 2014;37(6):E15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Filley AC, Henriquez M, Dey M. Recurrent glioma clinical trial, CheckMate‐143: the game is not over yet. Oncotarget. 2017;8(53):91779‐91794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Kirkpatrick DB. The first primary brain‐tumor operation. J Neurosurg. 1984;61(5):809‐813. [DOI] [PubMed] [Google Scholar]
  • 9. Sanai N, Polley MY, McDermott MW, Parsa AT, Berger MS. An extent of resection threshold for newly diagnosed glioblastomas. J Neurosurg. 2011;115(1):3‐8. [DOI] [PubMed] [Google Scholar]
  • 10. Lacroix M, Abi‐Said D, Fourney DR, et al. A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival. J Neurosurg. 2001;95(2):190‐198. [DOI] [PubMed] [Google Scholar]
  • 11. Eyupoglu IY, Hore N, Merkel A, Buslei R, Buchfelder M, Savaskan N. Supra‐complete surgery via dual intraoperative visualization approach (DiVA) prolongs patient survival in glioblastoma. Oncotarget. 2016;7(18):25755‐25768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Li YM, Suki D, Hess K, Sawaya R. The influence of maximum safe resection of glioblastoma on survival in 1229 patients: can we do better than gross‐total resection? J Neurosurg. 2016;124(4):977‐988. [DOI] [PubMed] [Google Scholar]
  • 13. Suchorska B, Weller M, Tabatabai G, et al. Complete resection of contrast‐enhancing tumor volume is associated with improved survival in recurrent glioblastoma‐results from the DIRECTOR trial. Neuro Oncol. 2016;18(4):549‐556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Hervey‐Jumper SL, Berger MS. Reoperation for recurrent high‐grade glioma: a current perspective of the literature. Neurosurgery. 2014;75(5):491‐499. discussion 8‐9 [DOI] [PubMed] [Google Scholar]
  • 15. Khan MB, Chakraborty S, Boockvar JA. Gross total resection of glioblastoma improves overall survival and progression‐free survival compared to subtotal resection or biopsy alone. Neurosurgery. 2016;79(6):N12‐N13. [DOI] [PubMed] [Google Scholar]
  • 16. Chaichana KL, Cabrera‐Aldana EE, Jusue‐Torres I, et al. When gross total resection of a glioblastoma is possible, how much resection should be achieved? World Neurosurg. 2014;82(1‐2):e257‐e265. [DOI] [PubMed] [Google Scholar]
  • 17. McGirt MJ, Mukherjee D, Chaichana KL, Than KD, Weingart JD, Quinones‐Hinojosa A. Association of surgically acquired motor and language deficits on overall survival after resection of glioblastoma multiforme. Neurosurgery. 2009;65(3):463‐469. discussion 9‐70 [DOI] [PubMed] [Google Scholar]
  • 18. Fazeny‐Dorner B, Wenzel C, Veitl M, et al. Survival and prognostic factors of patients with unresectable glioblastoma multiforme. Anticancer Drugs. 2003;14(4):305‐312. [DOI] [PubMed] [Google Scholar]
  • 19. Thomas JG, Rao G, Kew Y, Prabhu SS. Laser interstitial thermal therapy for newly diagnosed and recurrent glioblastoma. Neurosurg Focus. 2016;41(4):E12. [DOI] [PubMed] [Google Scholar]
  • 20. Sharma M, Habboub G, Behbahani M, Silva D, Barnett GH, Mohammadi AM. Thermal injury to corticospinal tracts and postoperative motor deficits after laser interstitial thermal therapy. Neurosurg Focus. 2016;41(4):E6. [DOI] [PubMed] [Google Scholar]
  • 21. Patel NV, Mian M, Stafford RJ, et al. Laser interstitial thermal therapy technology, physics of magnetic resonance imaging thermometry, and technical considerations for proper catheter placement during magnetic resonance imaging‐guided laser interstitial thermal therapy. Neurosurgery. 2016;79(Suppl 1):S8‐S16. [DOI] [PubMed] [Google Scholar]
  • 22. Barnett GH, Voigt JD, Alhuwalia MS. A systematic review and meta‐analysis of studies examining the use of brain laser interstitial thermal therapy versus craniotomy for the treatment of high‐grade tumors in or near areas of eloquence: an examination of the extent of resection and major complication rates associated with each type of surgery. Stereotact Funct Neurosurg. 2016;94(3):164‐173. [DOI] [PubMed] [Google Scholar]
  • 23. Silva D, Sharma M, Juthani R, Meola A, Barnett GH. Magnetic resonance thermometry and laser interstitial thermal therapy for brain tumors. Neurosurg Clin N Am. 2017;28(4):525‐533. [DOI] [PubMed] [Google Scholar]
