Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jan 3.
Published in final edited form as: Expert Opin Investig Drugs. 2023 Nov 6;32(10):921–930. doi: 10.1080/13543784.2023.2267982

Investigational Treatment Strategies in Glioblastoma: Progress Made and Barriers to Success

Thomas A Nelson 1, Jorg Dietrich 1
PMCID: PMC10764117  NIHMSID: NIHMS1952665  PMID: 37796104

Abstract

Introduction:

Glioblastoma, isocitrate dehydrogenase wildtype (IDHwt), remains an incurable disease despite considerable research effort. The current standard of care since 2005 comprises maximal safe resection followed by radiation with concurrent and adjuvant temozolomide; more recently, the addition of tumor treating fields was approved in the newly diagnosed and recurrent disease settings.

Areas Covered:

Searches of PubMed, Cochrane Library, and ClinicalTrials.gov provided a foundation for this review. We first describe early research including carmustine wafers, brachytherapy, anti-angiogenesis, and immune checkpoint inhibition for glioblastoma. Next, we discuss challenges precluding the translation of preclinical successes. This is followed by a description of promising treatments such as chimeric antigen receptor T-cell therapy as well as the recent qualified successes of cancer vaccinations. Non-immunotherapy trials are also highlighted, and ongoing or pending phase 2 and 3 clinical trials are codified in study tables.

Expert Opinion:

Unfortunately, hundreds of trials, including of agents effective in systemic malignancy, have not drastically changed management of glioblastoma. This may reflect unique resistance mechanisms and highlights a need for multimodality treatments beyond surgery, radiation, and conventional chemotherapy. Novel techniques, such as those in the emerging field of cancer neuroscience, may help uncover tolerable and effective regimens for this lethal malignancy.

Keywords: Cancer neuroscience, cancer vaccination, chimeric antigen receptor T-cell therapy, glioblastoma, immunotherapy

4.0. Introduction

Glioblastoma IDHwt (GBM) is the most common malignant primary brain tumor of adulthood affecting more than 12,000 individuals per year in the United States (US) and with an estimated worldwide increasing incidence of 0.5-5 per 100,000 (1-3). Existing therapies are not curative. Efforts to develop more effective treatments have yielded only marginal change to survival rates with multimodal therapies. At present, upfront maximal safe resection; concurrent chemoradiation with the alkylating chemotherapy agent TMZ; and 6 adjuvant 28-day cycles of TMZ has been the accepted standard of care for newly diagnosed GBM for the past two decades (4). This regimen demonstrated the value of chemotherapy to extend median overall survival (OS; 12.1 months with RT alone versus 14.6 months with the combination) in 2005, and it remains the backbone of treatment for most patients (4). In 2011, the US Food and Drug Administration (FDA) approved the Novocure Optune ® device, which uses alternating electric fields (tumor treating fields, TTF) to disrupt cancer proliferation, for recurrent GBM given comparable OS to physician’s choice chemotherapy (5). This modality was subsequently approved in 2015 as an adjunctive therapy for newly diagnosed GBM after demonstrating a median OS of 20.9 months with TTF + TMZ vs 16.0 months with TMZ alone (6). However, adoption of TTF has not been universal due to concerns about trial design, patient adherence, and convenience, and many providers elect to offer this treatment on a case-by-case basis (7). Accordingly, novel therapeutic targets and modalities supported by a deeper understanding of the molecular underpinnings of the disease and the importance of the tumor microenvironment in GBM evolution and progression are needed to reshape the landscape of treatment options in coming years.

Here we briefly describe recent work that has informed the current understanding of GBM and its management. We discuss recent advances and the existing challenges faced by clinicians and researchers in identifying new strategies and in bringing new discoveries from the research bench to a clinical application. We also explore promising efforts that take advantage of personalized treatment approaches and incorporate recent discoveries made in the growing field of cancer neuroscience, which have the potential to transform the management of GBM in the near future.

5.0. Prior Investigations and Historical Context

5.1. In-situ Treatment: Carmustine (BCNU) Wafers and Brachytherapy

For most of the 20th century, GBM was managed with resection alone. In the 1980s, radiotherapy became commonplace, but the adoption of chemotherapy for GBM was slow due to uncertainty of benefit, timing, dose, and preferred agents as well as associated toxicities (8). Once preclinical models in the 1990s demonstrated in vitro efficacy with agents such as carmustine, trials were undertaken to explore multiple systemic treatments (9,10). However, short drug half-life, low intra-tumoral drug concentration, limited drug penetrance across the blood-brain barrier, and systemic toxicities were felt to be limiting factors for widespread use. To address these issues, Brem et al explored the safety and efficacy of locally delivered carmustine, administered in the form of wafers implantable into the resection cavity at the time of surgery (11,12). Early data revealed an improvement in overall survival (OS), which was corroborated in a phase 3 trial that demonstrated a slight, but statistically significant, median OS benefit at 13.9 months with carmustine wafers vs 11.6 months with placebo; there was no improvement in progression-free survival (PFS) (11-13). However, while FDA-approved in GBM, concerns about generalizability due to strict patient selection, a requirement for institutional experience with implantation, associated toxicities, and an elevated risk for wound infection have limited the use of this treatment in current clinical practice. Conceptually similar to carmustine wafers is brachytherapy, which employs ionizing radiation delivered locally and continuously through beads or wafers placed into the tumor bed. Trials of varying total radiation dose, dose delivery rates, and placement techniques have yielded inconsistent benefit in GBM, and concerns about this modality mirror those expressed for carmustine wafers (14).

5.2. Anti-angiogenesis

Prior to routine use of molecular characterization for establishing a diagnosis of GBM, histologic evaluation served as the gold standard. One important factor separating low- and high-grade glioma is the presence of microvascular proliferation, which implicates angiogenesis in disease development. Genomic evaluations of GBM early in the 21st century confirmed the role of vascular endothelial growth factor (VEGF) in disease pathogenesis, and use of anti-VEGF treatment garnered significant interest in the neuro-oncology community (15-17). Unfortunately, multiple trials using VEGF targeting in GBM - as monotherapy or in combination, and at both diagnosis or disease recurrence - ultimately demonstrated no survival benefit (18-23). Specifically, no trial to date has conclusively shown prolongation of OS following administration of bevacizumab, the most commonly used VEGF targeting agent, and increases in PFS are likely attributable to reduction in contrast enhancement due to vascular normalization (24,25). Despite minor effects on disease progression, bevacizumab maintains an important role in patient management due to frequent treatment-associated symptomatic improvement and potential for reduced corticosteroid dependence (26).

