Next Article in Journal
NTPDase1/CD39 Ectonucleotidase Is Necessary for Normal Arterial Diameter Adaptation to Flow
Next Article in Special Issue
miRNAs in the Box: Potential Diagnostic Role for Extracellular Vesicle-Packaged miRNA-27a and miRNA-128 in Breast Cancer
Previous Article in Journal
The Intrinsically Disordered N Terminus in Atg12 from Yeast Is Necessary for the Functional Structure of the Protein
Previous Article in Special Issue
Prostate Cancer Stem Cells: Biology and Treatment Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glioblastoma Immunotherapy: A Systematic Review of the Present Strategies and Prospects for Advancements

1
Department of Medical and Surgical Specialties, Division of Neurosurgery, Radiological Sciences and Public Health, University of Brescia, Piazza Spedali Civili 1, 25123 Brescia, Italy
2
Department of Ophthalmology, University Hospital of Udine, P.le S. Maria della Misericordia 15, 33100 Udine, Italy
3
Neurosurgery Unit, Head-Neck and NeuroScience Department, University Hospital of Udine, P.le S. Maria della Misericordia 15, 33100 Udine, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(20), 15037; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242015037
Submission received: 13 September 2023 / Revised: 4 October 2023 / Accepted: 5 October 2023 / Published: 10 October 2023

Abstract

:
Glioblastoma (GBM) is characterized by aggressive growth and high rates of recurrence. Despite the advancements in conventional therapies, the prognosis for GBM patients remains poor. Immunotherapy has recently emerged as a potential treatment option. The aim of this systematic review is to assess the current strategies and future perspectives of the GBM immunotherapy strategies. A systematic search was conducted across major medical databases (PubMed, Embase, and Cochrane Library) up to 3 September 2023. The search strategy utilized relevant Medical Subject Heading (MeSH) terms and keywords related to “glioblastomas,” “immunotherapies,” and “treatment.” The studies included in this review consist of randomized controlled trials, non-randomized controlled trials, and cohort studies reporting on the use of immunotherapies for the treatment of gliomas in human subjects. A total of 1588 papers are initially identified. Eligibility is confirmed for 752 articles, while 655 are excluded for various reasons, including irrelevance to the research topic (627), insufficient method and results details (12), and being case-series or cohort studies (22), systematic literature reviews, or meta-analyses (3). All the studies within the systematic review were clinical trials spanning from 1995 to 2023, involving 6383 patients. Neuro-oncology published the most glioma immunotherapy-related clinical trials (15/97, 16%). Most studies were released between 2018 and 2022, averaging nine publications annually during this period. Adoptive cellular transfer chimeric antigen receptor (CAR) T cells were the primary focus in 11% of the studies, with immune checkpoint inhibitors (ICIs), oncolytic viruses (OVs), and cancer vaccines (CVs) comprising 26%, 12%, and 51%, respectively. Phase-I trials constituted the majority at 51%, while phase-III trials were only 7% of the total. Among these trials, 60% were single arm, 39% double arm, and one multi-arm. Immunotherapies were predominantly employed for recurrent GBM (55%). The review also revealed ongoing clinical trials, including 9 on ICIs, 7 on CVs, 10 on OVs, and 8 on CAR T cells, totaling 34 trials, with phase-I trials representing the majority at 53%, and only one in phase III. Overcoming immunotolerance, stimulating robust tumor antigen responses, and countering immunosuppressive microenvironment mechanisms are critical for curative GBM immunotherapy. Immune checkpoint inhibitors, such as PD-1 and CTLA-4 inhibitors, show promise, with the ongoing research aiming to enhance their effectiveness. Personalized cancer vaccines, especially targeting neoantigens, offer substantial potential. Oncolytic viruses exhibited dual mechanisms and a breakthrough status in the clinical trials. CAR T-cell therapy, engineered for specific antigen targeting, yields encouraging results, particularly against IL13 Rα2 and EGFRvIII. The development of second-generation CAR T cells with improved specificity exemplifies their adaptability.

1. Introduction

Glioblastoma (GBM) retains its status as the foremost prevalent and most malignant glial tumor. It accounts for over 50% of all primary brain tumors in the United States, presenting an annual incidence rate of approximately 3 cases per 100,000 individuals [1]. To date, GBM remains characterized by its high aggressiveness and unresponsiveness to nearly all current standard-of-care interventions, which encompass the combined use of chemotherapy and radiation post-surgical resection. Indeed, in spite of these aggressive treatment regimens, the median overall survival (OS) of afflicted patients extends no more than 15 months from their initial enrollment, just preceding the commencement of radiation therapy (RT) and concurrent chemotherapy (CT) involving temozolomide. This outcome is accompanied by a disheartening 5-year OS rate of less than 10%.
The difficulty of GBM treatment is primarily attributed to the limited population of therapy-resistant glioblastoma stem cells (GSCs) and the intricate tapestry of inter- and intra-tumor heterogeneity, which comprises various GBMs subtypes and stromal cells within the tumor microenvironment (TME) [2]. The GSCs bear significant responsibility for the recurrence of glioblastoma and its resistance to therapy, owing to their robust DNA repair mechanisms, [2] multi-drug-resistance traits, [3] and adept immune evasion tactics. The GBM TME plays a pivotal role in governing cellular behavior, driving GSC adaptability, and ultimately fostering therapeutic resistance [2,4,5]. Furthermore, GBM cells attract and modify immune cells distinct from microglia, reinforcing tumor growth and cultivating an immunosuppressive TME through the secretion of cytokines, extracellular vesicles, and the formation of connecting nanotubes [5].
The effectiveness of immunotherapy has already been firmly established across various solid tumor types, including melanoma, prostate cancer, non-small-cell lung cancer, and renal cell carcinoma. This success marks a notable advancement in the burgeoning field of immunotherapy, which hinges on the concept of re-educating and harnessing the patient’s immune response to combat tumors. These methods have progressively found their place in the treatment of a variety of cancers, including those afflicting the brain. Contemporary strategies for immunotherapy in cancer treatment primarily revolve around immune checkpoint blockade (ICB) agents. Furthermore, therapeutic vaccines, adoptive cell therapy, monoclonal antibodies (mAbs), and oncolytic viruses also constitute essential components of current immunotherapeutic approaches [1,4,6,7,8,9,10,11,12].
Numerous clinical trials have explored the application of immunotherapy in the context of GBMs. Nevertheless, due to the wide array of immunotherapy approaches employed, the varying selection of molecular targets, and the diversity of combination therapy strategies, questions persist concerning the effectiveness and safety of immunotherapies for GBM. Additionally, there is a noticeable absence of a recent comprehensive systematic review addressing the current strategies and future prospects of GBM immunotherapy [1,6,7].
This systematic literature review comprehensively examines a range of prominent immunotherapeutic strategies aimed at combating GBMs, including adoptive cellular transfer chimeric antigen receptor (CAR) T and NK cells, oncolytic viruses (OVs), cancer vaccines (CVs), immune checkpoint inhibitors (ICIs), and multifaceted combination therapies. Additionally, we provide a brief discussion on the rationale underpinning these approaches, while shedding light on their inherent limitations and the unique challenges that arise during the treatment of GBMs.

2. Methods

2.1. Literature Review

The systematic review was performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines [12]. Two authors performed a systematically comprehensive literature search of the PubMed, Ovid MEDLINE, and Ovid EMBASE databases. The first literature search was performed on 10 August 2023, and the search was updated on 3 September 2023. A combination of keyword searches was performed to generate a search strategy. The search keywords, including “glioblastoma”, “immunotherapy”, “target therapy”, “vaccines”, “monoclonal antibodies”, “overall survival”, and “progression free survival”, were used in both AND and OR combinations. Studies were retrieved using the following Medical Subject Heading (MeSH) terms and Boolean operators: (“glioma” OR “glioblastoma” OR “GBM”) AND (“immunotherapy” OR “target therapy”) AND (“outcomes” OR “prognosis” OR “progression free survival” OR “overall survival”). Other pertinent articles were identified through reference analysis of selected papers. A search filter was set to show only publications over the designated period: 1990–2023.
All studies were selected based on the following inclusion criteria: (1) English language; (2) clinical trials, including: single-arm or double-arm studies, and among them randomized controlled or non-randomized controlled trials; (3) studies on GBM immunotherapy strategies, both as stand-alone and combined therapies with CT and/or RT; and (4) studies including at least OS and progression free survival (PFS) among the outcomes analyzed. The following exclusion criteria were employed: (1) editorials, case reports, case series, cohort studies, literature reviews, and meta-analyses; (2) studies that did not clearly define the methods and/or results; and (3) studies that did not report data on PFS or OS.
The list of identified studies was imported into Endnote X9 and duplicates were removed. Two independent researchers (E.A. and P.P.P.) checked the results according to the inclusion and exclusion criteria. A third reviewer (M.Z.) resolved all disagreements. Then, the eligible articles were subject to full-text screening.

2.2. Data Extraction

For each study, we abstracted the following information: authors, year and journal of publication, title, name and phase of the clinical trial, number of patients, diagnosis, follow-up length, immunotherapy treatment, and outcomes.

2.3. Outcomes

Our primary outcomes were OS and PFS related to GBM immunotherapy.

2.4. Risk of Bias Assessment

The Newcastle–Ottawa scale (NOS) [13] was used to assess the quality of the included studies. A quality assessment was performed by assessing the selection criteria, comparability of the study, and outcome assessment. The ideal score was 9. Higher scores indicated a better quality of studies. Studies receiving 7 or more points were considered high-quality studies. Two authors (E.A. and P.P.P.) performed the quality assessment independently. When discrepancies arose, the papers were re-examined by the third author (Figure 1).

2.5. Statistical Analysis

Descriptive statistics were reported, including ranges and percentages. All statistical analyses were performed using the R statistical package v3.4.1 http://www.r-project.org (accessed on 6 September 2023).

3. Results and Discussion

3.1. Literature Review Results

A total of 1588 papers were identified after duplicate removal. After title and abstract analyses, 763 articles were identified for a full-text analysis. Eligibility was ascertained for 752 articles. The remaining 655 articles were excluded for the following reasons: (1) not relevant to the research topic (627 articles), (2) lack of method and/or results details (12 articles), case series and cohort studies (22 articles), and systematic literature review or meta-analysis (3 articles). All studies included in the analysis had at least one or more outcome measures available for one or more of the patient groups analyzed. Figure 2 shows the flowchart according to the PRISMA statement.
The PRISMA checklist is available as Appendix A (Figure A1).

3.2. Data Analysis

A summary of the included studies is presented in Table 1.
All the studies included in our systematic review were clinical trials, with study periods ranging from 1995 to 2023. In total, 6383 patients were enrolled in these trials. Neuro-oncology is the scientific journal that has published the highest number of clinical trials related to glioma immunotherapy (15/97, 16%), followed by Clinical Cancer Research (9/97, 9%), and Cancer Immunology Immunotherapy (6/97, 6%). The majority of these studies were published between 2018 and 2022, with 9 studies published each year during this period.
In terms of the classes of immunotherapeutic agents studied, least of the research focused on CAR T cells, accounting for 11/97 (11%) of the studies. Other significant categories included ICIs at 25/97 (26%), OVs at 12/97 (12%), and CVs at 49/97 (51%). Among these studies, phase-I trials were the most numerous, comprising 51% (49”97), while only 7% (7/97) were phase-III trials. Of the clinical trials, 60% (60/97) were single-arm studies, 39% (38/97) were double-arm studies, and only 1 was a multi-arm study. Among the double-arm and multi-arm trials, 85% (33/39) were randomized controlled trials. Among these, 61% (20/33) demonstrated improved OS and/or PFS compared to conventional treatment.
Immunotherapies were predominantly used for recurrent GBM in 55% of cases (53/97), while in the remaining cases, they were used as the initial treatment. In 25% of the latter cases (11/44), immunotherapies were used as the sole therapy without the addition of other adjuvant therapies, such as RT and/or CT.
A summary of the ongoing studies on GBM immunotherapies is presented in Table 2, Table 3, Table 4 and Table 5.
From the review, it emerged that 9 clinical trials were ongoing on ICIs, 7 on CVs, 10 on OVs, and 8 on CAR T cells, for a total of 34 clinical trials. Of these, most (18/34, 53%) were phase-I clinical trials, while only one was phase III.

3.3. Discussion

In this study, we conducted a systematic review of all the clinical trials published and ongoing between 1995 and 2023. Our findings reveal that the primary classes of immunotherapeutic agents under investigation include CAR T cells (11%), ICIs (26%), OVs (12%), and CVs (51%). Phase-I trials constituted the majority, accounting for 51% of the studies, whereas phase-III trials comprised only 7%. Double-arm studies constituted only 39%; however, among them, 85% were randomized controlled trials. Among these trials, 61% demonstrated improved overall survival (OS) and/or progression free survival (PFS) rates when utilizing GBM immunotherapy, both as a standalone strategy and in combination with conventional treatment. A total of 34 clinical trials are ongoing and the majority of them are phase-I CTs.

3.3.1. Immunosuppressive Mechanisms Employed by GBM

Glioblastoma, recognized as the most aggressive primary brain tumor, poses significant challenges due to its rapid growth, capacity to infiltrate brain tissue, molecular diversity, and resistance to treatment [110]. While immunotherapy holds promise in combating GBM, it encounters formidable obstacles related to the immunological environment of the central nervous system (CNS). Glioblastoma exhibits traits typical of immunotherapy-responsive tumors but deploys extensive immunosuppressive mechanisms, leveraging its CNS location. Overcoming intrinsic resistance, countering systemic immunosuppression, addressing adaptive resistance, and adapting to acquired resistance are crucial to dismantle GBM’s immunosuppressive machinery [3,111,112,113,114,115].
Intrinsic resistance in GBM is influenced by its molecular and clinical characteristics. Studies have revealed intratumor heterogeneity, where different subtypes coexist within the same tumor, complicating selective eradication of treatment-susceptible clones and paving the way for resistant ones. To navigate this complexity, immunotherapy aims to target multiple neoantigens derived from autologous tumor cells, minimizing the risk of antigenic overlap with normal tissue [113].
Immunotherapy’s success hinges on targeting neoantigens essential for tumor survival across GBM subtypes while sparing healthy tissue. Various approaches, such as OVs, CVs, and ICIs, address neoantigens effectively. However, CAR T- and NK-cell therapies require alternative strategies. The GBM microenvironment significantly contributes to immune evasion through immunosuppression. Microglia, the primary antigen-presenting cells in the CNS, promote tumor infiltration, progression, and invasiveness. Inflammatory cytokines secreted by microglia amplify GBM expansion. Moreover, the blood–brain barrier (BBB) typically prevents immune cell entry; however, GBM cells release chemotactic signals, such as CCL2, CCL5, CXCL, and SDF-1, to actively recruit tumor-associated macrophages (TAMs) across the BBB. TAMs in GBM are known to enhance immune checkpoints and support cancer stem cells. Inhibiting the activity of CCL2 and CCL5 has shown promise in reducing tumor migration and invasion [116,117].