  • 24. Kelly PJ, Daumas‐Duport C, Scheithauer BW, Kall BA, Kispert DB. Stereotactic histologic correlations of computed tomography‐ and magnetic resonance imaging‐defined abnormalities in patients with glial neoplasms. Mayo Clin Proc. 1987;62(6):450‐459. [DOI] [PubMed] [Google Scholar]
  • 25. Olar A, Aldape KD. Using the molecular classification of glioblastoma to inform personalized treatment. J Pathol. 2014;232(2):165‐177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Walker MD, Alexander E Jr, Hunt WE, et al. Evaluation of BCNU and/or radiotherapy in the treatment of anaplastic gliomas: a cooperative clinical trial. J Neurosurg. 1978;49(3):333‐343. [DOI] [PubMed] [Google Scholar]
  • 27. Walker MD, Green SB, Byar DP, et al. Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med. 1980;303(23):1323‐1329. [DOI] [PubMed] [Google Scholar]
  • 28. Walker MD, Strike TA, Sheline GE. An analysis of dose‐effect relationship in the radiotherapy of malignant gliomas. Int J Radiat Oncol Biol Phys. 1979;5(10):1725‐1731. [DOI] [PubMed] [Google Scholar]
  • 29. Salazar OM, Rubin P. The spread of glioblastoma multiforme as a determining factor in the radiation treated volume. Int J Radiat Oncol Biol Phys. 1976;1(7‐8):627‐637. [DOI] [PubMed] [Google Scholar]
  • 30. Hochberg FH, Pruitt A. Assumptions in the radiotherapy of glioblastoma. Neurology. 1980;30(9):907‐911. [DOI] [PubMed] [Google Scholar]
  • 31. Sulman EP, Ismaila N, Armstrong TS, et al. Radiation therapy for glioblastoma: American society of clinical oncology clinical practice guideline endorsement of the american society for radiation oncology guideline. J Clin Oncol. 2017;35(3):361‐369. [DOI] [PubMed] [Google Scholar]
  • 32. Cabrera AR, Kirkpatrick JP, Fiveash JB, et al. Radiation therapy for glioblastoma: executive summary of an American society for radiation oncology evidence‐based clinical practice guideline. Pract Radiat Oncol. 2016;6(4):217‐225. [DOI] [PubMed] [Google Scholar]
  • 33. Malmstrom A, Gronberg BH, Marosi C, et al. Temozolomide versus standard 6‐week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, phase 3 trial. Lancet Oncol. 2012;13(9):916‐926. [DOI] [PubMed] [Google Scholar]
  • 34. Zarnett OJ, Sahgal A, Gosio J, et al. Treatment of elderly patients with glioblastoma: a systematic evidence‐based analysis. JAMA Neurol. 2015;72(5):589‐596. [DOI] [PubMed] [Google Scholar]
  • 35. Patel M, Siddiqui F, Jin JY, et al. Salvage reirradiation for recurrent glioblastoma with radiosurgery: radiographic response and improved survival. J Neurooncol. 2009;92(2):185‐191. [DOI] [PubMed] [Google Scholar]
  • 36. Holt DE, Bernard ME, Quan K, et al. Salvage stereotactic radiosurgery for recurrent glioblastoma multiforme with prior radiation therapy. J Cancer Res Ther. 2016;12(4):1243‐1248. [DOI] [PubMed] [Google Scholar]
  • 37. Kim HR, Kim KH, Kong DS, et al. Outcome of salvage treatment for recurrent glioblastoma. J Clin Neurosci off j Neuro Soc Aust. 2015;22(3):468‐473. [DOI] [PubMed] [Google Scholar]
  • 38. Grossman R, Shimony N, Hadelsberg U, et al. Impact of resecting radiation necrosis and pseudoprogression on survival of patients with glioblastoma. World Neurosurg. 2016;89:37‐41. [DOI] [PubMed] [Google Scholar]
  • 39. Ruben JD, Dally M, Bailey M, Smith R, McLean CA, Fedele P. Cerebral radiation necrosis: incidence, outcomes, and risk factors with emphasis on radiation parameters and chemotherapy. Int J Radiat Oncol Biol Phys. 2006;65(2):499‐508. [DOI] [PubMed] [Google Scholar]
  • 40. Rusthoven KE, Olsen C, Franklin W, et al. Favorable prognosis in patients with high‐grade glioma with radiation necrosis: the University of Colorado reoperation series. Int J Radiat Oncol Biol Phys. 2011;81(1):211‐217. [DOI] [PubMed] [Google Scholar]
  • 41. Wesolowski JR, Rajdev P, Mukherji SK. Temozolomide (Temodar). AJNR Am J Neuroradiol. 2010;31(8):1383‐1384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Stupp R, Hegi ME, Mason WP, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5‐year analysis of the EORTC‐NCIC trial. Lancet Oncol. 2009;10(5):459‐466. [DOI] [PubMed] [Google Scholar]