5.3. Immune Checkpoint Inhibition

Harnessing the immune system to treat malignancy and systemic cancer has ushered tremendous progress in previously recalcitrant disease, including metastatic melanoma, renal cell cancer, and non-small cell lung cancer (27-31). Response to immune checkpoint inhibition (ICI) therapy has also been encouraging for central nervous system (CNS) metastases, opening exploration of primary brain tumor treatment (32-34). Unfortunately, ICI treatment either in head-to-head randomization against or in combination with standard therapy for GBM has not yielded a clear survival benefit (35-37). Notably, a subset of patients with GBM may experience prolonged and durable responses to ICI treatment; the reasons for disparate response are unclear but likely have to do with the immune milieu in the tumor microenvironment, and mutations infrequently enriched in GBM, such as BRAF or PTPN11, seem to be associated with improved outcomes (38). Work is underway to determine additional factors contributing to this inconsistent response and to assist in the identification and selection of patients who may benefit from ICI use, particularly in the recurrent setting. At present, ICI therapy is primarily available under the auspices of a clinical trial (e.g., NCT04977375, NCT03961971, NCT03491683, NCT04396860, and NCT04145115; see also: Table 1).

Table 1.

Recruiting and not yet recruiting international immunotherapy-based phase 2 or 3 clinical trials registered with ClinicalTrials.gov. Included trials have either OS or PFS as primary or secondary outcomes. The above trials exclude exclusively intra-operative or radiation therapy-based as well as purely diagnostic or observational studies. Terms: B7-H3 – B7 homolog 3 protein; CD – cluster of differentiation; EGFR – epidermal growth factor receptor; GBM - glioblastoma; ICI – immune checkpoint inhibitor; IGF - insulin-like growth factor; IL – interleukin; IL13Rα2 – IL-13 receptor subunit alpha 2; MMP2 – matrix metallopeptidase 2; NKG2D – CD129-R for natural killer cells type 2D; RT - radiotherapy; SoC - standard of care; TMZ - temozolomide

Type Intervention (Target) Country (Site) Clinical Trial Number Status Estimated Completion
CAR T cell-based CAR T cells (NKG2D) China (Jiangxi) and Taiwan NCT05131763 and NCT04717999 Recruiting and not yet recruiting (NCT04717999) December 2022 (NCT05131763) and September 2023 (NCT04717999)
CAR T cells (IL13Rα2) US (Duarte, CA) NCT04003649 Recruiting December 2023
CAR T cells (B7-H3) Multiple NCT04077866, NCT05241392, and NCT04385173 (China); NCT05366179, NCT05474378, and NCT05835687 (US) Recruiting March 2024 (NCT04385173) through March 2028 (NCT05835687)
CAR T cells (MMP2) US (Duarte, CA and Austin, TX) NCT04214392 and NCT05627323 Recruiting (NCT04214392) and not yet recruiting (NCT05627323) January and October 2024
CARv3-TEAM-E (EGFR) US (Boston, MA) NCT05660369 Recruiting June 2025
CAR T cells (IL-8R/CD70) US (Gainesville, FL) NCT05353530 Recruiting December 2027
Tris-CAR T cells (IL7Ra) China (Beijing) NCT05577091 Not yet recruiting November 2024
CAR T cells (EGFRvIII) China (Beijing) NCT05802693 Not yet recruiting November 2025
ICI-based AK105, anlotinib, and SoC China (Nanjing) NCT05033587 Recruiting May 2023
Pembrolizumab with and without NGM707 (myeloid target) International (17 sites) NCT04913337 Recruiting February 2025
Atezolizumab and cabozantinib plus SoC US (Houston, TX) NCT05039281s Recruiting December 2025
Sintilimab (anti-PD-1) and bevacizumab China (Zhengzhou) NCT05502991 Not yet recruiting December 2025
Other Cellular Therapies hSTAR (hematopoietic stem cell rescue) US (Bethesda, MD and Cleveland, OH) NCT05052957 Recruiting June 2024
IGV-001 combined agent (autologous GBM cells and ASO against IGF-1) US (23 sites) NCT04485949 Recruiting January 2025
Other T cell-based Activated autologous T cells US (Birmingham, AL, Louisville, KY, and Los Angeles, CA) NCT05664243 and NCT05341947 Recruiting (NCT05664243) and not yet recruiting (NCT05341947) June 2023 (NCT05341947) and December 2025 (NCT05664243)
Efineptakin alfa (long-acting IL-7) and pembrolizumab US (Rochester, MN) NCT05465954 Recruiting October 2024
DeltEx (gamma-delta T cells) plus SoC US (Birmingham, AL and Louisville, KY) NCT05664243 Recruiting December 2025
L19TNF (L19 antibody) plus SoC Switzerland (Zurich) NCT04443010 Recruiting October 2026
Peri-operative Immunotherapy Pembrolizumab with and without ACT001 plus surgery US (Houston, TX) NCT05053880 Recruiting November 2022
Pre-surgical pembrolizumab and radiation therapy US (Los Angeles, CA) NCT05879250 Recruiting December 2023
Surgical nivolumab and ipilimumab US (CA, MA, and NY) NCT04606316 Recruiting December 2023
Surgical pembrolizumab with or without olaparib US (Boston) NCT05463848 Recruiting December 2024
Vaccine Dendritic cell Italy (Milan), Germany (multiple sites), and Taiwan NCT04801147, NCT03395587, and NCT04115761 Recruiting December 2022 (NCT04115761) to September 2024 (NCT03395587)
SurVaxM (survivin) US (11 sites) NCT05163080 Recruiting August 2023
HSV-tk, valacyclovir, and SoC US (Houston, TX) NCT03596086 Recruiting December 2023
VBI-1901 (gB and pp65) US (10 sites) NCT03382977 Recruiting July 2025
TVI-Brain-1 (vaccination and activated T-cell enrichment) US (New Brunswick, NJ) NCT05685004 Not yet recruiting December 2025

6.0. Challenges

Despite identification of novel treatments with success in preclinical or animal models, translation of such promising strategies into patient care has consistently failed, and clinical trial results have been disappointing. There are several possible reasons why clinical studies fail to reproduce preclinical findings. One is that in vitro models and patient-derived xenografts, which are generated from cells cultured from a human tumor specimen and injected into an immunocompromised animal, by definition, are not spontaneously formed in vivo. Following xenografting, this newly introduced cellular material proliferates and integrates in a neural environment that inherently differs from a naturally occurring tumor. Microenvironmental factors that are believed to play a critical role in disease progression - such as glioblastoma stem cells (39), tumor-associated macrophages (40), and novel functional neural-tumor synapses (41-43) - may be entirely absent or affect the xenograft in unpredictable ways. Ongoing work is exploring how immune cells within the brain contribute to tumor growth, but it follows logically that this complex interplay is altered in immunodeficient recipient animals. A second factor is the striking cellular and molecular heterogeneity seen in GBM; most preclinical models use a limited number of commercially available or institutional cell lines that may not faithfully recapitulate tumoral divergence (44). Third is an incomplete understanding of the driver mutations of GBM that may be dynamic during disease evolution.