3.3.2. Immunotherapeutic Strategies

In pursuit of eradicating GBM cells, particularly the therapy-resistant subset, immunotherapy aims to activate the patient’s antitumor immune response. Our review reveals a variety of immunotherapeutic strategies, such as CVs, OVs, ICIs, and CAR T cells, which are evaluated in clinical studies either individually or in conjunction with standard GBM treatments.

Immune Checkpoint Inhibitors

Immune checkpoint inhibitors are monoclonal antibodies designed to counteract negative regulatory pathways that impede T-cell activation. These antibodies target surface receptors known as immune checkpoints. Under normal circumstances, immune checkpoint molecules can dampen cytotoxic T-cell function. However, when ICIs are employed, they disrupt this normal activation of immune checkpoints, restoring T-cell function and enhancing the immunotherapeutic effect. Presently, the primary focus of immune checkpoint inhibition revolves around two key receptors: programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). Notable advancements have been made in treating challenging cancers, such as melanoma, lung cancer, and renal cancer, using anti-PD-1 and anti-PD-L1 antibodies [75,93,118,119,120,121,122].
The NCT02017717 trial, a pioneering randomized phase-I clinical study for recurrent GBM, assessed the efficacy and tolerability of nivolumab (a PD-1 inhibitor) as monotherapy or in combination with ipilimumab (a CTLA-4 inhibitor). All patients underwent surgical resections, RT, and TMZ treatments before being allocated to three different treatment arms. Intriguingly, nivolumab monotherapy demonstrated a superior median overall survival (10.4 months) compared to the NIVO1 + IPI3 or NIVO3 + IPI1 combinations (9.2 and 7.3 months, respectively). Phase III of this trial involved 369 recurrent GBM patients randomized to receive either nivolumab or bevacizumab. While mOS and toxicity were comparable, bevacizumab-treated patients exhibited a shorter duration of radiologic responses [118].
A phase-II clinical trial assessed bevacizumab alone or combined with notecan for efficacy in recurrent GBM patients, with both treatment arms showing tolerable side effects and median overall survival rates of 9.2 and 8.7 months, respectively.
At present, two ongoing phase-III trials, NCT02667587 and NCT02617589, are investigating nivolumab’s potential as a treatment for MGMT-unmethylated glioblastoma. The NCT02667587 clinical trial compares SOC with nivolumab or placebo, while NCT02617589 compares nivolumab versus TMZ, each in combination with RT.
Pembrolizumab, another anti-PD1 checkpoint inhibitor, is being investigated as a glioma treatment. A phase-II trial involving neoadjuvant pembrolizumab administration before surgery, followed by post-surgery adjuvant treatment, demonstrated increased survival in recurrent GBM patients.
CTLA-4, an immune checkpoint that competes with CD80 and CD86 for binding, thereby suppressing T-cell function, is under evaluation in clinical trials (NCT02311920, NCT02829931) using ipilimumab therapies.
Another immune checkpoint receptor, LAG-3, inhibits T-cell activity while enhancing the suppressive function of Tregs. A phase-I trial (NCT02658981) is currently assessing BMS-986,016, a LAG-3 inhibitor, alone and in combination with nivolumab in recurrent GBM patients. TIM-3, an additional lymphocyte-expressed receptor capable of inducing T-cell exhaustion and immune response suppression, can lead to unfavorable outcomes [4,92,118,119,120,121,122].

Cancer Vaccines

Vaccines hold promise as tumor treatment, leveraging tumor antigens. They activate immune surveillance against GBM, fortifying the adaptive immune system. GBM vaccines fall into four categories: peptides, DNA, cells, and mRNA. Peptide and DNA vaccines administer tumor-specific antigens or DNA to provoke an adaptive immune response. Cell vaccines, specifically DC vaccines derived from PBMCs, prime with tumor antigens. mRNA vaccines employ viral vectors loaded with mRNA, generating potent immune responses. Despite years of development, vaccines remain under investigation. Of these, only three have advanced to phase-III clinical trials: Rindopepimut, DCvax, and PPV.
Rindopepimut, a peptide-based vaccine, targets EGFRvIII mutation exclusively in GBM, minimizing off-tumor toxicity risk. However, GBM’s tumor heterogeneity poses challenges, as EGFRvIII expression varies. Phase-II trials demonstrated improved progression free survival (PFS) and median survival, compared to historical controls. ACT IV, a double-blind phase-III trial, examined Rindopepimut’s effect on GBM patients with minimal residual disease (MRD). MRD was defined as <2 cm2 of enhancing tumor tissue post-surgery and chemoradiotherapy. The trial showed no significant difference in overall survival for MRD patients. IDH1 peptide vaccines, targeting the IDH1R132 H mutation, are in phase-I trials following promising preclinical results. Survivin, highly expressed in GBM, led to the development of SurVaxM, a peptide vaccine. Early results from a phase-II study combining SurVaxM with TMZ show improved PFS and OS. An antisense oligodeoxynucleotide against the IGF type-I receptor (IMV-001) demonstrated improved PFS in phase-I trials. PPV immunotherapy displayed safety and efficacy in a phase-I trial for recurrent GBM patients. However, a phase-III trial comparing PPV to best supportive care yielded unfavorable results. Neoantigens, identified through DNA and RNA sequencing, offer a personalized approach to GBM treatment. Two clinical trials demonstrated the potential of personalized GBM vaccination.
In the GAPVAC study, 16 newly diagnosed GBM patients received two synthesized vaccines: one targeting unmutated peptides (APVAC1) and the other neoantigens (APVAC2). Both vaccines elicited CD4+ and CD8+ T-cell responses, with varying immunogenicity. APVAC1 induced 50% immunogenicity, while APVAC2 achieved 84.7%. APVAC1 primarily stimulated CD8+ T-cell responses, while APVAC2 focused on CD4+ T-cell responses. Median PFS and OS were encouraging.
Another study administered a neoantigen vaccine (NeoVax) to newly diagnosed and MGMT unmethylated GBM patients. Two patients displayed immunogenicity and both CD4+ and CD8+ T-cell responses. Median PFS reached 7.6 months, with a median OS of 16.8 months. DCVax-L, a personalized peptide vaccination, demonstrated potential in a phase-III trial when combined with standard GBM therapy. mRNA-transfected DC vaccines were well-tolerated and significantly extended PFS compared to controls in a phase-II trial. Autologous DCs, pulsed with autologous whole tumor lysate and combined with standard chemoradiotherapy, proved feasible and safe in newly diagnosed GBM patients. In contrast, a multicentric phase-II study involving tumor lysate-charged autologous DCs (Audencel) failed to improve clinical outcomes for newly diagnosed GBM patients. AV-GBM-1, autologous DCs loaded with tumor-associated antigens from short-term autologous tumor cell cultures, is being evaluated in an ongoing phase-II clinical trial.
Dendritic cells (DCs), capable of promoting adaptive antitumor immune responses, are ideal for cellular vaccination. DC vaccines demonstrated effectiveness in preclinical and early stage clinical trials, showing significantly longer overall survival in GBM patients. Intratumoral and intradermal administrations yielded better results than intradermal alone. ICT-107, a DC-based vaccine designed for newly diagnosed GBM patients, proved safe with encouraging median PFS and OS in phase-I trials. A phase-III trial comparing ICT-107 to standard care was suspended due to funding issues.
The landscape of GBM treatment is evolving, with vaccines offering a promising avenue for intervention. While many vaccine candidates are in development, only a select few have progressed to phase-III trials. The complex nature of GBM, including its heterogeneity, presents both challenges and opportunities in vaccine development. Neoantigens, identified through advanced sequencing techniques, offer a personalized approach that shows significant potential. Additionally, DC-based vaccines have demonstrated effectiveness in the preclinical models and early stage clinical trials, with some promising results for patients. The ongoing quest for effective GBM vaccines continues, with researchers exploring various strategies to harness the power of the immune system in the fight against this devastating disease [24,46,53,55,56,64,123,124,125,126,127,128,129,130,131,132,133].

Oncolytic Viruses

In recent years, OVs have emerged as a novel therapeutic approach in treating various solid tumors, including GBM. OVs offer a dual mechanism of antitumor action, involving the direct killing of tumor-specific cells and the induction of systemic antitumor immunity, encompassing both innate and adaptive responses. OVs trigger immunogenic cell death in tumor cells, leading to the release of tumor-associated antigens (TAAs), damage-associated molecular patterns (DAMPs), and pathogen-associated molecular patterns (PAMPs). DAMPs and PAMPs serve as potent stimulators of innate immunity by activating pattern recognition receptors, such as Toll-like receptors. Furthermore, these molecules enhance antigen cross-presentation and adaptive immune responses.
OVs also elicit a proinflammatory immune response, increasing the production of CXCL9, CXCL10, and CXCL11, which promote the trafficking and infiltration of T cells into tumors. Various OVs, including adenovirus, herpes simplex virus, measles virus, parvovirus, poliovirus, and zika virus, have demonstrated efficacy against GBM in preclinical studies. Encouraging data from clinical trials have shown OVs to have a favorable safety profile and promising efficacy, with evidence of intratumoral viral replication and lymphocyte infiltration. For instance, a phase-I clinical trial (NCT01470794) demonstrated the safety and efficacy of Toca 511 in treating 56 recurrent high-grade glioma patients. In a follow-up phase-III study involving 23 eligible patients, the median overall survival (OS) reached 14.4 months, with one- and two-year survival rates of 65.2% and 34.8%, respectively. Notably, five patients achieved complete responses and survived for extended periods after Toca 511 treatment.
DNX-2401, a tumor-selective oncolytic adenovirus, exhibited antiglioma efficacy in preclinical studies. In a phase-I dose-escalation trial involving 37 patients with recurrent malignant gliomas, DNX-2401 demonstrated both safety and promising responses across different dose levels. Approximately 20% of patients survived beyond three years post-treatment, and some patients experienced over a 95% reduction in tumor size with a progression free survival exceeding three years. Analyses of post-treatment tumor specimens revealed viral replication and spread within the tumor, along with the induction of intratumoral CD8+ and T-bet+ T-cell infiltration and a reduced expression of transmembrane immunoglobulin mucin-3.
Both Toca 511 and DNX-2401 trials reported that approximately 20% of GBM patients exhibited complete responses after intratumoral OV administration, with rare virotherapy-associated severe adverse events. Oncolytic H-1 parvovirus, although showing only a slight improvement in the median OS when administered intratumorally to GBM patients, displayed an increase in infiltrating lymphocytes and IFN-γ levels.
PVSRIPO, a live attenuated poliovirus type-1 vaccine with a modified internal ribosome entry site, received a breakthrough therapy designation from the FDA based on a phase-I study in recurrent GBM patients. PVSRIPO recognizes the poliovirus receptor CD155, which is upregulated on malignant and antigen-presenting cells within the tumor microenvironment. The phase-I study demonstrated safety and sustained survival rates, with about 20% of patients remaining alive for 57–70 months after PVSRIPO injection. A phase-II randomized trial of PVSRIPO alone or in combination with lomustine in patients with recurrent grade-IV malignant glioma (NCT02986178) is ongoing.
Herpes simplex virus-1 (HSV-1), a double-stranded DNA virus, has been extensively explored as a treatment for various solid tumors, including GBM. Genetically engineered variants, such as G207 and G47 delta, have shown promise. In a phase-I clinical trial involving children and adolescents with recurrent or progressive high-grade gliomas, G207 demonstrated safety and enhance the immunological response.
G47 delta (DELYTACT), another oncolytic HSV-1 variant, was evaluated in adult patients with residual or recurrent GBM in a single-arm phase-II clinical trial in Japan. Minimal side effects were observed, and the survival rate after one year reached an impressive 84.4%. G47 delta (Delytact/Teserpaturev) received conditional approval from the Japan Ministry of Health, Labor and Welfare (MHLW) for the treatment of malignant gliomas in Japan [49,54,55,56,66,68,134,135].
Several ongoing clinical trials are exploring the use of OVs as therapeutic agents for recurrent high-grade gliomas. While these results are promising, further clinical trials are needed to establish the safety and efficacy of OVs as a therapy for GBM.

CAR T Cell

CAR T-cell therapy presents a promising strategy for overcoming the formidable challenges posed by the BBB and the intricate tumor microenvironment (TME) within the context of adoptive cell T-cell therapy (ACT). ACT involves the reintroduction of autologous or allogenic anti-tumor T cells engineered to target tumor-specific antigens highly expressed on tumor cells while sparing normal cells. In a meticulously orchestrated process, these T cells are meticulously engineered in vitro using a lentiviral vector, thereby expressing a high-affinity single-chain fragment variable (scFv) tailored to the target antigen. This scFv is intricately fused with transmembrane regions, co-stimulatory domains, and an intracellular signaling domain derived from the CD3 molecule of the T-cell receptor (TCR). This engineering empowers autologous T cells to keenly recognize antigens upon scFv binding, initiating CAR molecule clustering and ensuing activation. This activation, in turn, triggers a cascade of events, including cytokine release, proliferation, cytotoxicity, and metabolic shifts, thereby enabling CAR T cells to chiefly exert their anti-tumor functions through cytokine release and the involvement of the granzyme and perforin axis, as well as the Fas and Fas ligand axis, thus equipping them to effectively surmount TME-related immunosuppression.
Notably, CAR T cells meticulously engineered to target IL13 Rα2 have demonstrated a heightened degree of selectivity for this receptor over IL13Ra1/IL4Ra. Intracranial injections of these IL13-zetakine CAR T cells, executed within a glioma xenograft model, have yielded auspicious improvements in median overall survival. A groundbreaking first-in-human pilot safety and feasibility trial conducted in 2015 evaluated the application of IL13-zetakine CAR T cells in three patients with recurrent GBM. These CAR T cells were methodically delivered via an implanted reservoir/catheter system, directly into the resection cavity, thereby resulting in a precisely controlled, transient bout of brain inflammation. Although the CAR T cells were met with overall favorable tolerability, certain adverse events, including grade-3 headaches and transient grade-3 neurologic events, were observed. A subsequent analysis of tumor tissue indicated a tangible reduction in IL13 Rα2 expression, while an MRI analysis suggested an increase in tumor necrotic volume, thereby potentially extending overall survival. Following this pioneering trial, a second-generation IL13-zetakine CAR T cell was painstakingly developed, featuring a 4-1BB costimulatory domain and an ingeniously mutated IgG4-Fc linker, all designed to amplify antitumor potential while curbing off-target interactions. A patient afflicted with recurrent GBM subsequently received intracavitary infusions of these CAR T cells, resulting in a pronounced inhibition of local tumor progression. However, the emergence of novel intracranial tumors and spinal lesions was noted, with the fifth intraventricular infusion precipitating a substantial reduction in tumor size. Regrettably, this regression proved transient, lasting merely 7.5 months, and recurrence subsequently manifested at new locations, characterized by diminished IL13 Rα2 expression, thereby underscoring the indispensable need for combinatory therapeutic modalities aimed at addressing antigen-loss relapse.
EGFRvIII, a frequently encountered GBM mutation, was initially associated with abbreviated survival periods. In a phase-I clinical trial, 18 patients afflicted with recurrent GBM received third-generation EGFRvIII-directed CAR T cells, endowed with CD28 and 4-1BB costimulatory domains. Patients, upon infusion of these CAR T cells, expeditiously developed respiratory symptoms, particularly evident at higher dose levels, a phenomenon indicative of dose-limiting toxicity, despite the absence of discernible clinical benefits. The median overall survival rate reached 6.9 months, with select patients achieving survival exceeding a year. To effectively address instances of EGFRvIII-negative yet EGFR-positive glioblastomas, a novel approach surfaced, involving the fusion of EGFRvIII scFv with a bispecific T-cell engager (BiTE). These BiTEs, as synthetic bispecific antibodies, serve to heighten immune interactions while simultaneously augmenting antibody specificity. Recent investigations have explored SynNotch-CAR T cells, meticulously designed to target multiple antigens, thus yielding improvements in specificity, comprehensiveness, and longevity in comparison to conventional T-cell therapy, as employed in the context of glioblastoma.
B7-H3, alternatively known as CD276, plays a pivotal role in regulating T-cell functions and, notably, is found to be overexpressed across a diverse array of human cancer cells, a correlation that frequently coincides with negative clinical outcomes. This distinct profile renders it an enticing target for immunotherapeutic interventions. Nevertheless, significant challenges persist, not the least of which is the full elucidation of its receptor and the intricacies of immune regulation associated therewith.
HER2, conspicuously expressed in various CNS tumors, including GBM, yet conspicuously absent in normal CNS tissue, emerges as an attractive and distinctively selective target. Clinical trials employing HER2-specific CAR T cells have borne witness to well-tolerated treatments, despite the lack of substantial and tangible survival benefits. Recent preclinical inquiries have embarked upon the exploration of trivalent CAR T-cell therapy, an innovative approach that targets HER2, IL13 Rα2, and ephrin-A2, demonstrating encouraging improvements in terms of cytokine release and cytotoxicity when contrasted with the outcomes achieved using monospecific or bispecific CAR T cells, thereby strongly hinting at the potential utility of this multifaceted therapeutic approach within the context of glioblastoma therapy.
CAR T-cell therapy, undoubtedly, occupies a paramount and promising position in the realm of glioblastoma immunotherapy. Recent strides and advancements, such as the development of second-generation CAR T cells with heightened specificity and reduced off-target effects, serve as a testament to the adaptability and burgeoning potential of this therapeutic modality. Strikingly promising results have emerged from studies targeting antigens, such as IL13 Rα2 and EGFRvIII, as reflected in the significant tumor regression and improved patient survival rates. Moreover, the visionary concept of trivalent CAR T cells augurs a future where combination therapies that adeptly target multiple antigens may yield even more favorable treatment outcomes. Though formidable challenges persist, encompassing the phenomena of antigen loss and the imperative [74,136,137,138,139,140,141,142,143,144,145,146,147,148,149].