  • 43. Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352(10):997‐1003. [DOI] [PubMed] [Google Scholar]
  • 44. Malkoun N, Chargari C, Forest F, et al. Prolonged temozolomide for treatment of glioblastoma: preliminary clinical results and prognostic value of p53 overexpression. J Neurooncol. 2012;106(1):127‐133. [DOI] [PubMed] [Google Scholar]
  • 45. Darlix A, Baumann C, Lorgis V, et al. Prolonged administration of adjuvant temozolomide improves survival in adult patients with glioblastoma. Anticancer Res. 2013;33(8):3467‐3474. [PubMed] [Google Scholar]
  • 46. Gilbert MR, Wang M, Aldape KD, et al. Dose‐dense temozolomide for newly diagnosed glioblastoma: a randomized phase III clinical trial. J Clin Oncol. 2013;31(32):4085‐4091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Weller M, Tabatabai G, Kastner B, et al. MGMT promoter methylation is a strong prognostic biomarker for benefit from dose‐intensified temozolomide rechallenge in progressive glioblastoma: the DIRECTOR trial. Clin Cancer Res. 2015;21(9):2057‐2064. [DOI] [PubMed] [Google Scholar]
  • 48. Lai A, Tran A, Nghiemphu PL, et al. Phase II study of bevacizumab plus temozolomide during and after radiation therapy for patients with newly diagnosed glioblastoma multiforme. J Clin Oncol. 2011;29(2):142‐148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Clarke JL, Molinaro AM, Phillips JJ, et al. A single‐institution phase II trial of radiation, temozolomide, erlotinib, and bevacizumab for initial treatment of glioblastoma. Neuro Oncol. 2014;16(7):984‐990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Gilbert MR, Dignam JJ, Armstrong TS, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):699‐708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wong ET, Gautam S, Malchow C, Lun M, Pan E, Brem S. Bevacizumab for recurrent glioblastoma multiforme: a meta‐analysis. J Natl Compr Canc Netw. 2011;9(4):403‐407. [DOI] [PubMed] [Google Scholar]
  • 52. Chinot OL, de La Motte RT, Moore N, et al. AVAglio: Phase 3 trial of bevacizumab plus temozolomide and radiotherapy in newly diagnosed glioblastoma multiforme. Adv Ther. 2011;28(4):334‐340. [DOI] [PubMed] [Google Scholar]
  • 53. Chinot OL, Wick W, Mason W, et al. Bevacizumab plus radiotherapy‐temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370(8):709‐722. [DOI] [PubMed] [Google Scholar]
  • 54. Sandmann T, Bourgon R, Garcia J, et al. 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. 2015;33(25):2735‐2744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Tonder M, Weller M, Eisele G, Roth P. Carboplatin and etoposide in heavily pretreated patients with progressive high‐grade glioma. Chemotherapy. 2014;60(5‐6):375‐378. [DOI] [PubMed] [Google Scholar]
  • 56. Shapiro WR, Young DF. Chemotherapy of malignant glioma with CCNU alone and CCNU combined with vincristine sulfate and procarbazine hydrochloride. Trans Am Neurol Assoc. 1976;101:217‐220. [PubMed] [Google Scholar]
  • 57. Wick W, Puduvalli VK, Chamberlain MC, et al. Phase III study of enzastaurin compared with lomustine in the treatment of recurrent intracranial glioblastoma. J Clin Oncol. 2010;28(7):1168‐1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Batchelor TT, Betensky RA, Esposito JM, et al. Age‐dependent prognostic effects of genetic alterations in glioblastoma. Clin Cancer Res. 2004;10(1 Pt 1):228‐233. [DOI] [PubMed] [Google Scholar]
  • 59. Taal W, Oosterkamp HM, Walenkamp AM, et al. Single‐agent bevacizumab or lomustine versus a combination of bevacizumab plus lomustine in patients with recurrent glioblastoma (BELOB trial): a randomised controlled phase 2 trial. Lancet Oncol. 2014;15(9):943‐953. [DOI] [PubMed] [Google Scholar]
  • 60. Wick W, Gorlia T, Bendszus M, et al. Lomustine and bevacizumab in progressive glioblastoma. N Engl J Med. 2017;377(20):1954‐1963. [DOI] [PubMed] [Google Scholar]