The treatment of non-CNS tumors has dramatically improved in recent years in no small part due to targeted and immunotherapy-based approaches, which have not yet demonstrated robust or durable responses in human trials for GBM. Despite evidence of a likely evolution of GBM from neural stem or progenitor cells assumed to be associated with early TERT promoter mutations, it is unclear which subsequent alterations may consistently contribute to gliomagenesis to serve as critical therapeutic targets (45); the accumulation of multiple somatic mutations and avoidance of replicative senescence appear key for GBM formation. It is also likely that tumoral exploitation of multiple direct and indirect pathways for progression, including peritumoral neural hyperactivity and neurovascular decoupling, allow for subversion of therapeutic benefit (43,46,47).

Further complicating the treatment landscape is a challenge in interpreting existing clinical trial data. In 2021, the World Health Organization released the fifth edition of its Classification of Central Nervous System Tumors, an update to the 2016 schema. The most notable change for GBM is a requirement that tumors are isocitrate dehydrogenase (IDH) wild-type (wt); prior versions allowed for IDH mutations in the setting of characteristic histological findings such as pseudopalisading necrosis. This distinction is important as the behavior of IDH mutant tumors is significantly different from their wild-type counterparts. More specifically, overall survival (~1-2 years in IDH-wt GBM patients vs 3-4 years in the as-previously-classified IDH-mutant GBM patients) reflects a more indolent, albeit ultimately life-limiting, disease course (48,49). Therefore, interpretation of data predating this reclassification may be confounded by uneven distributions of IDH mutational status between treatment groups, especially from trials prior to the identification of this molecular alteration in 2008 (50).

As our knowledge of the molecular and genetic underpinnings and complexities of GBM evolves, it is relevant to contextualize our discoveries and incorporate genomic, molecular, histologic, and microenvironmental data for rational clinical trial design. A Society for Neuro-oncology (SNO) Think Tank was convened in 2020 to identify barriers to trial design. Some key limitations identified included overly restrictive inclusion and exclusion criteria that diminish both recruitment and generalizability; a paucity of appropriate control arms for phase 2 trials; and the absence of positive phase 2 data prior to initiation of a phase 3 design (51). The development of adaptive trials, such as GBM AGILE (NCT03970447), are attempts to address some of these issues, and umbrella or basket design trials will likely increase in popularity as well.

Lastly, it is important to consider the tolerability of novel treatments. The safety of any new intervention is scrutinized at all phases of trial design, though this is the primary focus of early phase or phase 1 trials; intuitively, highly cytotoxic chemotherapies may be efficacious in vitro but fail early into clinical investigation. However, this is true not only for novel drug design but also for radiation-, immunotherapy-, or device-based treatments.

7.0. On the Horizon

Acknowledging the challenges described above and the so far limited success with ICI therapy, significant interest remains in co-opting the immune system to treat primary brain tumors. Specifically, chimeric antigen receptor (CAR) T-cell therapy, bispecific antibodies, and tumor-directed vaccines are active areas of investigation. Table 1 highlights some ongoing studies of interest.

7.1. CAR T-cell Therapy

CAR T-cell therapy is a cellular immunotherapy approach in which an adaptive immune response is generated to cancer-associated surface antigens independent of major histocompatibility complex activity, thereby allowing recognition of a wider range of targets than natural T-cells (52). Following extraction of the patient’s or donor’s native T-cells, synthetic antigen receptors are added ex vivo that in turn allow for T-cell recognition and attack of the tumor. This technique has heralded a new era of treatment for hematologic malignancies and was first approved for advanced/refractory B-cell lymphoma and refractory pediatric B-cell acute lymphoblastic leukemia in 2017 (53,54). There is considerable interest in applying this modality to solid tumors as well, including those involving the CNS. However, selection of an appropriate, specific, and conserved neoantigen is difficult, and candidate cell-surface antigens are rare for tumors with low mutational burden such as GBM (55).

Fortunately, identification of novel tumor targets has permitted the development of new CAR T-cell products under investigation. One such antigen is the EGFRv3 alteration, seen in up to 30% patients with GBM, which has led to a phase 1 trial (NCT05660369) of CARv3-TEAM-E T cell therapy. Other neoantigens with recruiting clinical trials include B7-H3, an immunoregulatory protein of the B7 family possibly associated with tumor infiltration (NCT04077866, NCT05241392, and NCT04385173 in China and NCT05366179, NCT05474378, and NCT05835687 in the US) (56); a basket trial of NKG2D ligand (NCT05131763 in China); IL-13 receptor ɑ2, a high-affinity receptor expressed by the majority of GBMs with low non-tumor expression, with and without ICIs (NCT04003649 in the US) (57); and matrix metalloproteinase-2 (MMP2) that appears to contribute to chlorotoxin binding to GBM (NCT04214392 in the US) (58). Additional studies of these and other CAR T-cell and activated T-cell therapies that are not yet recruiting, including NCT05341947, NCT05353530, NCT05577091, NCT05627323, NCT05664243, NCT05802693, NCT04717999, and NCT05685004, highlighting the ongoing global interest in these treatments while other neoantigens are being discovered. Table 1 lists ongoing and not yet recruiting studies using immunotherapy approaches for the management of GBM.

Of note, unique toxicities related to tumor-associated inflammation may be seen with CAR T-cell therapies targeting the CNS and likely pose additional challenges to patients with glioblastoma (59,60). The most worrisome CNS adverse event is cerebral edema, which can be fatal; other short- and long-term effects may include delayed neurologic toxicities or non-neurologic symptoms that can be severe enough to halt a trial. As cancer immunotherapeutics are explored for solid tumors, strategies to abrogate or mitigate these on-target, off-tumor effects will be crucial (59).

7.2. Bispecific Antibodies

Similar in concept to CAR T-cell therapy are bispecific antibodies. These compounds build upon success seen with monoclonal antibodies - such as bevacizumab and rituximab - but bind to two different antigens or two epitopes of the same antigen simultaneously; the most common application is to engage both the CD3 receptor and tumor-specific antigens (61). Binding of these targets can recruit and activate immune cells, primarily T-cells, to a tissue of interest and eradicate specific cell populations, which is not possible using monoclonal antibodies alone. The development of these antibodies is technically challenging, and it took from 1985, when the first preclinical bispecific antibody model was created, until 2014 for a clinically accessible agent - blinatumomab - to receive FDA approval (62,63). However, notable progress has been made at antibody generation by applying novel hybridization and “knob-in-hole” techniques, yielding a more extensible and generalizable pathway for production of new agents (64,65). At the time of this publication, nine bispecific antibodies are FDA approved with seven used for malignancies such as lymphoma, multiple myeloma, and non-small cell lung cancer with many more in active clinical trials. The utility of this technique extends beyond malignancy, but one phase 1 trial (NCT03344250) is investigating the use of EGFR-CD3 bi-armed activated T-cells - administered during adjuvant TMZ cycles - following standard of care chemoradiation.