4. Conclusions

Our comprehensive analysis of the immunotherapy strategies for GBM treatment highlights the evolving landscape of therapeutic interventions for this challenging disease. A diverse range of approaches, including CAR T cells, ICIs, OVs, and CVs, are being explored to address the unique challenges presented by GBM. To develop a curative immunotherapy for GBM, it is crucial to overcome immunotolerance, stimulate robust responses to tumor antigens, and effectively counter the evolving escape mechanisms within the immunosuppressive microenvironment.
Immune checkpoint inhibitors, such as PD-1 and CTLA-4 inhibitors, have demonstrated promise in the clinical trials, and the ongoing research continues to seek ways to enhance their effectiveness. Cancer vaccines, particularly those targeting neoantigens, offer a personalized approach with significant potential. Oncolytic viruses have shown dual mechanisms of action and have displayed promise in clinical trials, with some achieving breakthrough status. CAR T-cell therapy shines as a beacon of hope. These cells, intricately engineered to target specific antigens, have produced encouraging results, especially when targeting IL13 Rα2 and EGFRvIII. The development of second-generation CAR T cells with improved specificity and reduced off-target effects exemplifies the adaptability and potential of this approach.
Despite this promising data from published and ongoing studies on GBM immunotherapies, the considerable variability in these results and the multitude of therapeutic targets make it challenging to reach consensus. Therefore, translating these results into clinical success for GBM patients remains a formidable task. Nevertheless, ongoing studies are actively exploring combination approaches and enhancing existing immunotherapeutic strategies, offering hope for a revolutionary breakthrough in cancer care.

Author Contributions

Conceptualization, E.A., M.Z., M.M.F. and P.P.P.; methodology, E.A., M.Z., C.T. and P.P.P.; validation, E.A., M.Z., L.D.M., C.T., T.I., M.M.F. and P.P.P.; formal analysis, E.A., C.T. and P.P.P.; investigation, E.A., C.T. and P.P.P.; resources, E.A., C.T. and P.P.P.; data curation, E.A. and P.P.P.; writing—original draft preparation, E.A. and P.P.P.; writing—review and editing, E.A., M.Z., L.D.M., C.T., T.I., M.M.F. and P.P.P.; visualization, E.A., M.Z., L.D.M., C.T., T.I., M.M.F. and P.P.P.; supervision, E.A., M.Z., T.I., M.M.F. and P.P.P.; project administration, E.A., M.Z., M.M.F. and P.P.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data are available from a publicly accessible repository.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AA = anaplastic astrocytoma, Ab = antibody, ACT = adoptive cellular therapy, AE = adverse event, AFTV = autologous formalin-fixed tumor vaccine, ATV = autologous tumor vaccine, ALECSAT = autologous lymphoid effector cells specific against tumor, CAR T = chimeric antigen receptor T cell, CHT = chemotherapy, CpG-ODN = oligodeoxynucleotide containing unmethylated cytosine-guanosine motif, CSC = cancer stem cell, CV = cancer vaccine, d = day, DC = dendritic cell, DIPG = diffuse intrinsic pontine glioma, DLT = dose-limiting toxicity, DMG = diffuse midline glioma, DCV = dendritic cell vaccine, EBRT = external beam radiotherapy, FC = fusion cell, FRT = fractionated radiotherapy, FSRT = fractionated stereotactic radiotherapy, GAAs = glioma-associated antigens, GBM = glioblastoma multiforme, GMCI = gene-mediated cytotoxic immunotherapy, GSC = glioma stem cell, HGG = high-grade glioma, HSPPC-96 = heat-shock protein 96—peptide complex, HUVEC = human umbilical vein endothelial cell, IC = intracranial, ICI = immune checkpoint inhibitors, mo = month, MTD = maximum tolerated dose, nGBM = newly diagnosed glioblastoma multiforme, N/A = not applicable, NDV = Newcastle disease virus, ORR = overall response rate, OS = overall survival, OV= oncolytic virus, PBMC = peripheral blood mononuclear cell, PD = progressive disease, PFS = progression free survival, rGBM = recurrent glioblastoma multiforme, RCHT = radiochemotherapy, RIT = radioimmunotherapy, RT = radiotherapy, SOC = standard of care treatment (RT + TMZ), TMZ = temozolomide, TT = tetanus toxoid, VDX = Veledimex, wk = week, y = year.

Appendix A

Figure A1. Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Review (PRISMA-ScR) checklist.
Figure A1. Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Review (PRISMA-ScR) checklist.
Ijms 24 15037 g0a1