  • 61. Pommier Y, Leo E, Zhang H, Marchand C. DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chem Biol. 2010;17(5):421‐433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Herrlinger U, Schafer N, Steinbach JP, et al. Bevacizumab plus irinotecan versus temozolomide in newly diagnosed o6‐methylguanine‐dna methyltransferase nonmethylated glioblastoma: the randomized GLARIUS trial. J Clin Oncol. 2016;34(14):1611‐1619. [DOI] [PubMed] [Google Scholar]
  • 63. Gilbert MR, Pugh SL, Aldape K, et al. NRG oncology RTOG 0625: a randomized phase II trial of bevacizumab with either irinotecan or dose‐dense temozolomide in recurrent glioblastoma. J Neurooncol. 2017;131(1):193‐199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Weathers SP, Gilbert MR. Advances in treating glioblastoma. F1000Prime Rep. 2014;6:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Vredenburgh JJ, Cloughesy T, Samant M, et al. Corticosteroid use in patients with glioblastoma at first or second relapse treated with bevacizumab in the BRAIN study. Oncologist. 2010;15(12):1329‐1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Norden AD, Young GS, Setayesh K, et al. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70(10):779‐787. [DOI] [PubMed] [Google Scholar]
  • 67. Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27(28):4733‐4740. [DOI] [PubMed] [Google Scholar]
  • 68. Diaz RJ, Ali S, Qadir MG, De La Fuente MI, Ivan ME, Komotar RJ. The role of bevacizumab in the treatment of glioblastoma. J Neurooncol. 2017;133(3):455‐467. [DOI] [PubMed] [Google Scholar]
  • 69. Oray M, Abu Samra K, Ebrahimiadib N, Meese H, Foster CS. Long‐term side effects of glucocorticoids. Expert Opin Drug Saf. 2016;15(4):457‐465. [DOI] [PubMed] [Google Scholar]
  • 70. Brem H, Piantadosi S, Burger PC, et al. Placebo‐controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer‐brain Tumor Treatment Group Lancet. 1995;345(8956):1008‐1012. [DOI] [PubMed] [Google Scholar]
  • 71. De Bonis P, Anile C, Pompucci A, et al. Safety and efficacy of Gliadel wafers for newly diagnosed and recurrent glioblastoma. Acta Neurochir. 2012;154(8):1371‐1378. [DOI] [PubMed] [Google Scholar]
  • 72. Gutenberg A, Lumenta CB, Braunsdorf WE, et al. The combination of carmustine wafers and temozolomide for the treatment of malignant gliomas. A comprehensive review of the rationale and clinical experience. J Neurooncol. 2013;113(2):163‐174. [DOI] [PubMed] [Google Scholar]
  • 73. Urbschat S, Sippl C, Engelhardt J, Kammers K, Oertel J, Ketter R. Importance of biomarkers in glioblastomas patients receiving local BCNU wafer chemotherapy. Mol Cytogenet. 2017;10(1):16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Pless M, Weinberg U. Tumor treating fields: concept, evidence and future. Expert Opin Investig Drugs. 2011;20(8):1099‐1106. [DOI] [PubMed] [Google Scholar]
  • 75. Burri SH, Gondi V, Brown PD, Mehta MP. The evolving role of tumor treating fields in managing glioblastoma: guide for oncologists. Am J Clin Oncol. 2018;41(2):191‐196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Zhu P, Zhu JJ. Tumor treating fields: a novel and effective therapy for glioblastoma: mechanism, efficacy, safety and future perspectives. Chin Clin Oncol. 2017;6(4):41. [DOI] [PubMed] [Google Scholar]
  • 77. Kim EH, Song HS, Yoo SH, Yoon M. Tumor treating fields inhibit glioblastoma cell migration, invasion and angiogenesis. Oncotarget. 2016;7(40):65125‐65136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Stupp R, Taillibert S, Kanner AA, et al. Maintenance therapy with tumor‐treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial. JAMA. 2015;314(23):2535‐2543. [DOI] [PubMed] [Google Scholar]
  • 79. Zhu JJ, Demireva P, Kanner AA, et al. Health‐related quality of life, cognitive screening, and functional status in a randomized phase III trial (EF‐14) of tumor treating fields with temozolomide compared to temozolomide alone in newly diagnosed glioblastoma. J Neurooncol. 2017;135(3):545‐552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Stupp R, Wong ET, Kanner AA, et al. NovoTTF‐100A versus physician's choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality. Eur J Cancer. 2012;48(14):2192‐2202. [DOI] [PubMed] [Google Scholar]