7.3. Vaccines

A longstanding and popular investigational cancer immunotherapy for GBM is tumor-directed vaccination. Although this technique also exploits an adaptive immune response, vaccines differ from CAR T-cell and bispecific antibody therapy due to the administration of an exogenous antigen meant to elicit a response to tumor. An important exception to this paradigm is dendritic cell (DC) vaccination, which requires isolation of DCs from the patient followed by exposure to tumor antigen and exogenous DC maturation prior to reinjection (66). As with the previously discussed immunotherapies, selection of antigen is the key step for vaccine treatment, and multiple targets (such as peptides to EGFRv3 and IDH-1) have been explored. As of this publication, there are 53 completed vaccine trials of which 43 had study sites in the United States; the first study was initiated in 1997. Treatment regimens have included vaccine monotherapy or combinations with standard of care or other experimental agents (67,68).

Although early vaccine trials demonstrated mixed results, two recent trials merit discussion. First is a single arm phase 2a study of SurVaxM, a peptide vaccine conjugate that targets the molecule survivin. This trial enrolled 64 patients with newly diagnosed GBM to receive four “priming” doses of the vaccine product plus sargramostim at two week intervals following resection and concurrent chemoradiation with TMZ (69). After the study treatment period, patients received adjuvant TMZ and maintenance SurVaxM until disease progression. Sixty-three patients were evaluable - as one patient received only one of four “priming” doses - with a six-month progression-free survival (PFS) of 95.2%, median PFS of 11.4 months, and median OS of 25.9 months (69). Within their population, there was a notable difference when stratified by O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status (MGMT promoter methylated n=33 and unmethylated n=29): median PFS was 17.9 vs 7.0 months and median OS was 41.4 vs 25.9 months, both favoring patients with methylated MGMT status (69). These results compare favorably with historical controls, but the lack of a dedicated control arm is a reasonable criticism for this trial. Recruitment is ongoing in a multicenter, randomized, double-blind clinical trial of SurVaxM (NCT05163080).

Second is a phase 3, randomized and double-blind trial of DCVax-L, a personalized vaccination engineered from patient DCs and antigens derived from their tumor sample (70). Patients with newly diagnosed (n=232) or recurrent (n=64) GBM who underwent near or gross total resection were enrolled in a 2:1 fashion to post-chemoradiation, upfront active treatment or placebo with all groups receiving standard of care adjuvant TMZ. The original primary endpoint was designed to be PFS. Importantly, this was a crossover from placebo trial in which every patient eventually received DCVax-L treatment. The initial report in 2018 did not describe PFS outcomes but emphasized OS (23.1 months), particularly in patients with MGMT promoter methylation (34.7 months) (71). When final results were published in 2023, the primary endpoint was changed from PFS to OS under the arguments that placebo arm loss due to crossover and difficulty in delineating pseudoprogression from true progression would render PFS a less useful measure. Overall, the newly diagnosed cohort experienced a median OS of 19.3 months vs 16.5 months in the control group, and in recurrent GBM there was a median OS of 13.2 vs 7.8 months in the control group (70). Both the newly diagnosed and recurrent cohorts therefore demonstrated a prolonged survival compared to existing treatment. To address concerns about the effects of crossover into the active treatment, the research team compared their outcomes to external controls comprising 15 outside trials in which patients received standard of care treatment. However, criticism has been leveled regarding the selection of trials as some with prolonged OS, including EF-14, were not incorporated. Furthermore, the exclusion of patients who underwent biopsy alone reduces the generalizability of these results. Despite the limitations to this study, the use of DC vaccination for GBM is promising, but future studies with formal randomization will need to confirm a true survival benefit with this approach.

Globally, recruitment is ongoing for 18 trials with at least one arm of vaccine-based therapy for adults with GBM: 6 in the United States, 4 in Germany, 4 in China, 2 each in Belgium and Italy, and 1 each in France and the Netherlands.

7.4. Other Agents

New chemotherapies, targeted therapies, repurposed medicines, and devices are under investigation around the world. These include inhibitors of EGFR, exportin 1, fatty acid synthesis, fibroblast growth factor receptor, glutamate, histone deacetylase, Janus kinase, phosphoinositide 3-kinase, poly (ADP-ribose) polymerase, protein arginine methyltransferase 5, receptor for advanced glycation end-products, sphingomyelin synthase 1, topoisomerase 2, and tyrosine kinase; botanical products including polyphenols and cannabinoids; chemotherapies used in other systemic malignancies; multiagent basket and umbrella trials (GBM AGILE, INSIGhT, and N2M2 NOA-20); sonodynamic therapies; nanoparticle delivery; and dietary interventions such as ketogenic diet. Table 2 highlights ongoing and not yet recruiting studies of these and other regimens.

Table 2.

Recruiting and not yet recruiting international phase 2 or 3 clinical trials registered with ClinicalTrials.gov. Included trials have either OS or PFS as primary or secondary outcomes. The above trials exclude exclusively intra-operative or radiation therapy-based as well as purely diagnostic or observational studies. Terms: CBD – cannabidiol; EGFR – epidermal growth factor receptor; FGFR – fibroblast growth factor receptor; GBM - glioblastoma; HDAC – histone deacetylase; JAK – Janus kinase; PARP - poly (ADP-ribose) polymerase; PI3K – phosphoinositide 3-kinase; PRMT5 - protein arginine methyltransferase 5; RAGE - receptor for advanced glycation endproducts; RT - radiotherapy; SoC - standard of care; STAT3 - signal transducer and activator of transcription 3; TCA – tricyclic antidepressant; THC – tetrahydrocannabinol; TKI – tyrosine kinase inhibitor; TMZ - temozolomide