References

  1. Yuan, B.; Wang, G.; Tang, X.; Tong, A.; Zhou, L. Immunotherapy of glioblastoma: Recent advances and future prospects. Hum. Vaccin. Immunother. 2022, 18, 2055417. [Google Scholar] [CrossRef]
  2. Vik-Mo, E.O.; Nyakas, M.; Mikkelsen, B.V.; Moe, M.C.; Due-Tønnesen, P.; Suso, E.M.; Sæbøe-Larssen, S.; Sandberg, C.; Brinchmann, J.E.; Helseth, E.; et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol. Immunother. 2013, 62, 1499–1509. [Google Scholar] [CrossRef]
  3. Markovic, D.S.; Glass, R.; Synowitz, M.; van Rooijen, N.; Kettenmann, H. Microglia stimulate the invasiveness of glioma cells by increasing the activity of metalloprotease-2. J. Neuropathol. Exp. Neurol. 2005, 64, 754–762. [Google Scholar] [CrossRef] [PubMed]
  4. Wahyuhadi, J.; Immadoel Haq, I.B.; Arifianto, M.R. Active Immunotherapy for Glioblastoma Treatment: A Systematic Review and Meta-Analysis. Cancer Control 2022, 29, 10732748221079474. [Google Scholar] [CrossRef] [PubMed]
  5. Agosti, E.; Panciani, P.P.; Zeppieri, M.; De Maria, L.; Pasqualetti, F.; Tel, A.; Zanin, L.; Fontanella, M.M.; Ius, T. Tumor Microenvironment and Glioblastoma Cell Interplay as Promoters of Therapeutic Resistance. Biology 2023, 12, 736. [Google Scholar] [CrossRef] [PubMed]
  6. Majc, B.; Novak, M.; Kopitar-Jerala, N.; Jewett, A.; Breznik, B. Immunotherapy of Glioblastoma: Current Strategies and Challenges in Tumor Model Development. Cells 2021, 10, 265. [Google Scholar] [CrossRef]
  7. Bausart, M.; Préat, V.; Malfanti, A. Immunotherapy for glioblastoma: The promise of combination strategies. J. Exp. Clin. Cancer Res. 2022, 41, 35. [Google Scholar] [CrossRef] [PubMed]
  8. Mende, A.L.; Schulte, J.D.; Okada, H.; Clarke, J.L. Current Advances in Immunotherapy for Glioblastoma. Curr. Oncol. Rep. 2021, 23, 21. [Google Scholar] [CrossRef]
  9. Chowdhury, S.; Bappy, M.H.; Clocchiatti-Tuozzo, S.; Cheeti, S.; Chowdhury, S.; Patel, V. Current Advances in Immunotherapy for Glioblastoma Multiforme and Future Prospects. Cureus 2021, 13, e20604. [Google Scholar] [CrossRef]
  10. Mahmoud, A.B.; Ajina, R.; Aref, S.; Darwish, M.; Alsayb, M.; Taher, M.; AlSharif, S.A.; Hashem, A.M.; Alkayyal, A.A. Advances in immunotherapy for glioblastoma multiforme. Front. Immunol. 2022, 13, 944452. [Google Scholar] [CrossRef]
  11. Hanaei, S.; Afshari, K.; Hirbod-Mobarakeh, A.; Mohajer, B.; Amir Dastmalchi, D.; Rezaei, N. Therapeutic efficacy of specific immunotherapy for glioma: A systematic review and meta-analysis. Rev. Neurosci. 2018, 29, 443–461. [Google Scholar] [CrossRef] [PubMed]
  12. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  13. Stang, A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur. J. Epidemiol. 2010, 25, 603–605. [Google Scholar] [CrossRef] [PubMed]
  14. Riva, P.; Arista, A.; Tison, V.; Sturiale, C.; Franceschi, G.; Spinelli, A.; Riva, N.; Casi, M.; Moscatelli, G.; Frattarelli, M. Intralesional radioimmunotherapy of malignant gliomas. An effective treatment in recurrent tumors. Cancer 1994, 73 (Suppl. S3), 1076–1082. [Google Scholar] [CrossRef] [PubMed]
  15. Riva, P.; Arista, A.; Franceschi, G.; Frattarelli, M.; Sturiale, C.; Riva, N.; Casi, M.; Rossitti, R. Local treatment of malignant gliomas by direct infusion of specific monoclonal antibodies labeled with 131I: Comparison of the results obtained in recurrent and newly diagnosed tumors. Cancer Res. 1995, 55 (Suppl. S23), 5952s–5956s. [Google Scholar] [PubMed]
  16. Pöpperl, G.; Götz, C.; Gildehaus, F.J.; Yousry, T.A.; Reulen, H.J.; Hahn, K.; Tatsch, K. [Initial experiences with adjuvant locoregional radioimmunotherapy using 131I-labeled monoclonal antibodies against tenascin (BC-4) for treatment of glioma (WHO III and IV)]. Nuklearmedizin 2002, 41, 120–128. [Google Scholar]
  17. Fukushima, T.; Yamamoto, M.; Oshiro, S.; Tsugu, H.; Hirakawa, K.; Soma, G.I. Recombinant mutant human tumor necrosis factor-alpha (TNF-SAM2) immunotherapy with ranimustine chemotherapy and concurrent radiation therapy for malignant astrocytomas. Anticancer Res. 2003, 23, 4473–4481. [Google Scholar]
  18. Yu, J.S.; Liu, G.; Ying, H.; Yong, W.H.; Black, K.L.; Wheeler, C.J. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res. 2004, 64, 4973–4979. [Google Scholar] [CrossRef]
  19. Steiner, H.H.; Bonsanto, M.M.; Beckhove, P.; Brysch, M.; Geletneky, K.; Ahmadi, R.; Schuele-Freyer, R.; Kremer, P.; Ranaie, G.; Matejic, D.; et al. Antitumor vaccination of patients with glioblastoma multiforme: A pilot study to assess feasibility, safety, and clinical benefit. J. Clin. Oncol. 2004, 22, 4272–4281. [Google Scholar] [CrossRef]
  20. Yamanaka, R.; Homma, J.; Yajima, N.; Tsuchiya, N.; Sano, M.; Kobayashi, T.; Yoshida, S.; Abe, T.; Narita, M.; Takahashi, M.; et al. Clinical evaluation of dendritic cell vaccination for patients with recurrent glioma: Results of a clinical phase I/II trial. Clin. Cancer Res. 2005, 11, 4160–4167. [Google Scholar] [CrossRef]
  21. De Vleeschouwer, S.; Fieuws, S.; Rutkowski, S.; Van Calenbergh, F.; Van Loon, J.; Goffin, J.; Sciot, R.; Wilms, G.; Demaerel, P.; Warmuth-Metz, M.; et al. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin. Cancer Res. 2008, 14, 3098–3104. [Google Scholar] [CrossRef] [PubMed]
  22. Izumoto, S.; Tsuboi, A.; Oka, Y.; Suzuki, T.; Hashiba, T.; Kagawa, N.; Hashimoto, N.; Maruno, M.; Elisseeva, O.A.; Shirakata, T.; et al. Phase II clinical trial of Wilms tumor 1 peptide vaccination for patients with recurrent glioblastoma multiforme. J. Neurosurg. 2008, 108, 963–971. [Google Scholar] [CrossRef] [PubMed]
  23. Ardon, H.; Van Gool, S.; Lopes, I.S.; Maes, W.; Sciot, R.; Wilms, G.; Demaerel, P.; Bijttebier, P.; Claes, L.; Goffin, J.; et al. Integration of autologous dendritic cell-based immunotherapy in the primary treatment for patients with newly diagnosed glioblastoma multiforme: A pilot study. J. Neurooncol. 2010, 99, 261–272. [Google Scholar] [CrossRef]
  24. Sampson, J.H.; Heimberger, A.B.; Archer, G.E.; Aldape, K.D.; Friedman, A.H.; Friedman, H.S.; Gilbert, M.R.; Herndon, J.E.; McLendon, R.E.; Mitchell, D.A.; et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 2010, 28, 4722–4729. [Google Scholar] [CrossRef] [PubMed]
  25. Okada, H.; Kalinski, P.; Ueda, R.; Hoji, A.; Kohanbash, G.; Donegan, T.E.; Mintz, A.H.; Engh, J.A.; Bartlett, D.L.; Brown, C.K.; et al. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J. Clin. Oncol. 2011, 29, 330–336. [Google Scholar] [CrossRef] [PubMed]
  26. Prins, R.M.; Soto, H.; Konkankit, V.; Odesa, S.K.; Eskin, A.; Yong, W.H.; Nelson, S.F.; Liau, L.M. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin. Cancer Res. 2011, 17, 1603–1615. [Google Scholar] [CrossRef]
  27. Akiyama, Y.; Oshita, C.; Kume, A.; Iizuka, A.; Miyata, H.; Komiyama, M.; Ashizawa, T.; Yagoto, M.; Abe, Y.; Mitsuya, K.; et al. α-type-1 polarized dendritic cell-based vaccination in recurrent high-grade glioma: A phase I clinical trial. BMC Cancer 2012, 12, 623. [Google Scholar] [CrossRef]
  28. Valle, R.D.; de Cerio, A.L.D.; Inoges, S.; Tejada, S.; Pastor, F.; Villanueva, H.; Gallego, J.; Espinos, J.; Aristu, J.; Idoate, M.A.; et al. Dendritic cell vaccination in glioblastoma after fluorescence-guided resection. World J. Clin. Oncol. 2012, 3, 142–149. [Google Scholar] [CrossRef]
  29. Cho, D.Y.; Yang, W.K.; Lee, H.C.; Hsu, D.M.; Lin, H.L.; Lin, S.Z.; Chen, C.C.; Harn, H.J.; Liu, C.L.; Lee, W.Y.; et al. Adjuvant immunotherapy with whole-cell lysate dendritic cells vaccine for glioblastoma multiforme: A phase II clinical trial. World Neurosurg. 2012, 77, 736–744. [Google Scholar] [CrossRef]
  30. Crane, C.A.; Han, S.J.; Ahn, B.; Oehlke, J.; Kivett, V.; Fedoroff, A.; Butowski, N.; Chang, S.M.; Clarke, J.; Berger, M.S.; et al. Individual patient-specific immunity against high-grade glioma after vaccination with autologous tumor derived peptides bound to the 96 KD chaperone protein. Clin. Cancer Res. 2013, 19, 205–214. [Google Scholar] [CrossRef]
  31. Phuphanich, S.; Wheeler, C.J.; Rudnick, J.D.; Mazer, M.; Wang, H.; Nuño, M.A.; Richardson, J.E.; Fan, X.; Ji, J.; Chu, R.M.; et al. Phase I trial of a multi-epitope-pulsed dendritic cell vaccine for patients with newly diagnosed glioblastoma. Cancer Immunol. Immunother. 2013, 62, 125–135. [Google Scholar] [CrossRef] [PubMed]
  32. Tanaka, M.; Tsuno, N.H.; Fujii, T.; Todo, T.; Saito, N.; Takahashi, K. Human umbilical vein endothelial cell vaccine therapy in patients with recurrent glioblastoma. Cancer Sci. 2013, 104, 200–205. [Google Scholar] [CrossRef] [PubMed]
  33. Pellegatta, S.; Eoli, M.; Frigerio, S.; Antozzi, C.; Bruzzone, M.G.; Cantini, G.; Nava, S.; Anghileri, E.; Cuppini, L.; Cuccarini, V.; et al. The natural killer cell response and tumor debulking are associated with prolonged survival in recurrent glioblastoma patients receiving dendritic cells loaded with autologous tumor lysates. Oncoimmunology 2013, 2, e23401. [Google Scholar] [CrossRef]
  34. Pollack, I.F.; Jakacki, R.I.; Butterfield, L.H.; Hamilton, R.L.; Panigrahy, A.; Potter, D.M.; Connelly, A.K.; Dibridge, S.A.; Whiteside, T.L.; Okada, H.; et al. Antigen-specific immune responses and clinical outcome after vaccination with glioma-associated antigen peptides and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in children with newly diagnosed malignant brainstem and nonbrainstem gliomas. J. Clin. Oncol. 2014, 32, 2050–2058. [Google Scholar] [CrossRef]
  35. Schuessler, A.; Smith, C.; Beagley, L.; Boyle, G.M.; Rehan, S.; Matthews, K.; Jones, L.; Crough, T.; Dasari, V.; Klein, K.; et al. Autologous T-cell therapy for cytomegalovirus as a consolidative treatment for recurrent glioblastoma. Cancer Res. 2014, 74, 3466–3476. [Google Scholar] [CrossRef]
  36. Bloch, O.; Crane, C.A.; Fuks, Y.; Kaur, R.; Aghi, M.K.; Berger, M.S.; Butowski, N.A.; Chang, S.M.; Clarke, J.L.; McDermott, M.W.; et al. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: A phase II, single-arm trial. Neuro Oncol. 2014, 16, 274–279. [Google Scholar] [CrossRef] [PubMed]
  37. Ishikawa, E.; Muragaki, Y.; Yamamoto, T.; Maruyama, T.; Tsuboi, K.; Ikuta, S.; Hashimoto, K.; Uemae, Y.; Ishihara, T.; Matsuda, M.; et al. Phase I/IIa trial of fractionated radiotherapy, temozolomide, and autologous formalin-fixed tumor vaccine for newly diagnosed glioblastoma. J. Neurosurg. 2014, 121, 543–553. [Google Scholar] [CrossRef]
  38. Hashimoto, N.; Tsuboi, A.; Kagawa, N.; Chiba, Y.; Izumoto, S.; Kinoshita, M.; Kijima, N.; Oka, Y.; Morimoto, S.; Nakajima, H.; et al. Wilms tumor 1 peptide vaccination combined with temozolomide against newly diagnosed glioblastoma: Safety and impact on immunological response. Cancer Immunol. Immunother. 2015, 64, 707–716. [Google Scholar] [CrossRef]
  39. Schijns, V.E.J.C.; Pretto, C.; Devillers, L.; Pierre, D.; Hofman, F.M.; Chen, T.C.; Mespouille, P.; Hantos, P.; Glorieux, P.; Bota, D.A.; et al. First clinical results of a personalized immunotherapeutic vaccine against recurrent, incompletely resected, treatment-resistant glioblastoma multiforme (GBM) tumors, based on combined allo- and auto-immune tumor reactivity. Vaccine 2015, 33, 2690–2696. [Google Scholar] [CrossRef]
  40. Sakai, K.; Shimodaira, S.; Maejima, S.; Udagawa, N.; Sano, K.; Higuchi, Y.; Koya, T.; Ochiai, T.; Koide, M.; Uehara, S.; et al. Dendritic cell-based immunotherapy targeting Wilms’ tumor 1 in patients with recurrent malignant glioma. J. Neurosurg. 2015, 123, 989–997. [Google Scholar] [CrossRef]
  41. Kalkanis, S.N.; Aghi, M.K.; Cloughsy, T.F.; Kaptain, G.; Portnow, J.; Vogelbaum, M.A.; Kesari, S.; Mikkelsen, T.; Elder, J.B.; Chen, C.C.; et al. Ddel-06preliminary Safety of Toca 511, A Retroviral Replicating Vector, In Patients with Recurrent High Grade Glioma across Three Separate Phase 1 Studies. Neuro-Oncology 2015, 17 (Suppl. S5), v74. [Google Scholar] [CrossRef]
  42. Westphal, M.; Heese, O.; Steinbach, J.P.; Schnell, O.; Schackert, G.; Mehdorn, M.; Schulz, D.; Simon, M.; Schlegel, U.; Senft, C.; et al. A randomised, open label phase III trial with nimotuzumab, an anti-epidermal growth factor receptor monoclonal antibody in the treatment of newly diagnosed adult glioblastoma. Eur. J. Cancer 2015, 51, 522–532. [Google Scholar] [CrossRef] [PubMed]
  43. Akasaki, Y.; Kikuchi, T.; Homma, S.; Koido, S.; Ohkusa, T.; Tasaki, T.; Hayashi, K.; Komita, H.; Watanabe, N.; Suzuki, Y.; et al. Phase I/II trial of combination of temozolomide chemotherapy and immunotherapy with fusions of dendritic and glioma cells in patients with glioblastoma. Cancer Immunol. Immunother. 2016, 65, 1499–1509. [Google Scholar] [CrossRef] [PubMed]