  • 81. Mrugala MM, Engelhard HH, Dinh Tran D, et al. Clinical practice experience with NovoTTF‐100A system for glioblastoma: The Patient Registry Dataset (PRiDe). Semin Oncol. 2014;41(Suppl 6):S4‐S13. [DOI] [PubMed] [Google Scholar]
  • 82. Bernard‐Arnoux F, Lamure M, Ducray F, Aulagner G, Honnorat J, Armoiry X. The cost‐effectiveness of tumor‐treating fields therapy in patients with newly diagnosed glioblastoma. Neuro Oncol. 2016;18(8):1129‐1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Rick J, Chandra A, Aghi MK. Tumor treating fields: a new approach to glioblastoma therapy. J Neurooncol. 2018;137(3):447‐453. [DOI] [PubMed] [Google Scholar]
  • 84. Hubbard SR, Till JH. Protein tyrosine kinase structure and function. Annu Rev Biochem. 2000;69:373‐398. [DOI] [PubMed] [Google Scholar]
  • 85. Brennan CW, Verhaak RG, McKenna A, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462‐477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. van den Bent MJ, Brandes AA, Rampling R, et al. Randomized phase II trial of erlotinib versus temozolomide or carmustine in recurrent glioblastoma: EORTC brain tumor group study 26034. J Clin Oncol. 2009;27(8):1268‐1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Uhm JH, Ballman KV, Wu W, et al. Phase II evaluation of gefitinib in patients with newly diagnosed Grade 4 astrocytoma: Mayo/North Central Cancer Treatment Group Study N0074. Int J Radiat Oncol Biol Phys. 2011;80(2):347‐353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Brown PD, Krishnan S, Sarkaria JN, et al. Phase I/II trial of erlotinib and temozolomide with radiation therapy in the treatment of newly diagnosed glioblastoma multiforme: North Central Cancer Treatment Group Study N0177. J Clin Oncol. 2008;26(34):5603‐5609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Raizer JJ, Abrey LE, Lassman AB, et al. A phase II trial of erlotinib in patients with recurrent malignant gliomas and nonprogressive glioblastoma multiforme postradiation therapy. Neuro Oncol. 2010;12(1):95‐103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Hegi ME, Diserens AC, Bady P, et al. Pathway analysis of glioblastoma tissue after preoperative treatment with the EGFR tyrosine kinase inhibitor gefitinib—a phase II trial. Mol Cancer Ther. 2011;10(6):1102‐1112. [DOI] [PubMed] [Google Scholar]
  • 91. Hasselbalch B, Lassen U, Poulsen HS, Stockhausen MT. Cetuximab insufficiently inhibits glioma cell growth due to persistent EGFR downstream signaling. Cancer Invest. 2010;28(8):775‐787. [DOI] [PubMed] [Google Scholar]
  • 92. Vollmann A, Vornlocher HP, Stempfl T, Brockhoff G, Apfel R, Bogdahn U. Effective silencing of EGFR with RNAi demonstrates non‐EGFR dependent proliferation of glioma cells. Int J Oncol. 2006;28(6):1531‐1542. [PubMed] [Google Scholar]
  • 93. Singh D, Chan JM, Zoppoli P, et al. Transforming fusions of FGFR and TACC genes in human glioblastoma. Science. 2012;337(6099):1231‐1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Sayal KK, Higgins GS, Hammond EM. Uncovering the influence of the FGFR1 pathway on glioblastoma radiosensitivity. Ann Transl Med. 2016;4(24):538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Di Stefano AL, Fucci A, Frattini V, et al. Detection, characterization, and inhibition of FGFR‐TACC fusions in IDH wild‐type glioma. Clin Cancer Res. 2015;21(14):3307‐3317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Guagnano V, Kauffmann A, Wohrle S, et al. FGFR genetic alterations predict for sensitivity to NVP‐BGJ398, a selective pan‐FGFR inhibitor. Cancer Discov. 2012;2(12):1118‐1133. [DOI] [PubMed] [Google Scholar]
  • 97. Olar A, Raghunathan A, Albarracin CT, et al. Absence of IDH1‐R132H mutation predicts rapid progression of nonenhancing diffuse glioma in older adults. Ann Diagn Pathol. 2012;16(3):161‐170. [DOI] [PubMed] [Google Scholar]
  • 98. Rohle D, Popovici‐Muller J, Palaskas N, et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science. 2013;340(6132):626‐630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Agios pharmaceuticals website. http://www.agios.com/pipeline‐idh.php.