Type Intervention or Trial (Target) Country (Site) Clinical Trial Number Status Estimated Completion
Chemotherapy (Targeted and Traditional) Glutamate inhibition - multiple agents Switzerland (Zurich) NCT05664464 Recruiting June 2023
Denifanstat (fatty acid synthesis inhibitor) and bevacizumab China (Beijing) NCT05118776 Recruiting July 2023
Multiple regimens (N2M2 NOA-20, Neuro Master Match) Germany (13 sites) NCT03158389 Recruiting September 2023
Paxalisib (PI3Kα inhibitor) with ketogenic diet and metformin US (New York, NY) NCT05183204 Recruiting December 2023
Cetuximab (EGFR) plus SoC US (New York, NY) NCT02861898 Recruiting December 2023
Endostatin, TMZ, and irinotecan China (Beijing) NCT04267978 Recruiting February 2024
LAM561 (sphingomyelin synthase 1) plus SoC International (20 sites) NCT04250922 Recruiting February 2024
Capecitabine plus SoC US (New York, NY) NCT03213002 Recruiting June 2024
Verteporfin (EGFR) US (Atlanta, GA) NCT04590664 Recruiting August 2024
Imipramine (TCA) and lomustine US (San Antonio, TX) NCT04863950 Recruiting September 2024
SKL27969 (PRMT5 inhibitor) US (MI, NC, and TX) NCT05388435 Recruiting September 2024
Antisecretory factor Sweden (Lund) NCT05669820 Recruiting January 2025
Debio 0123 (Wee1 kinase inhibitor) plus SoC International (10 sites) NCT05765812 Recruiting January 2025
INSIGhT – multiple regimens US (12 sites) NCT05095376 Recruiting March 2025
WP1066 (JAK2/STAT3 inhibitor) with RT US (Chicago) NCT05879250 Not yet recruiting March 2025
Selinexor (exportin 1) plus SoC US (21 sites) NCT05432804 Recruiting April 2025
NMS-03305293 (PARP inhibitor) plus TMZ International (5 sites) NCT04910022 Recruiting June 2025
Pazopanib (TKI) with TMZ France (Nice) NCT02331498 Recruiting August 2025
Berubicin (topoisomerase 2 inhibitor) plus SoC Poland (Gdansk and Warszawa) NCT04915404 Recruiting October 2025
BPM31510 (shifts glycolysis to oxidative phosphorylation) with RT and TMZ US (CA, NY, VA, and WA) NCT04752813 Recruiting December 2025
Pemigatinib (FGFR inhibitor) International (87 sites) NCT05267106 Recruiting January 2026
GBM AGILE - multiple regimens International (50 sites) NCT03970447 Recruiting June 2026
Lomustine plus SoC US (223 sites) NCT05095376 Recruiting August 2026
Lomustine with or without RT Europe NCT05904119 Recruiting September 2027
NBM-BMX (HDAC-8 inhibitor) Taiwan NCT06012695 Recruiting May 2028
Azeliragon (RAGE inhibitor) plus RT Pending NCT05986851 Not yet recruiting September 2024
APG-157 (multi-polyphenol botanical) and bevacizumab US (Rochester, MN and Omaha, NE) NCT06011109 Not yet recruiting December 2024
NG101m US (Houston, TX) NCT04373785 Not yet recruiting July 2027
Other Tofacitinib (JAK inhibitor) US (Dallas, TX) NCT05326464 Recruiting June 2024
Valganciclovir plus SoC Sweden (Stockholm) NCT04116411 Recruiting August 2024
AGuIX nanoparticles plus SoC France (6 sites) NCT04881032 Recruiting September 2024
Ultrasound with paclitaxel and carboplatin US (Chicago, IL) NCT04528680 Recruiting September 2024
Rhenium nanoliposomes Texas (3 sites) NCT01906385 Recruiting January 2025
TN-TC11G (THC and CBD) Spain (8 sites) NCT03529448 Recruiting March 2025
Liposomal curcumin plus SoC US (Baltimore, MD) NCT05768919 Recruiting February 2026
Nabiximols (cannabinoids) UK (18 sites) NCT05629702 Recruiting February 2026
Sonodynamic therapy (SONALA-001 and Exablate 4000 type 2) US (6 sites) NCT05370508 Recruiting April 2026
Ketogenic diet plus SoC US (CA, NC, and TX) NCT05708352 Recruiting June 2028
SonoCloud-9 ultrasound with carboplatin International (8 sites) NCT05902169 Not yet recruiting January 2028

The advent of novel treatment pathways will likely complement existing and experimental agents, as it has become clear that a personalized but multifaceted approach to GBM treatment is needed. For instance, dietary interventions and neuromodulatory techniques may be used in conjunction with vaccines and standard of care to address treatment evasion by GBM. As novel insights are gained from studying the tumor micro- and macroenvironment, additional targets may be identified that are relevant to disease development and progression.

8.0. Conclusion

Despite decades of difficulty in identifying breakthrough treatments for GBM, recent advances are cause for cautious optimism in the field of neuro-oncology. Immunotherapy, which has yielded noteworthy progress in systemic and hematologic malignancies, initially did not appear to have similar success in intracranial tumors. However, CAR T-cell therapy, bispecific antibodies, and tumor vaccines all have shown promise that indicate - with optimal target selection - new treatments may be within our reach. It has become evident that the identification and development of new therapies requires collaborative integration of preclinical and clinical investigations in an institutional environment conducive to and supportive of this scientific mission.

To ensure preclinical work successfully moves into the translational and clinical domains, there is a need to design both animal studies and clinical trials in rational but creative ways. The adoption and repurposing of techniques used in other scientific disciplines, such as electrophysiologic monitoring and network mapping, has the potential to broaden our understanding of GBM at all levels; genomic analyses and the burgeoning discipline of cancer neurosciences have begun to reveal new insights into factors facilitating tumor development, infiltration, and progression. Using these revelations to improve patient outcomes will likely require near-native recapitulation of the tumor microenvironment, diversity in cell lines studied, cross-institutional collaboration, expanded inclusion criteria for clinical trials, and adaptive study design. Although there is a long way to go, prospects for better treatment of GBM are on the horizon.

9.0. Expert Opinion

In recent years, there has been unprecedented activity to identify pathologic mechanisms of and novel treatments for GBM. Despite this furor, no major therapeutic breakthroughs have come to fruition. Shortcomings in clinical and translational efforts are attributed to the inherent biological sequestration of CNS neoplasms, which are immunologically privileged and consequently less responsive to immunotherapy; inter- and intratumoral heterogeneity that both underscores the molecular diversity of GBM and potentially drives resistance to treatment; biological disparities between animal models and in situ tumors; and the response of GBM to external stressors, such as chemotherapy or radiotherapy, which may produce a hypermutated phenotype and/or activate alternative bioenergetic pathways. These mechanisms all likely contribute meaningfully, but we feel that another key limitation is the prevalence of interventions or regimens targeting a single aspect or mechanism of disease.

Neuroscience, genomic, and molecular techniques continue to reveal new mechanistic knowledge, highlighting the extent to which GBM hijacks existing metabolic and proliferative pathways. In turn, this work has uncovered new treatment targets, leading to an ever-increasing arsenal of agents with compelling efficacy in the laboratory. We then eagerly test these new medicines or devices only to find discordance between preclinical and in-human results, but not as a consequence of poor academic rigor. Science requires logical and methodical testing to facilitate interpretation of results and ensure reproducibility; there is little doubt that most GBM research adheres to this expectation. However, the paradigm of the scientific method is typically applied by evaluating a single investigational agent per arm or clinical trial, which is meant to reduce the number of controllable variables. Unfortunately, as historical efforts have shown, it is unlikely that a single therapy will work completely in isolation, and the rational selection and testing of multiple agents - each affecting a different aspect of GBM growth - is likely to yield the greatest efficacy.