  44. Brown, C.E.; Alizadeh, D.; Starr, R.; Weng, L.; Wagner, J.R.; Naranjo, A.; Ostberg, J.R.; Blanchard, M.S.; Kilpatrick, J.; Simpson, J.; et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N. Engl. J. Med. 2016, 375, 2561–2569. [Google Scholar] [CrossRef] [PubMed]
  45. Cloughesy, T.F.; Landolfi, J.; Hogan, D.J.; Bloomfield, S.; Carter, B.; Chen, C.C.; Elder, J.B.; Kalkanis, S.N.; Kesari, S.; Lai, A.; et al. Phase 1 trial of vocimagene amiretrorepvec and 5-fluorocytosine for recurrent high-grade glioma. Sci. Transl. Med. 2016, 8, 341ra75. [Google Scholar] [CrossRef]
  46. Fenstermaker, R.A.; Ciesielski, M.J.; Qiu, J.; Yang, N.; Frank, C.L.; Lee, K.P.; Mechtler, L.R.; Belal, A.; Ahluwalia, M.S.; Hutson, A.D. Clinical study of a survivin long peptide vaccine (SurVaxM) in patients with recurrent malignant glioma. Cancer Immunol. Immunother. 2016, 65, 1339–1352. [Google Scholar] [CrossRef]
  47. Oji, Y.; Hashimoto, N.; Tsuboi, A.; Murakami, Y.; Iwai, M.; Kagawa, N.; Chiba, Y.; Izumoto, S.; Elisseeva, O.; Ichinohasama, R.; et al. Association of WT1 IgG antibody against WT1 peptide with prolonged survival in glioblastoma multiforme patients vaccinated with WT1 peptide. Int. J. Cancer 2016, 139, 1391–1401. [Google Scholar] [CrossRef]
  48. Wheeler, L.A.; Manzanera, A.G.; Bell, S.D.; Cavaliere, R.; McGregor, J.M.; Grecula, J.C.; Newton, H.B.; Lo, S.S.; Badie, B.; Portnow, J.; et al. Phase II multicenter study of gene-mediated cytotoxic immunotherapy as adjuvant to surgical resection for newly diagnosed malignant glioma. Neuro Oncol. 2016, 18, 1137–1145. [Google Scholar] [CrossRef]
  49. Alonso, M.M.; García-Moure, M.; Gonzalez-Huarriz, M.; Marigil, M.; Hernandez-Alcoceba, R.; Buñales, M.; Hervás, S.; Gallego, J.; Gomez-Manzano, C.; Fueyo, J.; et al. Abstract CT027: Oncolytic virus DNX-2401 with a short course of temozolomide for glioblastoma at first recurrence: Clinical data and prognostic biomarkers. Cancer Res. 2017, 77 (Suppl. S13), CT027. [Google Scholar] [CrossRef]
  50. O’Rourke, D.M.; Nasrallah, M.P.; Desai, A.; Melenhorst, J.J.; Mansfield, K.; Morrissette, J.J.D.; Martinez-Lage, M.; Brem, S.; Maloney, E.; Shen, A.; et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 2017, 9, eaaa0984. [Google Scholar] [CrossRef]
  51. Kong, D.S.; Nam, D.H.; Kang, S.H.; Lee, J.W.; Chang, J.H.; Kim, J.H.; Lim, Y.J.; Koh, Y.C.; Chung, Y.G.; Kim, J.M.; et al. Phase III randomized trial of autologous cytokine-induced killer cell immunotherapy for newly diagnosed glioblastoma in Korea. Oncotarget 2017, 8, 7003–7013. [Google Scholar] [CrossRef] [PubMed]
  52. Ursu, R.; Carpentier, A.; Metellus, P.; Lubrano, V.; Laigle-Donadey, F.; Capelle, L.; Guyotat, J.; Langlois, O.; Bauchet, L.; Desseaux, K.; et al. Intracerebral injection of CpG oligonucleotide for patients with de novo glioblastoma-A phase II multicentric, randomised study. Eur. J. Cancer 2017, 73, 30–37. [Google Scholar] [CrossRef] [PubMed]
  53. Inogés, S.; Tejada, S.; de Cerio, A.L.D. A phase II trial of autologous dendritic cell vaccination and radiochemotherapy following fluorescence-guided surgery in newly diagnosed glioblastoma patients. J. Transl. Med. 2017, 15, 104. [Google Scholar] [CrossRef] [PubMed]
  54. Geletneky, K.; Hajda, J.; Angelova, A.L. Oncolytic H-1 Parvovirus Shows Safety and Signs of Immunogenic Activity in a First Phase I/IIa Glioblastoma Trial. Mol. Ther. 2017, 25, 2620–2634. [Google Scholar] [CrossRef] [PubMed]
  55. Weller, M.; Butowski, N.; Tran, D.D. Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef]
  56. Zadeh, G.; Lang, F.; Daras, M. Atim-24. Interim Results of a Phase II Multicenter Study of The Conditionally Replicative Oncolytic Adenovirus Dnx-2401 with Pembrolizumab (Keytruda) for Recurrent Glioblastoma; Captive Study (Keynote-192). Neuro Oncol. 2018, 20 (Suppl. S6), vi6. [Google Scholar] [CrossRef]
  57. Phase 2 trial of SL-701 in relapsed/refractory (r/r) glioblastoma (GBM): Correlation of immune response with longer-term survival. J. Clin. Oncol. 2018, 36 (Suppl. S15), 2058. [CrossRef]
  58. Cloughesy, T.F.; Landolfi, J.; Vogelbaum, M.A. Durable complete responses in some recurrent high-grade glioma patients treated with Toca 511 + Toca FC. Neuro Oncol. 2018, 20, 1383–1392. [Google Scholar] [CrossRef]
  59. Fried, I.; Lossos, A.; Ben Ami, T. Preliminary results of immune modulating antibody MDV9300 (pidilizumab) treatment in children with diffuse intrinsic pontine glioma. J. Neurooncol. 2018, 136, 189–195. [Google Scholar] [CrossRef]
  60. Pellegatta, S.; Eoli, M.; Cuccarini, V. Survival gain in glioblastoma patients treated with dendritic cell immunotherapy is associated with increased NK but not CD8+ T cell activation in the presence of adjuvant temozolomide. Oncoimmunology 2018, 7, e1412901. [Google Scholar] [CrossRef]
  61. Yao, Y.; Luo, F.; Tang, C. Molecular subgroups and B7-H4 expression levels predict responses to dendritic cell vaccines in glioblastoma: An exploratory randomized phase II clinical trial. Cancer Immunol. Immunother. 2018, 67, 1777–1788. [Google Scholar] [CrossRef] [PubMed]
  62. Wick, W.; van den Bent, M.J. First results on the DCVax phase III trial: Raising more questions than providing answers. Neuro Oncol. 2018, 20, 1283–1284. [Google Scholar] [CrossRef] [PubMed]
  63. Desjardins, A.; Gromeier, M.; Herndon, J.E. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N. Engl. J. Med. 2018, 379, 150–161. [Google Scholar] [CrossRef] [PubMed]
  64. Buchroithner, J.; Erhart, F.; Pichler, J. Audencel Immunotherapy Based on Dendritic Cells Has No Effect on Overall and Progression-Free Survival in Newly Diagnosed Glioblastoma: A Phase II Randomized Trial. Cancers 2018, 10, 372. [Google Scholar] [CrossRef] [PubMed]
  65. Bota, D.A.; Chung, J.; Dandekar, M. Phase II study of ERC1671 plus bevacizumab versus bevacizumab plus placebo in recurrent glioblastoma: Interim results and correlations with CD4+ T-lymphocyte counts. CNS Oncol. 2018, 7, CNS22. [Google Scholar] [CrossRef]
  66. Phase I Study of DNX-2401 (Delta-24-RGD) Oncolytic Adenovirus: Replication and Immunotherapeutic Effects in Recurrent Malignant Glioma—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/29432077/ (accessed on 12 September 2023).
  67. Kieran, M.W.; Goumnerova, L.; Manley, P. Phase I study of gene-mediated cytotoxic immunotherapy with AdV-tk as adjuvant to surgery and radiation for pediatric malignant glioma and recurrent ependymoma. Neuro Oncol. 2019, 21, 537–546. [Google Scholar] [CrossRef]
  68. Todo, T.; Ino, Y.; Ohtsu, H.; Shibahara, J.; Tanaka, M. A phase I/II study of triple-mutated oncolytic herpes virus G47∆ in patients with progressive glioblastoma. Nat. Commun. 2022, 13, 4119. [Google Scholar] [CrossRef]
  69. Wen, P.Y.; Reardon, D.A.; Armstrong, T.S. A Randomized Double-Blind Placebo-Controlled Phase II Trial of Dendritic Cell Vaccine ICT-107 in Newly Diagnosed Patients with Glioblastoma. Clin. Cancer Res. 2019, 25, 5799–5807. [Google Scholar] [CrossRef]
  70. Chiocca, E.A.; Yu, J.S.; Lukas, R.V. Regulatable interleukin-12 gene therapy in patients with recurrent high-grade glioma: Results of a phase 1 trial. Sci. Transl. Med. 2019, 11, eaaw5680. [Google Scholar] [CrossRef]
  71. Cloughesy, T.; Petrecca, K.; Walbert, T. Ltbk-08. Toca 511 & Toca Fc versus Standard of Care in Patients with Recurrent High Grade Glioma. Neuro Oncol. 2019, 21 (Suppl. S6), vi284. [Google Scholar] [CrossRef]
  72. Migliorini, D.; Dutoit, V.; Allard, M. Phase I/II trial testing safety and immunogenicity of the multipeptide IMA950/poly-ICLC vaccine in newly diagnosed adult malignant astrocytoma patients. Neuro Oncol. 2019, 21, 923–933. [Google Scholar] [CrossRef] [PubMed]
  73. Eoli, M.; Corbetta, C.; Anghileri, E. Expansion of effector and memory T cells is associated with increased survival in recurrent glioblastomas treated with dendritic cell immunotherapy. Neuro-Oncol. Adv. 2019, 1, vdz022. [Google Scholar] [CrossRef] [PubMed]
  74. Goff, S.L.; Morgan, R.A.; Yang, J.C. Pilot Trial of Adoptive Transfer of Chimeric Antigen Receptor-transduced T Cells Targeting EGFRvIII in Patients with Glioblastoma. J. Immunother. 2019, 42, 126–135. [Google Scholar] [CrossRef] [PubMed]
  75. Cloughesy, T.F.; Mochizuki, A.Y.; Orpilla, J.R. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat. Med. 2019, 25, 477–486. [Google Scholar] [CrossRef] [PubMed]
  76. Cloughesy, T.F.; Brenner, A.; de Groot, J.F. A randomized controlled phase III study of VB-111 combined with bevacizumab vs bevacizumab monotherapy in patients with recurrent glioblastoma (GLOBE). Neuro-Oncology 2020, 22, 705–717. [Google Scholar] [CrossRef]
  77. Mueller, S.; Taitt, J.M.; Villanueva-Meyer, J.E. Mass cytometry detects H3.3K27M-specific vaccine responses in diffuse midline glioma. J. Clin. Investig. 2020, 130, 6325–6337. [Google Scholar] [CrossRef] [PubMed]
  78. Mishinov, S.V.; Budnik, A.Y.; Stupak, V.V. Autologous and Pooled Tumor Lysates in Combined Immunotherapy of Patients with Glioblastoma. Sovrem. Tekhnologii Med. 2020, 12, 34–41. [Google Scholar] [CrossRef]
  79. Awada, G.; Ben Salama, L.; De Cremer, J. Axitinib plus avelumab in the treatment of recurrent glioblastoma: A stratified, open-label, single-center phase 2 clinical trial (GliAvAx). J. Immunother. Cancer 2020, 8, e001146. [Google Scholar] [CrossRef]
  80. Smith, C.; Lineburg, K.E.; Martins, J.P. Autologous CMV-specific T cells are a safe adjuvant immunotherapy for primary glioblastoma multiforme. J. Clin. Investig. 2020, 130, 6041–6053. [Google Scholar] [CrossRef]
  81. Reardon, D.A.; Brandes, A.A.; Omuro, A. Effect of Nivolumab vs Bevacizumab in Patients with Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef]
  82. Reardon, D.A.; Desjardins, A.; Vredenburgh, J.J. Rindopepimut with Bevacizumab for Patients with Relapsed EGFRvIII-Expressing Glioblastoma (ReACT): Results of a Double-Blind Randomized Phase II Trial. Clin. Cancer Res. 2020, 26, 1586–1594. [Google Scholar] [CrossRef] [PubMed]
  83. Mitsuya, K.; Akiyama, Y.; Iizuka, A. Alpha-type-1 Polarized Dendritic Cell-based Vaccination in Newly Diagnosed High-grade Glioma: A Phase II Clinical Trial. Anticancer Res. 2020, 40, 6473–6484. [Google Scholar] [CrossRef] [PubMed]
  84. Weathers, S.P.; Penas-Prado, M.; Pei, B.L. Glioblastoma-mediated Immune Dysfunction Limits CMV-specific T Cells and Therapeutic Responses: Results from a Phase I/II Trial. Clin. Cancer Res. 2020, 26, 3565–3577. [Google Scholar] [CrossRef]
  85. Yuce Sari, S.; Aktas, B.Y.; Kertmen, N. Does Combined Fractionated Stereotactic Radiotherapy and Immunotherapy Change the Outcome of Recurrent High-Grade Gliomas? Cureus 2021, 13, e15852. [Google Scholar] [CrossRef] [PubMed]
  86. Duerinck, J.; Schwarze, J.K.; Awada, G. Intracerebral administration of CTLA-4 and PD-1 immune checkpoint blocking monoclonal antibodies in patients with recurrent glioblastoma: A phase I clinical trial. J. Immunother. Cancer 2021, 9, e002296. [Google Scholar] [CrossRef] [PubMed]
  87. Lim, J.; Park, Y.; Ahn, J.W. Autologous adoptive immune-cell therapy elicited a durable response with enhanced immune reaction signatures in patients with recurrent glioblastoma: An open label, phase I/IIa trial. PLoS ONE 2021, 16, e0247293. [Google Scholar] [CrossRef] [PubMed]
  88. Jacques, F.H.; Nicholas, G.; Lorimer, I.A.J. Avelumab in newly diagnosed glioblastoma. Neurooncol Adv. 2021, 3, vdab118. [Google Scholar] [CrossRef]
  89. Jiang, H.; Yu, K.; Cui, Y. Combination of Immunotherapy and Radiotherapy for Recurrent Malignant Gliomas: Results from a Prospective Study. Front. Immunol. 2021, 12, 632547. [Google Scholar] [CrossRef]
  90. Werlenius, K.; Stragliotto, G.; Strandeus, M. A randomized phase II trial of efficacy and safety of the immunotherapy ALECSAT as an adjunct to radiotherapy and temozolomide for newly diagnosed glioblastoma. Neuro-Oncol. Adv. 2021, 3, vdab156. [Google Scholar] [CrossRef]
  91. Reardon, D.A.; Kim, T.M.; Frenel, J.S. Treatment with pembrolizumab in programmed death ligand 1-positive recurrent glioblastoma: Results from the multicohort phase 1 KEYNOTE-028 trial. Cancer 2021, 127, 1620–1629. [Google Scholar] [CrossRef]
  92. Nayak, L.; Molinaro, A.M.; Peters, K. Randomized Phase II and Biomarker Study of Pembrolizumab plus Bevacizumab versus Pembrolizumab Alone for Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2021, 27, 1048–1057. [Google Scholar] [CrossRef]