  • 100. Karpel‐Massler G, Pareja F, Aime P, et al. PARP inhibition restores extrinsic apoptotic sensitivity in glioblastoma. PLoS ONE. 2014;9(12):e114583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Lu Y, Kwintkiewicz J, Liu Y, et al. Chemosensitivity of IDH1‐mutated gliomas due to an impairment in PARP1‐mediated DNA repair. Cancer Res. 2017;77(7):1709‐1718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Donawho CK, Luo Y, Luo Y, et al. ABT‐888, an orally active poly (ADP‐ribose) polymerase inhibitor that potentiates DNA‐damaging agents in preclinical tumor models. Clin Cancer Res. 2007;13(9):2728‐2737. [DOI] [PubMed] [Google Scholar]
  • 103. Jue TR, Nozue K, Lester AJ, et al. Veliparib in combination with radiotherapy for the treatment of MGMT unmethylated glioblastoma. J Transl Med. 2017;15(1):61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Robins HI, Zhang P, Gilbert MR, et al. A randomized phase I/II study of ABT‐888 in combination with temozolomide in recurrent temozolomide resistant glioblastoma: an NRG oncology RTOG group study. J Neurooncol. 2016;126(2):309‐316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Naidoo J, Page DB, Wolchok JD. Immune modulation for cancer therapy. Br J Cancer. 2014;111(12):2214‐2219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Preusser M, Lim M, Hafler DA, Reardon DA, Sampson JH. Prospects of immune checkpoint modulators in the treatment of glioblastoma. Nat Rev Neurol. 2015;11(9):504‐514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Carter T, Shaw H, Cohn‐Brown D, Chester K, Mulholland P. Ipilimumab and Bevacizumab in Glioblastoma. Clin Oncol (R Coll Radiol). 2016;28(10):622‐626. [DOI] [PubMed] [Google Scholar]
  • 108. Kim JE, Lim M. The role of checkpoints in the treatment of GBM. J Neurooncol. 2015;123(3):413‐423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single‐agent anti‐programmed death‐1 (MDX‐1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28(19):3167‐3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti‐PD‐1 antibody in cancer. N Engl J Med. 2012;366(26):2443‐2454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Robert C, Soria JC, Eggermont AM. Drug of the year: programmed death‐1 receptor/programmed death‐1 ligand‐1 receptor monoclonal antibodies. Eur J Cancer. 2013;49(14):2968‐2971. [DOI] [PubMed] [Google Scholar]
  • 112. Parsa AT, Waldron JS, Panner A, et al. Loss of tumor suppressor PTEN function increases B7‐H1 expression and immunoresistance in glioma. Nat Med. 2007;13(1):84‐88. [DOI] [PubMed] [Google Scholar]
  • 113. Xue S, Song G, Yu J. The prognostic significance of PD‐L1 expression in patients with glioma: a meta‐analysis. Sci Rep. 2017;7(1):4231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Maxwell R, Jackson CM, Lim M. Clinical trials investigating immune checkpoint blockade in glioblastoma. Curr Treat Options Oncol. 2017;18(8):51. [DOI] [PubMed] [Google Scholar]
  • 115. Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12(4):265‐277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Bregy A, Wong TM, Shah AH, Goldberg JM, Komotar RJ. Active immunotherapy using dendritic cells in the treatment of glioblastoma multiforme. Cancer Treat Rev. 2013;39(8):891‐907. [DOI] [PubMed] [Google Scholar]
  • 117. Prins RM, Soto H, Konkankit V, et al. Gene expression profile correlates with T‐cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res. 2011;17(6):1603‐1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Liau LM, Ashkan K, Tran DD, et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J Transl Med. 2018;16(1):142. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cancer Reports are provided here courtesy of Wiley

RESOURCES