Any potentially curative therapy for GBM will acknowledge and address multiple intra- and extra-tumoral factors. These may include de novo functional and hyperactive synapses between tumor and surrounding brain, which co-opt AMPA and/or NMDA receptors and non-activity dependent ionic dysregulation (43); tumoral preference for - but not dependence on - glycolysis (72); paracrine signaling, such as through neuroligin-3 and brain-derived neurotrophic factor (73,74); neuro-immunologic cross-talk; blood vessel dysregulation and angiogenesis; and cellular hyperproliferation, arising at least in part from insulin growth factor 1, PI3K, and MAPK (75-77). The repurposing of existing medications, such as those reducing glutamate activity or neurotrophins, may complement new targeted therapies or established agents. Furthermore, recent clinical trials have demonstrated great potential for immunotherapy, with cancer vaccines and CAR T-cell treatment as active areas of research with multiple ongoing trials. Lastly, the development or incorporation of neuromodulatory techniques may also dramatically affect our understanding and treatment of GBM, and these devices or agents may represent a growing field of exciting research. Importantly, assessment of treatment effect must take into account that even a modest benefit, when used in concert with other therapies affecting different pathways, may merit further investigation.

We therefore posit that a contemporary and highly efficacious treatment regimen will require a multimodality approach extending beyond surgery, radiation, and classical chemotherapy. Although it will be necessary to balance the toxicity or tolerability of any prospective treatment(s) with the potential benefit, the advances in GBM research in recent years is a source of hope for future patients.

3.0. Highlights.

  • Glioblastoma, IDH wildtype, (GBM) affects more than 12,000 individuals per year in the US with an estimated worldwide increasing incidence of 0.5-5 per 100,000, and existing therapies are not curative

  • Trials of brachytherapy, chemotherapy-impregnated wafers, high-dose chemotherapy, molecular-targeted therapies, anti-angiogenesis, and immune checkpoint inhibition have not resulted in changes to present management

  • Evolving understanding of GBM due to molecular characterization, genomics, and cancer neuroscience has the potential to improve outcomes

  • Recent work has demonstrated the potential benefit of dendritic cell and peptide GBM vaccines as well as CAR T-cell therapies

  • Many trials are underway of immunotherapy- and non-immunotherapy-based regimens, including those targeting neural hyperexcitability, tumor metabolism, and neuro-immune signaling

  • A multimodality, multi-pathway treatment regimen has the greatest likelihood of durable response in GBM but will require creative trial design on a solid scientific foundation

Funding Details

TN is supported by the National Institutes of Health (2K12CA090354-21), but this paper was not funded.

Footnotes

Declaration of Interests

JD has been a consultant for Amgen, Novartis, and Ono Therapeutics. JD also has received royalties as an author for UpToDate. The authors otherwise have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

10.0 References

  • 1.Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C, et al. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2015–2019. Neuro-Oncol. 2022. Oct 5;24(Supplement_5):v1–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Grech N, Dalli T, Mizzi S, Meilak L, Calleja N, Zrinzo A. Rising Incidence of Glioblastoma Multiforme in a Well-Defined Population. Cureus [Internet]. 2020. May 19 [cited 2023 Jul 31]; Available from: https://www.cureus.com/articles/31024-rising-incidence-of-glioblastoma-multiforme-in-a-well-defined-population [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lin D, Wang M, Chen Y, Gong J, Chen L, Shi X, et al. Trends in Intracranial Glioma Incidence and Mortality in the United States, 1975-2018. Front Oncol. 2021. Nov 1;11:748061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Stupp R, Weller M, Belanger K, Bogdahn U, Ludwin SK, Lacombe D, et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N Engl J Med. 2005; [DOI] [PubMed] [Google Scholar]
  • 5.Stupp R, Wong ET, Kanner AA, Steinberg D, Engelhard H, Heidecke V, 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. Sep;48(14):2192–202. [DOI] [PubMed] [Google Scholar]
  • 6.Stupp R, Taillibert S, Kanner A, Read W, Steinberg DM, Lhermitte B, et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA. 2017. Dec 19;318(23):2306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lassman AB, Joanta-Gomez AE, Pan PC, Wick W. Current usage of tumor treating fields for glioblastoma. Neuro-Oncol Adv. 2020. Jan 1;2(1):vdaa069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Walker MD, Green SB, Byar DP, Alexander E, Batzdorf U, Brooks WH, et al. Randomized Comparisons of Radiotherapy and Nitrosoureas for the Treatment of Malignant Glioma after Surgery. N Engl J Med. 1980. Dec 4;303(23):1323–9. [DOI] [PubMed] [Google Scholar]
  • 9.Yoshida J, Shibuya N, Kobayashi T, Kageyama N. Sensitivity to 1-(4-amino-2-methyl-5-pyrimidinyl)methyl-3-(2-chloroethyl)-3-nitrosourea hydrochloride (ACNU) of glioma cells in vivo and in vitro. Cancer. 1982. Aug 1;50(3):410–8. [DOI] [PubMed] [Google Scholar]
  • 10.Wolff JEA, Trilling T, Mölenkamp G, Egeler RM, Jürgens H. Chemosensitivity of glioma cells in vitro: a meta analysis. J Cancer Res Clin Oncol. 1999. Aug 6;125(8–9):481–6. [DOI] [PubMed] [Google Scholar]
  • 11.Brem H, Piantadosi S, Burger PC, Walker M, Selker R, Vick NA, et al. Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Lancet. 1995. Apr;345(8956):1008–12. [DOI] [PubMed] [Google Scholar]
  • 12.Brem H, Ewend MG, Piantadosi S, Greenhoot J, Burger PC, Sisti M. The safety of interstitial chemotherapy with BCNU-loaded polymer followed by radiation therapy in the treatment of newly diagnosed malignant gliomas: Phase I trial. J Neurooncol. 1995. Nov;26(2):111–23. [DOI] [PubMed] [Google Scholar]
  • 13.Westphal M, Hilt DC, Bortey E, Delavault P, Olivares R, Warnke PC, et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma1,2. 2003; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Barbarite E, Sick JT, Berchmans E, Bregy A, Shah AH, Elsayyad N, et al. The role of brachytherapy in the treatment of glioblastoma multiforme. Neurosurg Rev. 2017. Apr;40(2):195–211. [DOI] [PubMed] [Google Scholar]
  • 15.Godard S, Getz G, Delorenzi M, Farmer P, Kobayashi H, Desbaillets I, et al. Classification of Human Astrocytic Gliomas on the Basis of Gene Expression: A Correlated Group of Genes with Angiogenic Activity Emerges As a Strong Predictor of Subtypes1,2. [PubMed] [Google Scholar]
  • 16.Lamszus K, Ulbricht U, Matschke J, Brockmann MA, Fillbrandt R, Westphal M. Levels of Soluble Vascular Endothelial Growth Factor (VEGF) Receptor 1 in Astrocytic Tumors and Its Relation to Malignancy, Vascularity, and VEGF-A. [PubMed] [Google Scholar]
  • 17.Jain HV, Nör JE, Jackson TL. Modeling the VEGF–Bcl-2–CXCL8 Pathway in Intratumoral Agiogenesis. Bull Math Biol. 2008. Jan;70(1):89–117. [DOI] [PubMed] [Google Scholar]
  • 18.Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. A Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma. N Engl J Med. 2014. Feb 20;370(8):699–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Vredenburgh JJ, Desjardins A, Herndon JE, Marcello J, Reardon DA, Quinn JA, et al. Bevacizumab Plus Irinotecan in Recurrent Glioblastoma Multiforme. J Clin Oncol. 2007. Oct 20;25(30):4722–9. [DOI] [PubMed] [Google Scholar]
  • 20.Chauffert B, Feuvret L, Bonnetain F, Taillandier L, Frappaz D, Taillia H, et al. Randomized phase II trial of irinotecan and bevacizumab as neo-adjuvant and adjuvant to temozolomide-based chemoradiation compared with temozolomide-chemoradiation for unresectable glioblastoma: final results of the TEMAVIR study from ANOCEF. Ann Oncol. 2014. Jul;25(7):1442–7. [DOI] [PubMed] [Google Scholar]
  • 21.Balana C, De Las Penas R, Sepúlveda JM, Gil-Gil MJ, Luque R, Gallego O, et al. Bevacizumab and temozolomide versus temozolomide alone as neoadjuvant treatment in unresected glioblastoma: the GENOM 009 randomized phase II trial. J Neurooncol. 2016. May;127(3):569–79. [DOI] [PubMed] [Google Scholar]
  • 22.Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I, et al. Lomustine and Bevacizumab in Progressive Glioblastoma. N Engl J Med. 2017. Nov 16;377(20):1954–63. [DOI] [PubMed] [Google Scholar]
  • 23.Patel KS, Everson RG, Yao J, Raymond C, Goldman J, Schlossman J, et al. Diffusion Magnetic Resonance Imaging Phenotypes Predict Overall Survival Benefit From Bevacizumab or Surgery in Recurrent Glioblastoma With Large Tumor Burden. Neurosurgery. 2020. Nov;87(5):931–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.De Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y, et al. Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro-Oncol. 2010. Mar 1;12(3):233–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Jain RK. Normalizing Tumor Microenvironment to Treat Cancer: Bench to Bedside to Biomarkers. J Clin Oncol. 2013. Jun 10;31(17):2205–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Vredenburgh JJ, Cloughesy T, Samant M, Prados M, Wen PY, Mikkelsen T, et al. Corticosteroid Use in Patients with Glioblastoma at First or Second Relapse Treated with Bevacizumab in the BRAIN Study. The Oncologist. 2010. Dec 1;15(12):1329–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N Engl J Med. 2010. Aug 19;363(8):711–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med. 2019. Mar 21;380(12):1116–27. [DOI] [PubMed] [Google Scholar]
  • 29.Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus Docetaxel in Advanced Nonsquamous Non–Small-Cell Lung Cancer. N Engl J Med. 2015. Oct 22;373(17):1627–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, et al. Pembrolizumab for the Treatment of Non–Small-Cell Lung Cancer. N Engl J Med. 2015. May 21;372(21):2018–28. [DOI] [PubMed] [Google Scholar]
  • 31.Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med. 2015. Jul 2;373(1):23–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hendriks LEL, Henon C, Auclin E, Mezquita L, Ferrara R, Audigier-Valette C, et al. Outcome of Patients with Non–Small Cell Lung Cancer and Brain Metastases Treated with Checkpoint Inhibitors. J Thorac Oncol. 2019. Jul;14(7):1244–54. [DOI] [PubMed] [Google Scholar]
  • 33.Crinò L, Bronte G, Bidoli P, Cravero P, Minenza E, Cortesi E, et al. Nivolumab and brain metastases in patients with advanced non-squamous non-small cell lung cancer. Lung Cancer. 2019. Mar;129:35–40. [DOI] [PubMed] [Google Scholar]
  • 34.Tringale KR, Reiner AS, Sehgal RR, Panageas K, Betof Warner AS, Postow MA, et al. Efficacy of immunotherapy for melanoma brain metastases in patients with concurrent corticosteroid exposure. CNS Oncol. 2023. Mar 1;12(1):CNS93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick A, et al. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020. Jul 1;6(7):1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Omuro A, Brandes AA, Carpentier AF, Idbaih A, Reardon DA, Cloughesy T, et al. Radiotherapy combined with nivolumab or temozolomide for newly diagnosed glioblastoma with unmethylated MGMT promoter: An international randomized phase III trial. Neuro-Oncol. 2023. Jan 5;25(1):123–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Lim M, Weller M, Idbaih A, Steinbach J, Finocchiaro G, Raval RR, et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro-Oncol. 2022. Nov 2;24(11):1935–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zhao J, Chen AX, Gartrell RD, Silverman AM, Aparicio L, Chu T, et al. Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat Med. 2019. Mar;25(3):462–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Almairac F, Turchi L, Sakakini N, Debruyne DN, Elkeurti S, Gjernes E, et al. ERK-Mediated Loss of miR-199a-3p and Induction of EGR1 Act as a “Toggle Switch” of GBM Cell Dedifferentiation into NANOG- and OCT4-Positive Cells. Cancer Res. 2020. Aug 15;80(16):3236–50. [DOI] [PubMed] [Google Scholar]
  • 40.Wu A, Wei J, Kong LY, Wang Y, Priebe W, Qiao W, et al. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro-Oncol. 2010. Nov;12(11):1113–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Venkataramani V, Tanev DI, Strahle C, Studier-Fischer A, Fankhauser L, Kessler T, et al. Glutamatergic synaptic input to glioma cells drives brain tumour progression. Nature. 2019. Sep 26;573(7775):532–8. [DOI] [PubMed] [Google Scholar]
  • 42.Venkatesh HS, Tam LT, Woo PJ, Lennon J, Nagaraja S, Gillespie SM, et al. Targeting neuronal activity-regulated neuroligin-3 dependency in high-grade glioma. Nature. 2017. Sep;549(7673):533–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Venkatesh HS, Morishita W, Geraghty AC, Silverbush D, Gillespie SM, Arzt M, et al. Electrical and synaptic integration of glioma into neural circuits. Nature. 2019. Sep 26;573(7775):539–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cassidy JW, Caldas C, Bruna A. Maintaining Tumor Heterogeneity in Patient-Derived Tumor Xenografts. Cancer Res. 2015. Aug 1;75(15):2963–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lee JH, Lee JE, Kahng JY, Kim SH, Park JS, Yoon SJ, et al. Human glioblastoma arises from subventricular zone cells with low-level driver mutations. Nature. 2018. Aug;560(7717):243–7. [DOI] [PubMed] [Google Scholar]
  • 46.Chow DS, Horenstein CI, Canoll P, Lignelli A, Hillman EMC, Filippi CG, et al. Glioblastoma Induces Vascular Dysregulation in Nonenhancing Peritumoral Regions in Humans. Am J Roentgenol. 2016. May;206(5):1073–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Montgomery MK, Kim SH, Dovas A, Zhao HT, Goldberg AR, Xu W, et al. Glioma-Induced Alterations in Neuronal Activity and Neurovascular Coupling during Disease Progression. Cell Rep. 2020. Apr;31(2):107500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Tesileanu CMS, Dirven L, Wijnenga MMJ, Koekkoek JAF, Vincent AJPE, Dubbink HJ, et al. Survival of diffuse astrocytic glioma, IDH1/2 wildtype, with molecular features of glioblastoma, WHO grade IV: a confirmation of the cIMPACT-NOW criteria. Neuro-Oncol. 2020. Apr 15;22(4):515–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hartmann C, Hentschel B, Wick W, Capper D, Felsberg J, Simon M, et al. Patients with IDH1 wild type anaplastic astrocytomas exhibit worse prognosis than IDH1-mutated glioblastomas, and IDH1 mutation status accounts for the unfavorable prognostic effect of higher age: implications for classification of gliomas. Acta Neuropathol (Berl). 2010. Dec;120(6):707–18. [DOI] [PubMed] [Google Scholar]
  • 50.Parsons DW, Jones S, Zhang X, Lin JCH, Leary RJ, Angenendt P, et al. An Integrated Genomic Analysis of Human Glioblastoma Multiforme. Science. 2008. Sep 26;321(5897):1807–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bagley SJ, Kothari S, Rahman R, Lee EQ, Dunn GP, Galanis E, et al. Glioblastoma Clinical Trials: Current Landscape and Opportunities for Improvement. Clin Cancer Res. 2022. Feb 15;28(4):594–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.June CH, Sadelain M. Chimeric Antigen Receptor Therapy. N Engl J Med. 2018. Jul 5;379(1):64–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med. 2018. Feb;378(5):439–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. N Engl J Med. 2017. Dec 28;377(26):2531–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015. Apr 3;348(6230):69–74. [DOI] [PubMed] [Google Scholar]
  • 56.Li Y, Yang X, Wu Y, Zhao K, Ye Z, Zhu J, et al. B7-H3 promotes gastric cancer cell migration and invasion. Oncotarget. 2017. Sep 22;8(42):71725–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Brown CE, Aguilar B, Starr R, Yang X, Chang WC, Weng L, et al. Optimization of IL13Rα2-Targeted Chimeric Antigen Receptor T Cells for Improved Anti-tumor Efficacy against Glioblastoma. Mol Ther. 2018. Jan;26(1):31–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Deshane J, Garner CC, Sontheimer H. Chlorotoxin Inhibits Glioma Cell Invasion via Matrix Metalloproteinase-2. J Biol Chem. 2003. Feb;278(6):4135–44. [DOI] [PubMed] [Google Scholar]
  • 59.Flugel CL, Majzner RG, Krenciute G, Dotti G, Riddell SR, Wagner DL, et al. Overcoming on-target, off-tumour toxicity of CAR T cell therapy for solid tumours. Nat Rev Clin Oncol. 2023. Jan;20(1):49–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Mahdi J, Dietrich J, Straathof K, Roddie C, Scott BJ, Davidson TB, et al. Tumor inflammation-associated neurotoxicity. Nat Med. 2023. Apr;29(4):803–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Spiess C, Zhai Q, Carter PJ. Alternative molecular formats and therapeutic applications for bispecific antibodies. Mol Immunol. 2015. Oct;67(2):95–106. [DOI] [PubMed] [Google Scholar]
  • 62.Perez P, Hoffman RW, Shaw S, Bluestone JA, Segal DM. Specific targeting of cytotoxic T cells by anti-T3 linked to anti-target cell antibody. Nature. 1985. Jul;316(6026):354–6. [DOI] [PubMed] [Google Scholar]
  • 63.Przepiorka D, Ko CW, Deisseroth A, Yancey CL, Candau-Chacon R, Chiu HJ, et al. FDA Approval: Blinatumomab. Clin Cancer Res. 2015. Sep 15;21(18):4035–9. [DOI] [PubMed] [Google Scholar]
  • 64.Zhu Z, Presta LG, Zapata G, Carter P. Remodeling domain interfaces to enhance heterodimer formation: Domain interjace remodeling. Protein Sci. 1997. Apr;6(4):781–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Lu RM, Hwang YC, Liu IJ, Lee CC, Tsai HZ, Li HJ, et al. Development of therapeutic antibodies for the treatment of diseases. J Biomed Sci. 2020. Dec;27(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012. Apr;12(4):265–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Datsi A, Sorg RV. Dendritic Cell Vaccination of Glioblastoma: Road to Success or Dead End. Front Immunol. 2021. Nov 2;12:770390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yang T, Shi Y, Liang T, Xing H, Ma W, Li YM, et al. Peptide vaccine against glioblastoma: from bench to bedside. Holist Integr Oncol. 2022. Dec 16;1(1):21. [Google Scholar]
  • 69.Ahluwalia MS, Reardon DA, Abad AP, Curry WT, Wong ET, Figel SA, et al. Phase IIa Study of SurVaxM Plus Adjuvant Temozolomide for Newly Diagnosed Glioblastoma. J Clin Oncol. 2023. Mar 1;41(7):1453–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Liau LM, Ashkan K, Brem S, Campian JL, Trusheim JE, Iwamoto FM, et al. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination With Extension of Survival Among Patients With Newly Diagnosed and Recurrent Glioblastoma: A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol. 2023. Jan 1;9(1):112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liau LM, Ashkan K, Tran DD, Campian JL, Trusheim JE, Cobbs CS, 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. May 29;16(1):142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Agnihotri S, Zadeh G. Metabolic reprogramming in glioblastoma: the influence of cancer metabolism on epigenetics and unanswered questions. Neuro-Oncol. 2016. Feb;18(2):160–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Wang X, Prager BC, Wu Q, Kim LJY, Gimple RC, Shi Y, et al. Reciprocal Signaling between Glioblastoma Stem Cells and Differentiated Tumor Cells Promotes Malignant Progression. Cell Stem Cell. 2018. Apr;22(4):514–528.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Venkatesh HS, Johung TB, Caretti V, Noll A, Tang Y, Nagaraja S, et al. Neuronal Activity Promotes Glioma Growth through Neuroligin-3 Secretion. Cell. 2015. May;161(4):803–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tirrò E, Massimino M, Romano C, Martorana F, Pennisi MS, Stella S, et al. Prognostic and Therapeutic Roles of the Insulin Growth Factor System in Glioblastoma. Front Oncol. 2021. Feb 2;10:612385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Langhans J, Schneele L, Trenkler N, Von Bandemer H, Nonnenmacher L, Karpel-Massler G, et al. The effects of PI3K-mediated signalling on glioblastoma cell behaviour. Oncogenesis. 2017. Nov 29;6(11):398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zohrabian VM, Forzani B, Chau Z, Murali R, Jhanwar-Uniyal M. Rho/ROCK and MAPK Signaling Pathways Are Involved in Glioblastoma Cell Migration and Proliferation. ANTICANCER Res. 2009; [PubMed] [Google Scholar]

RESOURCES