  93. Sahebjam, S.; Forsyth, P.A.; Tran, N.D. Hypofractionated stereotactic re-irradiation with pembrolizumab and bevacizumab in patients with recurrent high-grade gliomas: Results from a phase I study. Neuro-Oncology 2021, 23, 677–686. [Google Scholar] [CrossRef] [PubMed]
  94. Bota, D.A.; Taylor, T.H.; Lomeli, N. A Prospective, Cohort Study of SITOIGANAP to Treat Glioblastoma When Given in Combination with Granulocyte-Macrophage Colony-Stimulating Factor/Cyclophosphamide/Bevacizumab/Nivolumab or Granulocyte-Macrophage Colony-Stimulating Factor/Cyclophosphamide/Bevacizumab/Pembrolizumab in Patients Who Failed Prior Treatment with Surgical Resection, Radiation, and Temozolomide. Front. Oncol. 2022, 12, 934638. [Google Scholar] [CrossRef] [PubMed]
  95. Bota, D.A.; Taylor, T.H.; Piccioni, D.E. Phase 2 study of AV-GBM-1 (a tumor-initiating cell targeted dendritic cell vaccine) in newly diagnosed Glioblastoma patients: Safety and efficacy assessment. J. Exp. Clin. Cancer Res. 2022, 41, 344. [Google Scholar] [CrossRef] [PubMed]
  96. Hu, J.L.; Omofoye, O.A.; Rudnick, J.D. A Phase I Study of Autologous Dendritic Cell Vaccine Pulsed with Allogeneic Stem-like Cell Line Lysate in Patients with Newly Diagnosed or Recurrent Glioblastoma. Clin. Cancer Res. 2022, 28, 689–696. [Google Scholar] [CrossRef]
  97. Sampson, J.H.; Singh Achrol, A.; Aghi, M.K. Targeting the IL4 receptor with MDNA55 in patients with recurrent glioblastoma: Results of a phase IIb trial. Neuro Oncol. 2023, 25, 1085–1097. [Google Scholar] [CrossRef]
  98. Omuro, A.; Reardon, D.A.; Sampson, J.H. Nivolumab plus radiotherapy with or without temozolomide in newly diagnosed glioblastoma: Results from exploratory phase I cohorts of CheckMate 143. Neuro-Oncol. Adv. 2022, 4, vdac025. [Google Scholar] [CrossRef]
  99. Lim, M.; Weller, M.; Idbaih, A. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro-Oncology 2022, 24, 1935–1949. [Google Scholar] [CrossRef]
  100. Parney, I.F.; Anderson, S.K.; Gustafson, M.P. Phase I trial of adjuvant mature autologous dendritic cell/allogeneic tumor lysate vaccines in combination with temozolomide in newly diagnosed glioblastoma. Neuro-Oncol. Adv. 2022, 4, vdac089. [Google Scholar] [CrossRef]
  101. Chiocca, E.A.; Gelb, A.B.; Chen, C.C. Combined immunotherapy with controlled interleukin-12 gene therapy and immune checkpoint blockade in recurrent glioblastoma: An open-label, multi-institutional phase I trial. Neuro-Oncology 2022, 24, 951–963. [Google Scholar] [CrossRef]
  102. Ogino, H.; Taylor, J.W.; Nejo, T. Randomized trial of neoadjuvant vaccination with tumor-cell lysate induces T cell response in low-grade gliomas. J. Clin. Investig. 2022, 132, e151239. [Google Scholar] [CrossRef]
  103. Muragaki, Y.; Ishikawa, E.; Maruyama, T. A multicenter, randomized, placebo-controlled phase IIb trial of an autologous formalin-fixed tumor vaccine for newly diagnosed glioblastomas. J. Neurosurg. 2023, 139, 344–354. [Google Scholar] [CrossRef] [PubMed]
  104. Mahase, S.S.; Roytman, M.; Roth O’Brien, D. Concurrent immunotherapy and re-irradiation utilizing stereotactic body radiotherapy for recurrent high-grade gliomas. Cancer Rep. 2023, 6, e1788. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, Z.; Zhou, J.; Yang, X. Safety and antitumor activity of GD2-Specific 4SCAR-T cells in patients with glioblastoma. Mol. Cancer 2023, 22, 3. [Google Scholar] [CrossRef] [PubMed]
  106. Guo, C.; Yang, Q.; Xu, P. Adjuvant Temozolomide Chemotherapy with or without Interferon Alfa Among Patients with Newly Diagnosed High-grade Gliomas: A Randomized Clinical Trial. JAMA Netw. Open 2023, 6, e2253285. [Google Scholar] [CrossRef] [PubMed]
  107. Liau, L.M.; Ashkan, K.; Brem, S. 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, 9, 112–121. [Google Scholar] [CrossRef]
  108. Burger, M.C.; Forster, M.T.; Romanski, A. Intracranial injection of NK cells engineered with a HER2-targeted chimeric antigen receptor in patients with recurrent glioblastoma. Neuro-Oncology 2023, noad087. [Google Scholar] [CrossRef]
  109. Lepski, G.; Bergami-Santos, P.C.; Pinho, M.P. Adjuvant Vaccination with Allogenic Dendritic Cells Significantly Prolongs Overall Survival in High-Grade Gliomas: Results of a Phase II Trial. Cancers 2023, 15, 1239. [Google Scholar] [CrossRef]
  110. Jackson, C.M.; Choi, J.; Lim, M. Mechanisms of immunotherapy resistance: Lessons from glioblastoma. Nat. Immunol. 2019, 20, 1100–1109. [Google Scholar] [CrossRef]
  111. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef]
  112. Yeung, Y.T.; Bryce, N.S.; Adams, S. p38 MAPK inhibitors attenuate pro-inflammatory cytokine production and the invasiveness of human U251 glioblastoma cells. J. Neurooncol. 2012, 109, 35–44. [Google Scholar] [CrossRef] [PubMed]
  113. Rempel, S.A.; Dudas, S.; Ge, S.; Gutiérrez, J.A. Identification and localization of the cytokine SDF1 and its receptor, CXC chemokine receptor 4, to regions of necrosis and angiogenesis in human glioblastoma. Clin. Cancer Res. 2000, 6, 102–111. [Google Scholar]
  114. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef] [PubMed]
  115. Butowski, N.; Colman, H.; De Groot, J.F. Orally administered colony stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: An Ivy Foundation Early Phase Clinical Trials Consortium phase II study. Neuro-Oncology 2016, 18, 557–564. [Google Scholar] [CrossRef] [PubMed]
  116. Förnvik, K.; Maddahi, A.; Persson, O.; Osther, K.; Salford, L.G.; Nittby Redebrandt, H. C1-inactivator is upregulated in glioblastoma. PLoS ONE 2017, 12, e0183086. [Google Scholar] [CrossRef] [PubMed]
  117. Current State of Immunotherapy for Glioblastoma—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/29643471/ (accessed on 12 September 2023).
  118. Shen, S.; Chen, L.; Liu, J. Current state and future of co-inhibitory immune checkpoints for the treatment of glioblastoma. Cancer Biol. Med. 2020, 17, 555–568. [Google Scholar] [CrossRef]
  119. Zhang, N.; Wei, L.; Ye, M.; Kang, C.; You, H. Treatment Progress of Immune Checkpoint Blockade Therapy for Glioblastoma. Front. Immunol. 2020, 11, 592612. [Google Scholar] [CrossRef]
  120. Fife, B.T.; Bluestone, J.A. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol. Rev. 2008, 224, 166–182. [Google Scholar] [CrossRef]
  121. Kim, J.E.; Patel, M.A.; Mangraviti, A. Combination Therapy with Anti-PD-1, Anti-TIM-3, and Focal Radiation Results in Regression of Murine Gliomas. Clin. Cancer Res. 2017, 23, 124–136. [Google Scholar] [CrossRef]
  122. Harris-Bookman, S.; Mathios, D.; Martin, A.M. Expression of LAG-3 and efficacy of combination treatment with anti-LAG-3 and anti-PD-1 monoclonal antibodies in glioblastoma. Int. J. Cancer 2018, 143, 3201–3208. [Google Scholar] [CrossRef]
  123. Liau, L.M.; Ashkan, K.; Tran, D.D. 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, 142. [Google Scholar] [CrossRef]
  124. Narita, Y.; Arakawa, Y.; Yamasaki, F. A randomized, double-blind, phase III trial of personalized peptide vaccination for recurrent glioblastoma. Neuro-Oncology 2019, 21, 348–359. [Google Scholar] [CrossRef] [PubMed]
  125. Weller, M.; Kaulich, K.; Hentschel, B. Assessment and prognostic significance of the epidermal growth factor receptor vIII mutation in glioblastoma patients treated with concurrent and adjuvant temozolomide radiochemotherapy. Int. J. Cancer 2014, 134, 2437–2447. [Google Scholar] [CrossRef] [PubMed]
  126. Schuster, J.; Lai, R.K.; Recht, L.D. A phase II, multicenter trial of rindopepimut (CDX-110) in newly diagnosed glioblastoma: The ACT III study. Neuro-Oncology 2015, 17, 854–861. [Google Scholar] [CrossRef] [PubMed]
  127. Sampson, J.H.; Aldape, K.D.; Archer, G.E. Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro-Oncology 2011, 13, 324–333. [Google Scholar] [CrossRef]
  128. Schumacher, T.; Bunse, L.; Pusch, S. A vaccine targeting mutant IDH1 induces antitumour immunity. Nature 2014, 512, 324–327. [Google Scholar] [CrossRef]
  129. Andrews, D.W.; Judy, K.D.; Scott, C.B. Phase Ib Clinical Trial of IGV-001 for Patients with Newly Diagnosed Glioblastoma. Clin. Cancer Res. 2021, 27, 1912–1922. [Google Scholar] [CrossRef]
  130. Matsumoto, K.; Noguchi, M.; Satoh, T. A phase I study of personalized peptide vaccination for advanced urothelial carcinoma patients who failed treatment with methotrexate, vinblastine, adriamycin and cisplatin. BJU Int. 2011, 108, 831–838. [Google Scholar] [CrossRef]
  131. Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef]
  132. Patente, T.A.; Pinho, M.P.; Oliveira, A.A.; Evangelista, G.C.M.; Bergami-Santos, P.C.; Barbuto, J.A.M. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front. Immunol. 2018, 9, 3176. [Google Scholar] [CrossRef]
  133. Rutkowski, S.; De Vleeschouwer, S.; Kaempgen, E. Surgery and adjuvant dendritic cell-based tumour vaccination for patients with relapsed malignant glioma, a feasibility study. Br. J. Cancer. 2004, 91, 1656–1662. [Google Scholar] [CrossRef] [PubMed]
  134. Aiken, R.; Chen, C.; Cloughesy, T. Atim-33. Interim Results of a Phase II Multi-Center Study of Oncolytic Adenovirus Dnx-2401 with Pembrolizumab for Recurrent Glioblastoma; Captive Study (Keynote-192). Neuro-Oncology 2019, 21 (Suppl. S6), vi8–vi9. [Google Scholar] [CrossRef]
  135. Clinica Universidad de Navarra, Universidad de Navarra. Phase I Trial of DNX-2440 Oncolytic Adenovirus in Patients with Recurrent Glioblastoma. clinicaltrials.gov; 2023. Available online: https://clinicaltrials.gov/study/NCT03714334 (accessed on 15 August 2023).
  136. Benmebarek, M.R.; Karches, C.H.; Cadilha, B.L.; Lesch, S.; Endres, S.; Kobold, S. Killing Mechanisms of Chimeric Antigen Receptor (CAR) T Cells. Int. J. Mol. Sci. 2019, 20, 1283. [Google Scholar] [CrossRef] [PubMed]
  137. Larson, R.C.; Maus, M.V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 2021, 21, 145–161. [Google Scholar] [CrossRef]
  138. Gardner, R.A.; Finney, O.; Annesley, C. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 2017, 129, 3322–3331. [Google Scholar] [CrossRef]
  139. Jiang, X.; Lu, X.; Liu, R.; Zhang, F.; Zhao, H. HLA Tetramer Based Artificial Antigen-Presenting Cells Efficiently Stimulate CTLs Specific for Malignant Glioma. Clin. Cancer Res. 2007, 13, 7329–7334. [Google Scholar] [CrossRef]
  140. Thaci, B.; Brown, C.E.; Binello, E.; Werbaneth, K.; Sampath, P.; Sengupta, S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro-Oncology 2014, 16, 1304–1312. [Google Scholar] [CrossRef]
  141. Tu, M.; Wange, W.; Cai, L.; Zhu, P.; Gao, Z.; Zheng, W. IL-13 receptor α2 stimulates human glioma cell growth and metastasis through the Src/PI3K/Akt/mTOR signaling pathway. Tumour Biol. 2016, 37, 14701–14709. [Google Scholar] [CrossRef]
  142. Brown, C.E.; Warden, C.D.; Starr, R. Glioma IL13Rα2 is associated with mesenchymal signature gene expression and poor patient prognosis. PLoS ONE 2013, 8, e77769. [Google Scholar] [CrossRef]
  143. Kong, S.; Sengupta, S.; Tyler, B. Suppression of human glioma xenografts with second-generation IL13R-specific chimeric antigen receptor-modified T cells. Clin. Cancer Res. 2012, 18, 5949–5960. [Google Scholar] [CrossRef]
  144. Felsberg, J.; Hentschel, B.; Kaulich, K. Epidermal Growth Factor Receptor Variant III (EGFRvIII) Positivity in EGFR-Amplified Glioblastomas: Prognostic Role and Comparison between Primary and Recurrent Tumors. Clin. Cancer Res. 2017, 23, 6846–6855. [Google Scholar] [CrossRef] [PubMed]
  145. Yang, S.; Wei, W.; Zhao, Q. B7-H3, a checkpoint molecule, as a target for cancer immunotherapy. Int. J. Biol. Sci. 2020, 16, 1767–1773. [Google Scholar] [CrossRef] [PubMed]
  146. Majzner, R.G.; Theruvath, J.L.; Nellan, A. CAR T Cells Targeting B7-H3, a Pan-Cancer Antigen, Demonstrate Potent Preclinical Activity Against Pediatric Solid Tumors and Brain Tumors. Clin. Cancer Res. 2019, 25, 2560–2574. [Google Scholar] [CrossRef] [PubMed]
  147. Tang, X.; Wang, Y.; Huang, J. Administration of B7-H3 targeted chimeric antigen receptor-T cells induce regression of glioblastoma. Signal Transduct. Target. Ther. 2021, 6, 125. [Google Scholar] [CrossRef] [PubMed]
  148. Pieńkowski, T.; Zielinski, C.C. Trastuzumab treatment in patients with breast cancer and metastatic CNS disease. Ann. Oncol. 2010, 21, 917–924. [Google Scholar] [CrossRef]
  149. Bielamowicz, K.; Fousek, K.; Byrd, T.T. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro-Oncology 2018, 20, 506–518. [Google Scholar] [CrossRef]
Figure 1. Modified Newcastle–Ottawa scale.
Figure 1. Modified Newcastle–Ottawa scale.
Ijms 24 15037 g001
Figure 2. PRISMA flowchart.
Figure 2. PRISMA flowchart.
Ijms 24 15037 g002
Table 1. Summary of the studies included in the systematic literature review.
Table 1. Summary of the studies included in the systematic literature review.
AuthorYearTrial NamePhasePatients (N)Diagnosis (Target Glioma)Follow Up (Months, Median Value)TreatmentEndpoints
OSPFS
Riva et al. [14]1994N/AI24Recurrent HGGN/AAdjuvant RIT: murine monoclonal anti-Tenascin Ab (BC-2 and BC-4) labeled with 131I injected directly into the tumor through a cathetermOS: 16 moN/A
Riva et al. [15]1995N/AI50GBMN/AAdjuvant RIT: murine monoclonal anti-Tenascin Ab (BC-2 and BC-4) labeled with 131I locally infused in the site of neoplastic disease through a catheter + RT and CHTmOS: 20 moN/A
Pöpperl et al. [16]2002N/AI12 GBMN/AAdjuvant RIT: murine monoclonal anti-Tenascin Ab (BC-2 and BC-4) labeled with 131I locally infused in the surgical cavity (compared with an historical control group (n = 85) treated with standard therapy)mOS: 18.5, mOS in historical control group: 9.7 moN/A
Fukushima et al. [17]2003N/AI26GBMN/ART + MCNU (ranimustine) + TNF-SAM2 (recombinant human mutant TNF-α) (compared with an historical control group (n = 26) treated with standard therapy)mOS: 330 wkN/A
Yu et al. [18]2004N/AI14GBMN/AAutologous tumor lysate-pulsed DCVmOS: 133 wkN/A
Steiner et al. [19]2004N/AII10GBM49(1) ATV-NDV (NDV-modified autologous tumor vaccine) + SOC
(2) Standard therapy
(1) mOS: 100 wk, OS-1 y/2 y/3 y: 91%/39%/4%
(2) mOS: 49 wk, OS-1 y/2 y/3 y: 45%/11%/0%
(1) mPFS: 40 wk, PFS-1 y/2 y: 21%/4%
(2) mPFS: 26 wk, PFS-1 y/2 y: 8%/1%
Yamanaka et al. [20]2005N/AI/II35Recurrent GBM24(1) Autologous tumor lysate-pulsed DCV + KLH or KLH/OK-432
(2) EBRT + nitrosourea-based CHT
(1) OS-2 y: 23.5%
(2) OS-2 y: 3.7%
N/A
Vleeschouwer et al. [21]2008N/AI56Recurrent GBM16Adjuvant autologous resected GBM lysate-pulsed (mature) DCVmOS: 9.6 mo OS-12 mo/24 mo/36 mo: 37.4%/14.8%/11.1%mPFS: 3 mo, PFS-12 mo: 10.7%
Izumoto et al. [22]2008N/AII21Recurrent GBMN/AWT1-235 peptide vaccinationN/AmPFS: 20 wk, PFS-6 mo: 33.3%
Ardon et al. [23]2010N/AN/A8GBMN/ASOC + autologous GBM lysate-loaded DCVN/APFS-6 mo: 75%, mOS: 24 mo
Sampson et al. [24]2010NCT00643097II35GBMN/A(1) Newly diagnosed GBM EGFRvIII + PEPvIII-KLH
(2) TMZ
N/A(1) mPFS: 14.2 mo, mOS: 26 mo
(2) mPFS: 6.3 mo, mOS: 15 mo
Okada et al. [25]2011N/AI/II22Recurrent GBMN/AVaccination with α-type 1 polarized DCs (αDC1) loaded with GAAs + immunoadjuvant poly-ICLCN/APFS-12 mo: 41%
Prins et al. [26]2011NCT00068510I23GBMN/AAutologous tumor lysate-pulsed DCV + Imiquimod or Poly-ICLC adjuvantmOS: 31.4 mo, OS-1 y/2 y/3 y: 91%/55%/47%N/A
Akiyama et al. [27]2012UMIN000000914I9GBMN/A5 synthetic peptides-pulsed DCV + KLH (compared with an historical control group treated with standard therapy)mOS 19 mo, mOS 16 mo of the historical control groupN/A
Valle et al. [28]2012N/AI5GBMN/AAutologous tumor lysate-pulsed DCVmOS: 27 mo, 2 y-OS: 80%mPFS: 16.1 mo
Cho et al. [29]2012N/AII34Newly diagnosed GBM33(1) SOC + adjuvant autologous tumor lysate-pulsed DCV
(2) SOC (surgery + RT + CHT)
(1) mOS: 31.9 mo OS-1 y/2 y/3 y: 88.8%/44.4%/16.7%
(2) mOS: 15 mo, OS-1 y/2 y/3 y: 75%/18.8%/0%
(1) mPFS: 8.5 mo
(2) mPFS: 8 mo
Crane et al. [30]2013NCT00293423I12Recurrent GBMN/AAdjuvant autologous HSPPC-96 vaccinemOS in responder: 47 wk, mOS in non-responder: 16 wkN/A
Phuphanich et al. [31]2013N/AI21GBM40Autologous tumor lysate-pulsed DCVmOS: 38.4 moOS-6 mo/12 mo/24 mo/36 mo: 100%/100%/93.7%/55.6%/38.4%PFS-6 mo/12 mo/18 mo/24 mo: 100%/62.5%/43.8%/43.8%
Tanaka et al. [32]2013N/AI17Recurrent GBMN/AGlutaraldehyde-fixed HUVEC vaccine (human umbilical vein endothelial cell)OS-6 mo/1 y/5 y: 88.2%/47.1%/17.6%mPFS: 5.5 mo
Pellegatta et al. [33]2013N/AN/A15Recurrent GBMN/AAutologous tumor lysate-pulsed DCVmOS: 8 momPFS: 4.4 mo
Vik-Mo et al. [2]2013NCT00846456I/II17GBMN/A(1) DCV with mRNA from GSC
(2) without DCV
N/A(1) mPFS: 694 d, mOS: 759 d
(2) mPFS: 236 d, mOS: 585 d
Pollack et al. [34]2014NCT01130077I26Pediatric BSG N/AAutologous tumor lysate-pulsed DCVmOS in BSG patients: 12.7 mo; median OS in HGG patients: 25.1 moN/A
Schuessler et al. [35]2014ACTRN12609000338268I11Recurrent GBMN/AACT with CMV-specific autologous cytotoxic T cellsmOS: 403 dmPFS: 246 d
Bloch et al. [36]2014NCT00293423II41Recurrent GBMN/AAdjuvant autologous HSPPC-96 vaccine (peptide complexes bound to chaperon HSP-96, overexpressed in HGG)OS-6 mo/12 mo: 90.2%/29.3%, mOS: 42.6 wkN/A
Ishikawa et al. [37]2014UMIN000001426I/II24Newly diagnosed GBM30Autologous formalin-fixed GBM tumor vaccine (AFTV) + FRT and TMZOS-2 y/3 y: 47%/38%, mOS: 22.2 moPFS-2 y: 33%, mPFS: 8.2 mo
Hashimoto et al. [38]2015N/AI7GBMN/AWT1-peptide vaccination + TMZN/APFS: range 5.2–49-1 mo
Schijns et al. [39]2015N/AI48Recurrent GBM10(1) Gliovac (or ERC 1671) vaccine
(2) SOC
(1) OS-6 mo: 100%, OS-40 wk: 77%
(2) OS-6 mo: 33%, OS-40 wk: 10%
N/A
Sakai et al. [40]2015N/AI10Recurrent GBM21Autologous tumor lysate-pulsed DCVmOS: 26 mo, OS-21 mo: 50%N/A
Kalkanis et al. [41]2015NCT01156584I54Recurrent HGGN/A(1) Toca 511 + Toca FC
(2) External control
(1) mOS: 13.6 mo
(2) mOS: 7.1 mo
N/A
Westphal et al. [42]2015N/AIII142Newly diagnosed GBMN/A(1) Nimotuzumab + SOC
(2) SOC
(1) mOS: 19.5 mo (residual tumor) and 23.3 mo (no residual tumor)
(2) mOS: 16,7 mo (residual tumor) and 21 mo (no residual tumor)
(1) PFS-12 mo: 25.6%, mPFS: 5.6 mo (residual tumor) and 10.6 mo (no residual tumor)
(2) PFS-12 mo: 20.3% mPFS: 4 mo (residual tumor) and 9.9 mo (no residual tumor)
Akasaki et al. [43]2016N/AI/II32Recurrent and newly diagnosed GBMN/ATMZ + immunotherapy with fusion cell (FC): autologous cultured GBM cell were fused with autologous DC using polyethylene glycolRecurrent GBM: mOS: 18 mo; newly diagnosed GBM: mOS: 30.5 moRecurrent GBM: PFS: 10.3 mo; newly diagnosed GBM: mPFS: 18.3 mo
Brown et al. [44]2016NCT02208362I92Recurrent GBMN/AIL13 Rα2-specific CAR T cellsN/APFS: 7.5 mo
Cloughesy et al. [45]2016NCT01156584I54Recurrent GBMN/A(1) Toca 511 + Toca FC
(2) External control
(1) mOS: 13.6 mo
(2) mOS: 7.1 mo
N/A
Fenstermaker et al. [46]2016NCT01250470I9Recurrent GBM SURVIVIN+N/ASVN53-67/M57-KLH (SurVaxM): conjugated survivin peptide mimic vaccine with KLHmOS: 86.6 wk, OS-1 y: 77.8%mPFS: 17.6 wk
Oji et al. [47]2016UMIN000002001II50GBMN/AWT1-235 peptide vaccinationOS significantly prolonged (p = 0.001)PFS significantly prolonged (p = 0.028)
Wheeler et al. [48]2016NCT00589875II182Newly diagnosed GBM36(1) GMCI + SOC
(2) SOC
(1) OS-1 y/2 y/3 y: 67%/35%/19%
(2) OS-1 y/2 y/3 y: 57%/22%/8%
N/A
Alonso et al. [49]2017NCT01956734I61Recurrent GBMN/ADNX-2401 + TMZOS-9 mo: 100%N/A
O’Rourke et al. [50]2017NCT02209376I10Recurrent GBM EGFRvIII+N/ACAR T-EGFRvIII+mOS: 251 dN/A
Kong et al. [51]2017NCT00807027III180Newly diagnosed GBMN/A(1) ACT w/expansion of autologous CIK (cytokine-induced killer cells) + SOC
(2) SOC
(1) mOS: 22.5 mo, OS-12 mo/18 mo/24 mo: 78.2%/57.2%/38.2%
(2) mOS: 16.9 mo, OS-12 mo/18 mo/24 mo: 75.2%/45.1%/38.5%
(1) mPFS: 8.1 mo, PFS-12 mo/18 mo/24 mo: 28.3%/25.6%/18.4%
(2) mPFS: 5.4 mo, PFS-12 mo/18 mo/24 mo: 22.6%/21.2%/13.4%
Ursu et al. [52]2017N/AII81GBMN/A(1) CpG-ODN (administrated locally around the surgical cavity) + SOC
(2) SOC
(1) OS-2 y: 31%
(2) OS-2 y: 26%
(1) mPFS: 9 mo
(2) mPFS: 8.5 mo
Inogés et al. [53]2017NCT01006044II31Newly diagnosed GBMN/AAutologous whole tumor lysate-pulsed DCV + RT and CHTmOS: 23.4 momPFS: 12.7 mo
Geletneky et al. [54]2017NCT01301430I/II18Progressive primary or recurrent GBM6H-1 PV (H-1 parvovirus)mOS: 15.5 moPFS: 4 mo
Weller et al. [55] 2017NCT01480479III745Newly diagnosed GBM EGFRvIII+12(1) Rindopepimut (with KLH) + GM-CSF and TMZ
(2) KLH and TMZ
(1) mOS: 20.1 mo
(2) mOS: 20 mo
N/A
Zadeh et al. [56]2018NCT02798406II49Recurrent GBMN/ADNX-2401, pembrolizumabOS-9 mo: 100%N/A
Peereboom et al. [57]2018NCT02078648I/II74Recurrent GBM HLA-A2+N/ASL701/GM-CSF + poly-ICLC and bevacizumabOS-12: 37%
Cloughesy et al. [58]2018NCT01470794I56Recurrent GBM36Toca 511 + Toca FCmOS: 11.9 mo (95% CI, 10.7 mo to 15.1 mo)N/A
Fried et al. [59]2018N/AI9Pediatric DIPGN/APidilizumab (MDV9300) + RTmOS: 15.6 momPFS: 9.3 mo
Pellegatta et al. [60]2018N/AII24GBM17Autologous tumor lysate-pulsed DCV + TMZmOS: 20.1, OS-1 y/2 y: 75%/37%mPFS: 10.5 mo, PFS-6 mo: 79%, PFS-12 mo: 37.5%
Yao et al. [61]2018N/AII47GBMN/AAutologous tumor lysate-pulsed DCV vs. placeboOS significantly prolonged (p < 0.01)PFS significantly prolonged (p = 0.03)
Wick et al. [62]2018NCT02149225I16Newly diagnosed GBM HLA-A*02:01 or HLA-A*24:02+N/AAPVAC1 or APVAC2 (multi-peptide vaccines)/GM-CSF + poly-ICLC + TMZmOS: 29 momPFS: 14.2 mo
Desjardins et al. [63]2018NCT01491893I61Recurrent supratentorial GBMN/APVSRIPO (compared with an historical control group (n = 104) treated with standard therapy)OS-6 mo/12 mo/24 mo/36 mo/48 mo/60 mo: 90%/54%/21%/21%/21%/21%N/A
Buchroithner et al. [64]2018N/AII76Newly diagnosed GBMN/A(1) Tumor lysate-charged autologous DCV (Audencel) + SOC
(2) SOC
(1) mOS: 564 d
(2) mOS: 568 d
(1) PFS-12 mo: 28.4%
(2) PFS-12 mo: 24.5%
Bota et al. [65]2018N/AII9Recurrent GBMN/A(1) Gliovac (ERC 1671) vaccine + bevacizumab
(2) Placebo + bevacizumab
(1) mOS: 12.1 mo, OS-12 mo: 50%
(2) mOS: 7.6 mo, OS-12 mo: 26%
(1) mPFS: 7.3 mo
(2) mPFS: 5.4 mo
Lang et al. [66]2018NCT00805376I37Recurrent HGGN/A(1) DNX-2401 intratecal injection
(2) DNX-2401 intrathecal infusion + resection
(1) mOS: 9.5 mo
(2) mOS: 13.0 mo
N/A
Kieran et al. [67]2019NCT00634231I12GBMN/AAdV-tk plus valacyclovir + RTOS: 24 moPFS: 37.3 and 47.7 mo
Todo et al. [68]2019UMIN000015995II30Recurrent GBMN/AG47 delta (compared with an historical control group treated with standard therapy)OS-12: 84.4%N/A
Wen et al. [69]2019NCT01280552II124Newly diagnosed GBM40(1) ICT-107 (peptide-pulsed DC vaccine)
(2) Un-pulsed DCs
(1) mOS: 17.0 mo
(2) mOS: 15.0 mo (HR = 0.87, p = 0.58)
N/A
Chiocca et al. [70]2019NCT02026271I31Recurrent HGG13hIL-12 vector (Ad–RTS–hIL–12) injected in resection cavity + Veledimex (VDX, oral activator for hIL-12)mOS in VDX 20 mg cohort: 12.7 mo, OS-12 mo/18 mo/24 mo in VDX 20 mg cohort: 60%/26.7%/13.3%, OS-12 mo in 10 mg/ 20 mg/ 30 mg/ 40 mg cohorts: 0%/60%/0%/30%N/A
Cloughesy et al. [71]2019NCT02414165II/III403Recurrent GBM23Toca 511 + Toca FCmOS: 11.1 moN/A
Migliorini et al. [72]2019NCT01920191I/II19Newly diagnosed GBMN/ARCHT + IMA950 multipeptide vaccine (w/ adjuvant poly-ICLC)mOS: 10 mo (all patients) and 9.5 mo (GBM patients)mPFS: 10 mo (all pt.) and 9.5 (GBM patients), PFS-6 mo/9 mo: 84% (all patients) and 63% (GBM patients)
Eoli et al. [73]2019NCT04002804I/II20Recurrent GBM9(1) Autologous tumor lysate-pulsed DCV + TMZ
(2) Autologous tumor lysate-pulsed DCV + TT preconditioning of the vaccine site
(1) OS-9: 33%
(2) OS-9: 62.5%
N/A
Goff et al. [74]2019NCT01454596I18Recurrent GBM EGFRvIII+N/AAutologous EGFRvIII-specific CAR T cellsmOS: 6.9 momPFS: 1.3 mo
Cloughesy et al. [75]2019N/AI32Recurrent GBM16(1) Neoadjuvant pembrolizumab + adjuvant pembrolizumab
(2) Adjuvant pembrolizumab
N/A(1) mPFS: 99.5 d
(2) mPFS: 72.5 d
Cloughesy et al. [76]2020NCT02511405III256Recurrent GBMN/A(1) VB-111, bevacizumab
(2) Bevacizumab
(1) mOS: 6.8 mo
(2) mOS: 7.9 mo
N/A
Mueller et al. [77]2020NCT02960230I29New GBM with H3.3 K27M mutation18(1) H3.3 K27M peptide vaccine
(2) H3.3 K27M peptide vaccine + Nivolumab
(1) mOS: 16.1 mo
(2) mOS: 9.8 mo
N/A
Mishinov et al. [78]2020N/AI58GBMN/A(1) Autologous tumor lysate-pulsed DCV + standard treatment
(2) Allogeneic pooled lysates from more tumors—pulsed DCV + standard treatment
(3) Maximum safe tumor resection + RT + CHT
(1) mOS: 16 mo
(2) mOS: 15 mo
(3) mOS: 14.5 mo
N/A
Awada et al. [79]2020NCT03291314II54Recurrent GBM25(1) Axitinib + avelumab
(2) Axitinib
(1) mOS: 26.6 wk
(2) mOS: 18 wk
(1) PFS-6 mo: 22.2%
(2) PFS-6 mo: 18.5%
Smith et al. [80]2020ACTRN12615000656538I25Newly diagnosed GBM12ACT with CMV-specific autologous cytotoxic T cellsmOS: 21 mo, OS-6 mo/1 y/2 y: 92%/84%/36%mPFS: 10 mo, PFS-6 mo/1 y/2 y: 72%/28%/16%
Reardon et al. [81]2020NCT02017717III369Recurrent GBM10(1) Nivolumab
(2) Bevacizumab
(1) mOS: 9.8 mo OS-6 mo/12 mo/18 mo:72.3%/42%/21%
(2) mOS: 10 mo, OS-6 mo/12 mo/18 mo:78.2%/42%/21%
(1) mPFS: 1.5 mo, PFS-6 mo/12 mo/18 mo:15.7%/10.5%/5.8%
(2) PFS-6 mo/12 mo/18 mo:29.6%/17.4%/8.9%
Reardon et al. [82]2020NCT01498328II73Recurrent GBM EGFRvIII+N/A(1) Rindopepimut (with KLH) + GM-CSF and bevacizumab
(2) KLH and bevacizumab
(1) PFS-6 mo: 28%, OS-24 mo: 20%
(2) PFS-6 mo: 16%, OS-24 mo: 3%
(1) OS-24 mo: 20%
(2) OS-24 mo: 3%
Mitsuya et al. [83]2020NTC0190103II16Newly diagnosed GBM725 synthetic peptides-pulsed DCVmOS: 19 moN/A
Weathers et al. [84]2020NCT02661282I/II20GBM12ACT with CMV-specific autologous cytotoxic T cellsOS-1 y: 50%, mOS: 12 moPFS-6 mo: 19%, mPFS: 1.3 mo
Yuce Sari et al. [85]2021N/AI8Recurrent GBM21FSRT + neoadjuvant, concomitant, adjuvant nivolumabmOS: 21.3 mo (from diagnosis) and 12.6 mo (from progression), OS-1 y/2 y: 88%/33% (from diagnosis) and 73%/0% (from progression)mPFS: 2.3 mo
Duerinck et al. [86]2021NCT03233152I27Recurrent GBM22IC (in the brain tissue lining the resection cavity) nivolumab and ipilimumab + intravenous nivolumab (compared with an historical control group (n = 469) treated with standard therapy)mOS: 38 wk, OS-6 mo/1 y/2 y: 74.1%/40.7%/27%N/A
Lim et al. [87]2021KCT0003815 I/II14Recurrent GBM24ACT (adoptive immune cell therapy) with activated NK cells and T lymphocytes from PBMCmOS: 22. mo, OS-2 y: 35.7%mPF6: 10 mo
Jacques et al. [88]2021NCT03047473II30Newly diagnosed GBM42Avelumab (ICI) + SOCmOS: 15.3 momPFS: 9.7 mo
Jiang et al. [89]2021NCT03392545I30Recurrent GBM and DMG (H3K27M-mutant)23Cyclophosphamide (Treg depletion) IC, immunoadjuvant Poly I:C + systemic immunoadjuvant Poly I:C and GM-CSF, low-dose re-irradiationmOS: 362 dmPFS: 88 d
Werlenius et al. [90]2021NCT02799238II62Newly diagnosed GBMN/A(1) ALECSAT + SOC
(2) SOC
(1) mOS: 19.2 mo
(2) mOS: 18.3 mo
(1) mPFS: 7.8 mo
(2) mPFS: 7.9 mo
Reardon et al. [91]2021NCT02054806I26Recurrent GBM PD-L1+14PembrolizumabmOS: 13.1 mo, OS-6 mo/1 y/2 y: 75.8%/58%/31%mPFS: 2.8 mo, PFS-6 mo/1 y/2 y: 37.7%/16.8%/8.4%
Nayak et al. [92]2021NCT02337491II80Recurrent GBM49(1) Pembrolizumab + bevacizumab
(2) Pembrolizumab
(1) mOS: 8.8 mo, OS-6 mo/12 mo/18 mo: 79.7%/44.3%/16.9%
(2) mOS: 10.3 mo, OS-6 mo/12 mo/18 mo: 70%/30%/23.3%
(1) mPFS: 4.1 mo, PFS-6 mo/12 mo/18 mo: 26%/14%/10%
(2) mPFS: 1.4 mo, PFS-6 mo/12 mo/18 mo: 6.7%/6.7%/3.3%
Sahebjam et al. [93]2021NCT02313272I32Recurrent GBM3Pembrolizumab + RT + bevacizumab(1) Bevacizumab-naïve: mOS: 13.4 mo, OS-6 mo/1 y/2 y: 91.7%/58.3%/16.7%
(2) Bevacizumab-resistant: mOS: 9.3 mo, OS-6 mo/1 y: 87.5%/25%
(1) Bevacizumab-naïve: mPFS: 7.9 mo, PFS-6 mo/1 y: 66.7%/29.2%
(2) Bevacizumab-resistant: mPFS: 6,5 mo, mOS: 9.3 mo, PFS-6 mo/1 y: 87.5%/25%
Bota et al. [94]2022N/AN/A21Recurrent GBMN/ABevacizumab + pembrolizumab or nivolumab + ERC 1671 vaccinemOS: 19.63 mo, mOS-6 mo/1 y/2 y: 90.5%/61.1%/45.3%mPFS: 9.14 mo, PFS-6 mo/1 y/2 y: 76.2%/47.62%/21.4%
Bota et al. [95]2022NCT03400917II57GBMN/AAutologous tumor lysate-pulsed DCV + GM-CSFmOS: 14 mo, OS-6 mo/12 mo/18 mo/24 mo: 87.5%/55.4%/38.5%/25.2%mPFS: 8.5 mo, PFS-6 mo/12 mo/18 mo/24 mo: 69.7%/26.8%/16.1%/10%
Hu et al. [96]2022NCT02010606I36GBMN/AAutologous tumor lysate-pulsed DCV + SOCmOS: 20.36 momPFS: 8.75 mo
Sampson et al. [97]2022NCT02858895II44Recurrent GBMN/AMDNA55 (IL4 R targeting toxin) administered intratumorally using convection-enhanced delivery(1) mOS: 11.6 mo, OS-1 y: 46%
(2) Subgroup (n = 32) of IL4 R high and low patients treated with high-dose MDNA55]: mOS: 15 mo, OS-1 y: 55%
N/A
Omuro et al. [98]2022NCT02017717I117Newly diagnosed GBMN/A(1) PART A: COHORT 1 c (n = 31) nivolumab + RT + TMZ; COHORT 1 d (n = 30) nivolumab + RT
(2) PART B: COHORT 1 c (n = 28) nivolumab + RT + TMZ; COHORT 1 d (n = 28) nivolumab + RT
(1) PART A: COHORT 1 c mOS: 22 mo, COHORT 1 d mOS: 14.4 mo
(2) PART B: COHORT 1 c mOS: 15 mo, COHORT 1 d mOS: 14 mo
(1) PART A: COHORT 1 c mPFS: 10 mo, COHORT 1 d mPFS: 5.6 mo
(2) PART B: COHORT 1 c mPFS: 6.4 mo, COHORT 1 d mPFS: 6 mo
Lim et al. [99]2022NCT02667587III716Newly diagnosed GBM with UNmethylated MGMT promoterN/A(1) Nivolumab + SOC
(2) Placebo + SOC
(1) mOS: 28.9 mo
(2) mOS: 32.1 mo
(1) mPFS: 10.6 mo
(2) mPFS: 10.3 mo
Parney et al. [100]2022NCT01957956I20Newly diagnosed GBM35Allogenetic GBM lysate-pulsed (mature) DCV + TMZmOS: 19 mo, OS-2 y/4 y: 25%/10%mPFS: 9.7 mo
Chiocca et al. [101]2022NCT03636477I21Recurrent GBMN/ANivolumab + peritumoral injection of hIL-12 vector (Ad–RTS–hIL–12) + VDXmOS all patients: 9.8 mo, mOS in VDX 10 mg: 16.9 mo, mOS in VDX 20 mg: 8.5 moN/A
Ogino et al. [102]2022NCT02549833I17GBM21GBM6-AD + Poly-ICLC adjuvant:
(1) neoadjuvant vaccination + surgery + adjuvant vaccination
(2) surgery + adjuvant vaccination
N/ANo significant differences between PFS between two arms
Muragaki et al. [103]2023UMIN000010602II57Newly diagnosed supratentorial GBMN/A(1) Autologous formalin-fixed GBM tumor vaccine (AFTV) + immune adjuvants
(2) Identical placebo without fixed tumor tissue
(1) mOS: 25.6 mo, OS-3 y: 38%, PFS-3 y: 81%, OS-3 y: 80%
(2) mOS: 31 mo, OS-3 y: 41%, PFS-3 y: 46%, OS-3 y: 54%
N/A
Mahase et al. [104]2023N/AI21Recurrent GBMN/A(1) ICI + SBRT (fractionated stereotactic radiosurgery)
(2) ICI (Pembrolizumab and Nivolumab)
(1) mOS: 7 mo
(2) mOS: 6 mo
(1) mPFS: 2.8 mo
(2) mPFS: 1 mo
Liu et al. [105]2023NCT03170141I8GBM GD2+24Autologous GD2-specific 4SCAR-T cellsmOS: 10 moN/A
Guo et al. [106]2023NCT01765088III199Newly diagnosed HGG66(1) IFN-α + TMZ
(2) TMZ
(1) mOS: 26.7 mo and 24.7 mo (subgroup with UNmet MGMT prom), OS-2 y/5 y: 57.4%/18.1%
(2) mOS: 18.8 mo and 17.4 mo (subgroup with UNmet MGMT prom), OS-2 y/5 y: 37.3%/9.1%
(1) mPFS: 14.8 mo, PFS-2 y/5 y: 27.9%/9.6%
(2) mPFS: 12.9 mo, PFS-2 y/5 y: 18.5%/4.8%
Liau et al. [107]2023NCT00045968III331GBMN/AAutologous tumor lysate-pulsed DCV + SOCmOS: 19.3 mo (nGBM) and 13.2 mo (rGBM), OS-48 mo/60 mo: 15.7%/13% (nGBM), OS-24 mo/30 mo: 20.7%/9.7% (rGBM)N/A
Burge et al. [108]2023NCT03383978I9Recurrent GBM IDH wild type HER2 +N/AIC injection (into the margin of surgical cavity) of HER2-targeted CAR-NK cells NK-92/5.28 z (ACT, adoptive cell therapy)mOS: 31 wkmPFS: 7 wk
Lepski et al. [109]2023N/AI/II37Recurrent GMBN/AAllogenic DC vaccination for GBMOS: 26.9 mo, OS-6 mo/12 mo/18 mo/24 mo: 61.3%/46.6%/34.9%/26%N/A
Abbreviations: Ab = antibody, ACT = adoptive cellular therapy, AFTV = autologous formalin-fixed tumor vaccine, ATV = autologous tumor vaccine, ALECSAT = autologous lymphoid effector cells specific against tumor, CAR T = chimeric antigen receptor T cell, CHT = chemotherapy, CpG-ODN = oligodeoxynucleotides containing unmethylated cytosine–guanosine motif, d = day, DC = dendritic cell, DIPG = diffuse intrinsic pontine glioma, DMG = diffuse midline glioma, DCV = dendritic cell vaccine, EBRT = external beam radiotherapy, FC = fusion cell, FRT = fractionated radiotherapy, FSRT = fractionated stereotactic radiotherapy, GAAs = glioma-associated antigens, GBM = glioblastoma multiforme, GMCI = gene-mediated cytotoxic immunotherapy, HGG = high-grade glioma, HSPPC-96 = heat-shock protein 96—peptide complex, HUVEC = human umbilical vein endothelial cell, IC = intracranial, ICIs = immune checkpoint inhibitors, mo = month, nGBM = newly diagnosed glioblastoma multiforme, N/A = not applicable, NDV = Newcastle disease virus, OS = overall survival, PBMC = peripheral blood mononuclear cell, PD = progressive disease, PFS = progression free survival, rGBM = recurrent glioblastoma multiforme, RCHT = radiochemotherapy, RIT = radioimmunotherapy, RT = radiotherapy, SOC = standard of care treatment (RT + TMZ), TMZ = temozolomide, TT = tetanus toxoid, VDX = veledimex, wk = week, y = year.
Table 2. Summary of the ongoing studies on GBM immunotherapies testing ICIs.
Table 2. Summary of the ongoing studies on GBM immunotherapies testing ICIs.
Trial NamePhasePatients (N)TreatmentOutcomes
NCT02311920I32Ipilimumab and/or nivolumab in combination with temozolomidePFS and OS
NCT02336165II159Durvalumab monotherapy, with bevacizumab or with radiotherapyOS and PFS
NCT02658981I63Anti-LAG3 or urelumab alone in combination with nivolumabMTD
NCT03673787I/II87 Atezolizumab in combination with ipatasertibDLT
NCT03743662II94Nivolumab with radiation therapy and bevacizumabT-lymphocyte density and safety
NCT03961971I15Anti-Tim-3 in combination with anti-PD-1 and stereotactic radiosurgerySerious adverse events
NCT04145115II37Ipilimumab and nivolumabDLT
NCT04396860II/III485Ipilimumab and nivolumab plus radiation therapyEfficacy and safety
NCT04606316I60Nivolumab in combination with ipilimumab and surgeryTumor infiltrating
Abbreviations: DLT = dose-limiting toxicity, MTD = maximum tolerated dose, OS = overall survival, PFS = progression free survival.
Table 3. Summary of the ongoing studies on GBM immunotherapies testing CVs.
Table 3. Summary of the ongoing studies on GBM immunotherapies testing CVs.
Trial NamePhasePatients (N)TreatmentOutcomes
NCT02366728II100CMV pp65 DC vaccine +111In-labeled DC vaccine + Td Toxoid + basiliximabOS
NCT02465268II175pp65-shLAMP DC dendritic cell vaccine with GM-CSFOS
NCT02924038I30IMA-950 (peptide vaccine comprising multiple GAAs) and poly-ICLC ± varlilumab (immunostimulatory antiCD27 antibody)Safety and T-cell responses
NCT02960230I29H3.3K27 M peptide vaccine plus Td and poly-ICLCSafety and OS
NCT03018288II108pembrolizumab ± HSPPC-96 vaccine1-year OS
NCT03400917II55AV-GBM-1 (autologous dendritic cells loaded with tumor associated antigens from a short-term cell culture of autologous tumor cells)OS
NCT04116658II52EO2401 peptide vaccineSafety and tolerability
Abbreviations: GBM = glioblastoma multiforme, OS = overall survival.
Table 4. Summary of the ongoing studies on GBM immunotherapies testing OVs.
Table 4. Summary of the ongoing studies on GBM immunotherapies testing OVs.
Trial NamePhasePatients (N)TreatmentOutcomes
NCT01301430II18Human mesenchymal stem cells TG6002 (modified vaccinia virus) and 5-FCSafety and DLT
NCT01470794I58DNX-2440 conditionally replication-competent adenovirus with O × 40 ligand (T-cell stimulator)DLT
NCT02062827I36DNX-2401 (Delta-24-RGD adenovirus) ± surgeryMTD
NCT02798406II48DNX-2401 (Delta-24-RGD adenovirus) and i.v. pembrolizumab (anti-PD-1 antibody)ORR by interval tumor size change
NCT02986178II62G207 (modified oncolytic strain of HSV-1) single-dose inoculationOS at 24 months
NCT03152318I108PVSRIPO (genetically recombinant nonpathogenic poliovirus:rhinovirus chimera) ± lomustineMTD
NCT03294486II78Ad5-DNX-2401 (oncolytic adenovirus) in bone marrowProgression at 6 months
NCT03714334I24rQNestin 34.5 v.2 (oncolytic HSV-1) + cyclophosphamideTreatment-related adverse events
NCT03896568I36Ad-RTS-hIL-12 + veledimexSafety, DLT, and rate of tumor
NCT00390299I40M032 (modified strain of HSV-1) by intratumoral infusionToxicity and MTD
Abbreviations: DLT = dose-limiting toxicity, MTD = maximum tolerated dose, ORR = overall response rate.
Table 5. Summary of the ongoing studies on GBM immunotherapies testing CAR T cells.
Table 5. Summary of the ongoing studies on GBM immunotherapies testing CAR T cells.
Trial NamePhasePatients (N)TreatmentOutcomes
NCT01109095I16HER2 CMV-specific CAR T cellsDLT
NCT01454596I/II18EGFRvIII-directed CAR T cells with cyclophosphamide, fludarabine and aldesleukinAEs and PFS
NCT02208362I92IL13 Rα2-targeted CAR T cellsAEs and DLT
NCT02209376I11EGFRvIII-directed CAR T cellsAEs
NCT04003649I60IL13 Rα2-targeted CAR T cells with or without nivolumab and ipilimumabAEs, DLT, feasibility, and OS
NCT04077866I/II40B7-H3-targeted CAR T cells with or without temozolomideOS and PFS
NCT04385173I12B7-H3-targeted CAR T cells with temozolomideAEs, MTD, OS, and PFS
NCT04661384I30IL13 Rα2-targeted CAR T cellsAEs and OS
Abbreviations: AEs = adverse events, CAR T = chimeric antigen receptor T cell, DLT = dose-limiting toxicity, MTD = maximum tolerated dose, OS = overall survival, PFS = progression free survival.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Agosti, E.; Zeppieri, M.; De Maria, L.; Tedeschi, C.; Fontanella, M.M.; Panciani, P.P.; Ius, T. Glioblastoma Immunotherapy: A Systematic Review of the Present Strategies and Prospects for Advancements. Int. J. Mol. Sci. 2023, 24, 15037. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242015037

AMA Style

Agosti E, Zeppieri M, De Maria L, Tedeschi C, Fontanella MM, Panciani PP, Ius T. Glioblastoma Immunotherapy: A Systematic Review of the Present Strategies and Prospects for Advancements. International Journal of Molecular Sciences. 2023; 24(20):15037. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242015037

Chicago/Turabian Style

Agosti, Edoardo, Marco Zeppieri, Lucio De Maria, Camilla Tedeschi, Marco Maria Fontanella, Pier Paolo Panciani, and Tamara Ius. 2023. "Glioblastoma Immunotherapy: A Systematic Review of the Present Strategies and Prospects for Advancements" International Journal of Molecular Sciences 24, no. 20: 15037. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms242015037

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop