Skip to main content
Neuro-Oncology Advances logoLink to Neuro-Oncology Advances
. 2025 Sep 9;7(Suppl 4):iv72–iv83. doi: 10.1093/noajnl/vdaf051

Translational advancements in tumor vaccine therapies for glioblastomas

Rohan Jha 1, Lennard Spanehl 2,3, Jason A Chen 4, Florian A Gessler 5, Omar Arnaout 6, Pablo A Valdes 7, Bryan D Choi 8, Pier Paolo Peruzzi 9, Joshua D Bernstock 10,11,3,, Ennio A Chiocca 12,3,
PMCID: PMC12418593  PMID: 40933034

Abstract

Glioblastoma (GBM) presents significant therapeutic challenges due to the limited efficacy of current treatments. This resistance is multifactorial, stemming from tumor heterogeneity, an immunosuppressive tumor microenvironment, and the restrictive blood-brain barrier, which limits therapeutic access. In response, immunotherapies, particularly tumor vaccines, have emerged as strategies to harness the immune system against these tumors. This review provides an overview of recent advancements and notable clinical trials in tumor vaccine development for GBM. Additionally, it discusses recent preclinical advancements focused on enhancing immune recruitment and response. Identified strategies include peptide, cellular, and nucleic acid vaccines targeting tumor-specific antigens to induce antitumor T-cell responses. Clinical data and preclinical studies exploring various vaccine candidates, adjuvants, and delivery methods demonstrate encouraging results, with some showing improved progression-free and overall survival rates. Despite these advancements, it is clear that further research into personalized vaccines and combination therapies is necessary to enhance immune responses and improve clinical outcomes.

Keywords: clinical trials, glioblastoma (GBM), high-grade gliomas, personalized vaccines, vaccine, vaccine adjuvants


Key Points.

  • Vaccines are a promising treatment for glioblastoma.

  • Personalized vaccines and combination therapy might enhance efficacy.

Gliomas are the most common malignant primary brain tumors, accounting for approximately 80% of all malignant primary CNS tumors.1,2 Among these gliomas, IDH-wildtype glioblastoma (GBM) is associated with very poor outcomes; the median overall survival is approximately 10–12 months, with 5% to 10% survival 5 years postdiagnosis, even with optimal standard of care therapies.1,3 Only 3 therapeutics approved by the U.S. Food and Drug Administration (FDA) have demonstrated a survival benefit: carmustine wafer implants (Gliadel),4 temozolomide and tumor-treating field (TTF) devices.5,6 However, (Gliadel) is costly and has shown high complication rates with only slight improvements in overall survival (OS).7 Consequently, the current standard of care for newly diagnosed GBM is centered on maximally safe surgical resection, followed by radiation therapy and temozolomide, with or without TTF.5,8 Nevertheless, recurrence occurs in nearly all patients, typically at around 7 months posttreatment. Recurrent tumors exhibit increased resistance to treatment, resulting in an even poorer prognosis.9

The resistance of GBMs to treatment, including chemotherapy, radiotherapy, and immunotherapy, is attributed to several factors, such as their substantial molecular heterogeneity and the tumor microenvironment. The blood-brain barrier (BBB) further impedes the efficacy of treatments, contributing in part to the high recurrence rate.10,11 Traditional chemotherapeutics and molecular therapies are only partially and transiently effective against certain subpopulations of the tumor.12,13 This leads to negative selection for resistant subpopulations and phenotype switching, which further fosters therapy resistance.14 Additionally, these gliomas are associated with both local and systemic immunosuppressive states, with various mechanisms and pathways acting to prevent robust immune cell infiltration.15 Notably and relevant to the presentation of tumor antigen(s), endogenous dendritic cells (DC) comprise less than 1% of the total cellular composition surrounding GBM.15–17

Due to the limited success of current standard-of-care therapies, other therapeutic avenues are being explored for GBM treatment, including immune-targeted therapies. These therapies aim to harness the body’s immune system to selectively eliminate tumor cells by generating a tumor-specific response.18 Immune therapeutics for cancer fall into several categories, including immune checkpoint inhibitors, T-cell-targeted therapies, oncolytic viruses (OVs), cytokine-mediated therapies, gene therapies, and therapeutic vaccines.19 We will focus this review primarily on therapeutic vaccines and discuss these grouped by the vaccine type. Tables 13 provide an overview of notable clinical trials utilizing tumor vaccine therapies for GBM, some of which we will discuss in further detail.

Table 1.

Notable Phase I Clinical Trials Utilizing Tumor Vaccine Therapies for High-Grade Gliomas

Trial name Name Sponsor Study start date Completed/expected completion date Enrollment Phase Status
NCT00639639 Vaccine Therapy in Treating Patients With Newly Diagnosed Glioblastoma Multiforme (ATTAC) Duke University 2006-02-06 2022-06-01 42 Phase I COMPLETED
NCT00626483 Basiliximab in Treating Patients With Newly Diagnosed Glioblastoma Multiforme Undergoing Targeted Immunotherapy and Temozolomide-Caused Lymphopenia (REGULATe) Duke University 2007-04-24 2016-07-06 34 Phase I COMPLETED
NCT01621542 Clinical Study of WT2725 in Patients With Advanced Malignancies Sumitomo Pharma America, Inc. 2012-07-31 2017-05-17 64 Phase I COMPLETED
NCT01957956 Vaccine Therapy and Temozolomide in Treating Patients With Newly Diagnosed Glioblastoma Mayo Clinic 2013-11-11 2016-11-16 21 Phase I COMPLETED
NCT02010606 Phase I Study of a Dendritic Cell Vaccine for Patients With Either Newly Diagnosed or Recurrent Glioblastoma Cedars-Sinai Medical Center 2014-01-08 2021-07-10 39 Phase I COMPLETED
NCT02149225 GAPVAC Phase I Trial in Newly Diagnosed Glioblastoma Patients Immatics Biotechnologies GmbH 2014-10 2018-06 16 Phase I COMPLETED
NCT02287428 Personalized NeoAntigen Cancer Vaccine w RT Plus Pembrolizumab for Patients With Newly Diagnosed GBM Dana-Farber Cancer Institute 2014-11 2026-06 56 Phase I RECRUITING
NCT02287428 Personalized NeoAntigen Cancer Vaccine w RT Plus Pembrolizumab for Patients With Newly Diagnosed GBM Dana-Farber Cancer Institute 2014-11 2026-06 56 Phase I RECRUITING
NCT02498665 A Study of DSP-7888 Dosing Emulsion in Adult Patients With Advanced Malignancies Sumitomo Pharma America, Inc. 2015-11 2018-09 24 Phase I COMPLETED
NCT02924038 A Study of Varlilumab and IMA950 Vaccine Plus Poly-ICLC in Patients With WHO Grade II Low-Grade Glioma (LGG) University of California, San Francisco 2017-04-03 2030-12-31 14 Phase I ACTIVE, NOT RECRUITING
NCT03223103 Safety and Immunogenicity of Personalized Genomic Vaccine and Tumor Treating Fields (TTF to Treat Glioblastoma Albert Einstein College of Medicine 2018-03-01 2025-05 13 Phase I ACTIVE, NOT RECRUITING
NCT03299309 PEP-CMV in Recurrent MEdulloblastoma/Malignant Glioma (PRiME) Duke University 2018-06-29 2025-04 30 Phase I ACTIVE, NOT RECRUITING
NCT04015700 Neoantigen-based Personalized DNA Vaccine in Patients With Newly Diagnosed, Unmethylated Glioblastoma Washington University School of Medicine 2020-07-14 2024-12-31 9 Phase I ACTIVE, NOT RECRUITING
NCT04573140 A Study of RNA-lipid Particle (RNA-LP) Vaccines for Newly Diagnosed Pediatric High-Grade Gliomas (pHGG) and Adult Glioblastoma (GBM) (PNOC020) University of Florida 2021-10-26 2027-07 28 Phase I RECRUITING
NCT05283109 ETAPA I: Peptide-based Tumor-Associated Antigen Vaccine in GBM (ETAPA I) Duke University 2023-08-30 2028-02 36 Phase I RECRUITING
NCT05743595 Neoantigen-based Personalized DNA Vaccine With Retifanlimab PD-1 Blockade Therapy in Patients With Newly Diagnosed, Unmethylated Glioblastoma Washington University School of Medicine 2023-10-27 2027-10-31 12 Phase I RECRUITING

Table 3.

Notable Phase III Clinical Trials Utilizing Tumor Vaccine Therapies for High-Grade Gliomas

Trial name Name Sponsor Study start date Completed/expected completion date Enrollment Phase Status
NCT00045968 Study of a Drug [DCVax-L] to Treat Newly Diagnosed GBM Brain Cancer (GBM) Northwest Biotherapeutics 2006-12 2022-11 348 Phase III COMPLETED
NCT01480479 Phase III Study of Rindopepimut/GM-CSF in Patients With Newly Diagnosed Glioblastoma (ACT IV) Celldex Therapeutics 2011-11 2016-11 745 Phase III COMPLETED
NCT03149003 A Study of DSP-7888 Dosing Emulsion in Combination With Bevacizumab in Patients With Recurrent or Progressive Glioblastoma Following Initial Therapy Sumitomo Pharma America, Inc. 2017-12-08 2021-08-30 221 Phase III COMPLETED

Table 2.

Notable Phase II Clinical Trials Utilizing Tumor Vaccine Therapies for High-Grade Gliomas

Trial name Name Sponsor Study start date Completed/expected completion date Enrollment Phase Status
NCT02465268 Vaccine Therapy for the Treatment of Newly Diagnosed Glioblastoma Multiforme (ATTAC-II) University of Florida 2016-08-09 2023-11-30 175 Phase II COMPLETED
NCT02649582 Adjuvant Dendritic Cell-Immunotherapy Plus Temozolomide in Glioblastoma Patients (ADDIT-GLIO) University Hospital, Antwerp 2015-12 2025-12 20 Phase I/II RECRUITING
NCT03879512 Autologous Dendritic Cells, Metronomic Cyclophosphamide and Checkpoint Blockade in Children With Relapsed HGG Wuerzburg University Hospital 2018-02-07 2025-01-31 25 Phase I/II RECRUITING
NCT02455557 SurVaxM Vaccine Therapy and Temozolomide in Treating Patients With Newly Diagnosed Glioblastoma Roswell Park Cancer Institute 2015-05-04 2024-12-30 66 Phase II ACTIVE, NOT RECRUITING
NCT05163080 SurVaxM Plus Adjuvant Temozolomide for Newly Diagnosed Glioblastoma (SURVIVE) (SURVIVE) MimiVax, LLC 2021-11-18 2024-08-18 247 Phase II ACTIVE, NOT RECRUITING
NCT02078648 Safety and Efficacy Study of SL-701, a Glioma-Associated Antigen Vaccine to Treat Recurrent Glioblastoma Multiforme Stemline Therapeutics, Inc. 2014-05 2018-01-22 74 Phase I/II COMPLETED
NCT04013672 Study of Pembrolizumab Plus SurVaxM for Glioblastoma at First Recurrence Case Comprehensive Cancer Center 2020-03-19 2024-05 40 Phase II ACTIVE, NOT RECRUITING
NCT03750071 VXM01 Plus Avelumab Combination Study in Progressive Glioblastoma Vaximm GmbH 2018-11-21 2022-12-31 30 Phase I/II ACTIVE, NOT RECRUITING
NCT01920191 Phase I/II Trial of IMA950 Multi-Peptide Vaccine Plus Poly-ICLC in Glioblastoma University Hospital, Geneva 2013-08 2016-03 19 Phase I/II COMPLETED
NCT03665545 Pembrolizumab in Association With the IMA950/Poly-ICLC for Relapsing Glioblastoma (IMA950-106) University Hospital, Geneva 2018-10-25 2023-12-31 18 Phase I/II ACTIVE, NOT RECRUITING
NCT01280552 A Study of ICT-107 Immunotherapy in Glioblastoma Multiforme (GBM) Precision Life Sciences Group 2011-01 2015-12 124 Phase II COMPLETED
NCT04116658 First-in-Human, Phase 1b/2a Trial of a Multipeptide Therapeutic Vaccine in Patients With Progressive Glioblastoma (ROSALIE) Enterome 2020-07-13 2025-12 100 Phase I/II ACTIVE, NOT RECRUITING
NCT00905060 HSPPC-96 Vaccine With Temozolomide in Patients With Newly Diagnosed GBM (HeatShock) University of California, San Francisco 2009-06-29 2014-06-03 70 Phase II COMPLETED
NCT01814813 Vaccine Therapy With Bevacizumab Versus Bevacizumab Alone in Treating Patients With Recurrent Glioblastoma Multiforme That Can Be Removed by Surgery Alliance for Clinical Trials in Oncology 2013-05 2023-05-01 90 Phase II TERMINATED
NCT03018288 Radiation Therapy Plus Temozolomide and Pembrolizumab With and Without HSPPC-96 in Newly Diagnosed Glioblastoma (GBM) National Cancer Institute (NCI) 2017-09-21 2022-12-20 90 Phase II COMPLETED

Tumor vaccines aim to harness and stimulate the intrinsic immune response by activating effector immune cells to recognize and infiltrate tumors through tumor-specific antigens (TSAs), which are unique to tumor cells, or tumor-associated antigens (TAAs), which are expressed in normal tissues but overexpressed in tumor cells.19,20 Frequently mutated or highly expressed proteins in GBM, such as the TSA epidermal growth factor receptor variant III (EGFRvIII), or TAAs such as telomerase reverse transcriptase (TERT) have been investigated as potential vaccine targets.21 The drawbacks of targeting TSAs include substantial patient heterogeneity, and inconsistent immunogenicity and presentation of neoepitopes, thus often requiring personalized (and therefore expensive and time-consuming) development strategies. On the other hand, TAAs are limited by central tolerance, and cross-reactivity with normal tissues can lead to autoimmunity and side effects including brain inflammation.21,22 By targeting TSAs and/or TAAs, antitumor immune responses can be amplified to induce antitumor T-cell activity and reverse the immunosuppressive tumor environment. The FDA has approved cancer tumor vaccine therapeutics for solid tumors including metastatic melanomas, prostate cancer, and bladder cancer, all of which have shown significant improvements in outcomes.23 Tumor vaccines rely on both the tumor antigen and an immune adjuvant to elicit a robust response. Successful construction of tumor vaccines requires the selection of delivery platform (including cell, peptide, nucleic acid, or viral-based), appropriate antigen(s), and adjuvants.

Dendritic Cell Vaccines

Most contemporary research and successes with cellular vaccines for GBM rely on DCs. DCs are among the most potent antigen-presenting cells (APCs), known for their ability to robustly induce innate and acquired immunity as well as immunity conversion.24 Specifically, DCs phagocytose, process, and present antigens to T-cells via major histocompatibility complex (MHC) molecules, initiating antigen-specific T-cell responses. It is important to distinguish between 2 major populations of DCs, DC1 and DC2, which are phenotypically and functionally different. While lymphoid-related DC2 activate mainly TH2 cells and have shown to generate immune-tolerating responses, myeloid-related DC1 present antigens to MHC class I molecules and exert an immunostimulatory function via TH1 cells.25,26 This process links the innate and adaptive immune systems. When T-cells are primed and activated, they migrate to the tumor site to exert antitumor effects, effectively transforming immunologically “cold” gliomas into more “hot” sites.27,28

The goal of DC vaccines is to amplify this natural process by pulsing autologous or allogeneic DCs with tumor lysate to elicit antigen-specific cytotoxic T-cell responses. For patient-derived approaches, autologous DCs are induced with tumor antigen, treated with cytokines to promote maturation, and then prepared for reinjection into the patient.29,30 The forms of tumor antigens can vary and have included peptides, polypeptides, mRNA, DNA, and whole-tumor lysates.31

DC vaccines can be classified as autologous or allogeneic. Autologous vaccines rely on gathering unique TAAs from the patient’s own tumor.32 This approach theoretically offers tumor specificity, although consistent superior vaccine efficacy has yet to be demonstrated. It also involves more resource utilization and challenges in harvesting and producing sufficient dosages for clinical use. Alternatively, allogeneic vaccines are based on shared antigens found in a high percentage of gliomas, potentially offering a more broadly applicable approach.

DCVax-L is a DC vaccine that utilizes an autologous tumor lysate to initiate a robust immune response.33 The vaccine was evaluated in a phase III clinical trial at multiple sites across multiple countries (NCT00045968), enrolling a total of 331 patients. An initial report from 2018 demonstrated that the median OS was 19.3 months, compared to 16.5 months in a cohort of randomized internal patients and post hoc-introduced synthetic control group.33 The median OS in patients with recurrent GBM was 13.2 months (compared to 7.8 months in the external control cohort). Furthermore, long-term survival appeared to be improved with a 2-year survival rate of 20.7% (compared to 9.6%) and a 5-year survival rate of 13.0% (compared to 5.7%). The study demonstrated negligible toxicity, with only 5 serious adverse events in over 2000 administered doses. Further analysis suggested that the potential survival benefit was maximal in patients age 65 or older, those with subtotal resection at the time of surgery, and those with increased MGMT promoter methylation.33 In addition, multiparametric MRI prediction models were developed to assess treatment response.34 Despite these results, the study failed to meet its primary endpoint, PFS, as reported in a second recent report, with a median PFS of 6.2 months (CI: 5.7–7.4) in the DCVax-L group, and 7.6 months (CI: 5.7–7.4) for the placebo group.35 Though the OS data was reported as significant, their analysis relied on post hoc retrospective analysis with cross-trial comparisons. The change of the primary endpoint to OS, raises concern about the validity of the vaccine.36 Thus, clear conclusion of the efficacy of this vaccine on gliomastoma cannot be drawn, and the post hoc nature of these results is largely hypothesis-generating and requires confirmation in further trials.

Peptide Vaccines

Peptide vaccines rely on the injection and presentation of tumor-specific peptide antigens. These short-chain amino acids contain epitopes specific to the tumor, acting as antigenic targets presented to T-cells in lymphoid tissue. Often, these peptides are conjugated to a carrier protein, which increases immunogenicity and strengthens the adaptive immune response to the tumor.21,37 While single peptides have traditionally been used, recent advances have shifted focus to multi-peptide targets.38

As previously mentioned, GBM is characterized by its tumor heterogeneity and specific genomic mutations, which can lead to antigens that are either unique (TSAs) or expanded in tumors (TAAs) when compared to normal cells. Therefore, these TSAs/TAAs are candidates for eliciting an immune response against tumor cells. Also, single-peptide vaccinations can result in antigen escape, allowing tumors to evade the immune response.39 Strategies to overcome these barriers include multi-peptide proteins and patient-personalized target antigens, which are discussed in further detail below.

Epidermal Growth Factor Receptor Class III variant (EGFR vIII) is a deletion mutation that is heterogeneously expressed in ~1/4–1/3 of GBMs and linked to poor long-term survival, while not found in normal tissue.40 Due to the tumor specificity of EGFR vIII, it served as the target of the rindopepimut (CDX-110), a peptide spanning the mutation site of EGFR vIII conjugated to the immune adjuvant hemocyanin.41 In a phase I clinical trial, the vaccine was shown to eliminate tumor cells while being safe, immunogenic, and tumor-specific.42 In a follow-up phase II study with 65 patients, the median OS was 22 months, with a 36-month OS of 26%, with the authors reporting significantly improved survival compared to historical controls. However, the treatment group was a relatively healthy group, especially compared to the historical controls. As a follow-up, rindopepimut was tested in a phase III trial (NCT01480479) with 745 patients. However, the trial was terminated for futility as the median OS did not improve significantly after a preplanned interim analysis. Data from the study showed that a strong anti-EGFR vIII immune response was generated, suggesting that failure to generate a robust immune response was not the primary limitation for the lack of improved survival. Failure of this study may have been due to over selection of patients and comparison of outcomes with potentially inaccurate historical control in the earlier phases of the study. In light of the failure of this study, there have been substantial efforts to identify methods to improve responses. One approach is T-cell bispecific antibodies (TCBs), which bind both EGFRvIII and the T-cell receptor to specifically activate and recruit T-cells.43 Other approaches include tandem chimeric antigen receptor (CAR) T cells that target both EGFR vIII and IL-13Ra2, which has demonstrated some efficacy in in vitro models.44 Similarly, bispecific T cell engager (BiTE) antibody constructs, designed to engage CD3+ T cells to EGFR vIII, demonstrated safety tolerance in a phase I trial, though the robustness of its antitumor activity remains to be evaluated in larger studies.45

WT1-Based Vaccines

Wilms’ Tumor 1 (WT1) gene is a TAA overexpressed in multiple malignancies, including GBM.46 WT2725 is a WT1-derived oligopeptide vaccine designed to include cytotoxic T-cell responses against advanced malignancies known to overexpress WT1.47 Several phase I dose-escalation studies (NCT01621542, NCT02498665) in patients with WT1-associated tumors, including GBM, did not identify any dose-limiting toxicities of WT1 vaccines, suggesting they are well-tolerated at therapeutic doses.46 Another phase I/II study is currently recruiting, where it aims to evaluate the overall and progression-free survival (PFS) of patients with WT1 mRNA DC vaccination in conjunction with adjuvant temozolomide (NCT02649582). Finally, a large phase III study with 221 patients with GBM is also currently underway, where the DSP-7888 vaccine underwent a dose-escalation trial and is administered in conjunction with bevacizumab (NCT03149003). The interim results suggest that dose-limiting toxicity was not experienced by the cohort, with a larger OS (10.2 [8.2–11.4] months vs. 9.4 [7.4–10.3] months), 1-year OS (37.9 [28.7–47.0]% vs. 31.6 [22.9–40.7]%), and PFS (5.3 [3.9–5.6] months vs. 3.8 [3.7–5.6] months) in patients taking the vaccine, although these differences were not statistically significant.48 Analysis of data across these trials has suggested that maintenance of WT1 expression during the vaccination period could be driving the observed trends of increased PFS and OS.49

Survivin-Based Vaccines

Survivin is an antiapoptotic protein capable of inhibiting caspase activation and is overexpressed in most cancers, including GBM.50 SurVaxM is a peptide vaccine that targets survivin and has been shown to stimulate both T-cell immunity against survivin and inhibit the activity of the survivin pathway.51 In a phase II clinical trial with 64 patients with newly diagnosed GBM (NCT02455557), SurVaxM was well tolerated, with a median PFS of 11.4 months and a median OS of 25.9 months. A larger phase II trial (NCT05163080) is currently ongoing, with an estimated 247 patients enrolled.

Other clinical trials have aimed to combine SurVaxM with other immune peptides to increase the efficacy of the elicited immune response. SL-701 is a vaccine comprised of synthetic peptides targeting interleukin-13 receptor alpha-2, ephrinA2, and survivin.52 In a phase II study with 74 patients (NCT02078648), SL-701 with or without imiquimod and bevacizumab was well tolerated. Results suggested that the median OS was 11 months with imiquimod and 11.7 months with bevacizumab, with a disease control rate of 22% and 54%, respectively.52 Another phase I trial is evaluating the efficacy of a combination of survivin vaccination in conjunction with CMV pp65 and P30-linked EphA2 (NCT05283109). It has enrolled an estimated 36 patients thus far and is estimated to be completed in 2028.53 In another phase II study, 40 patients were treated with SurVaxM and pembrolizumab (NCT04013672). The study is expected to be completed in late 2024 as well.

Heat Shock Protein-Based Vaccines

Heat shock proteins (HSP), which have a role in reassembling misfolded proteins and guiding the degradation of unreducible ones, are found to be upregulated in tumor tissues.54 Vaccines based on HSPs may, in theory, lead to more robust T-cell immune responses due to the highly specific interactions between APCs compared to other tumor vaccines. HSPPC-96 is a vaccine that was developed based on HSPs, and its efficacy was evaluated in a phase II trial (NCT00905060) with 70 patients. When administered in combination with temozolomide, the median OS was 23.8 months.55 A follow-up randomized phase III trial, where HSPPC-96 was administered with bevacizumab, was completed in 2023, and results are pending (NCT01814813). In addition, there is another phase II trial in progress evaluating the efficacy of HSPPC-96 in conjunction with pembrolizumab (NCT03018288).

Multipeptide Vaccines

As highlighted earlier, vaccines targeting multiple peptides to reduce negative selection and immunologic escape are being developed and tested. IMA950 is a multi-peptide vaccine adjuvanted with poly-ICLC, containing 11 TAA peptides.56,57 In a phase I/II clinical trial (NCT01920191), a total of 19 patients were recruited and demonstrated a median OS of 21 months. The trial also demonstrated safety and multi-peptide sustained T cell responses in a subset of patients. A follow-up clinical trial was carried out, where IMA950 was combined with pembrolizumab, in 18 patients (NCT03665545).58 The study concluded in late 2023, and results are currently pending. Another follow-up trial underway is studying the safety and efficacy of varlilumab and IMA950 plus poly-ICLC in grade II low-grade gliomas (NCT02924038).59

ICT-107 is a vaccine based on 6 synthetic peptides targeting those overexpressed in GBM tumor cells.60 In a phase II study, 124 patients with newly diagnosed GBM were randomized to receive ICT-107 or unpulsed controls. Results suggested that the vaccine was well tolerated and efficient, with the median OS increased by 2.0 and median PFS increased by 2.2 months compared to controls. Given the possible signal, especially in HLA A2 positive patients, a phase III trial was planned (NCT01280552) but was terminated due to insufficient funding. Finally, EO2401 is a multi-peptide vaccine that is based on the homologies between TAAs and microbiome-derived peptides.61 These peptides correspond to HLA-A2-restricted peptides with molecular mimicry of 3 TAAs that are often upregulated in GBM. In a phase I/II clinical trial with 100 patients (NCT04116658) where EO2401 was combined with nivolumab and bevacizumab, the interim results suggested that the median OS was 14.5 months, and the median PFS was 5.5 months.61 The trial is expected to be completed in 2025.

Personalized Peptide Vaccines

Peptide-based personalized vaccines rely on targeting of TSAs, which are often unique to patients. These approaches analyze the transcriptomes and immunopeptides of individual tumors, and the relevant targets from a premanufactured library of neoepitopes are chosen as candidates.62 In short, patients were treated with a subset of targets best suited for their tumors. In a phase I clinical trial with 15 patients (NCT02149225), patients were treated with these personalized vaccines derived from biomarker data or tumor genome sequencing, which led to a median PFS of 14.2 months and a median OS of 29.0 months.63 Similarly, in another study using neoantigen prediction, patients were treated with vaccines composed of personalized neoantigen synthetic peptides.64 In the study, 8 patients with GBM underwent personalized neoantigen-targeting vaccines, and the investigators found that 2 patients who did not receive dexamethasone during priming showed increased levels of neoantigen-specific T cells targeted to the tumor. The median PFS was 7.6 months, whereas the median OS was 16.8 months. The study has been expanded to a clinical trial (NCT02287428), which has now recruited 56 patients, with the study estimated to be completed in 2026. Multiple other phase I trials are also currently in progress, evaluating the efficacy of personalized approaches. These approaches range from personalized neoantigen DNA vaccines (where tumor-specific mutations are used to create synthetic DNA plasmids, which produce proteins for immune activation) (NCT04015700), personalized genomic vaccines in conjunction with TTFs (NCT03223103), personalized vaccines with checkpoint blockade in children (NCT03879512), neoantigens with pembrolizumab (NCT02287428), and personalized DNA vaccines with retifanlimab PD-1 blockade (NCT05743595).

Nucleic Acid Vaccines for GBM

Nucleic acid vaccines, which are predominantly messenger RNA (mRNA) vaccines, involve incorporating antigen-specific transcripts into APCs, which subsequently express the antigens to generate both innate and adaptive immune responses.65 Nucleic acids can be introduced to APCs in either an in vivo or ex vivo fashion; at present, ex vivo loading of DCs with mRNA has been the most commonly used approach. mRNA derived from patient tumor cells can be amplified and pulsed into DCs in vitro, requiring only a relatively small number of cells as the source material.66

CMV-Based Vaccines

The cytomegalovirus (CMV) structural protein (pp65) is a phosphoprotein found commonly in GBM (>90%) but not in normal glial tissues.67,68 As a result, it was a candidate antigen sequence for a vaccine, where DCs were electroporated with mRNA encoding CMV pp65. The vaccine was evaluated in a phase I clinical trial in conjunction with temozolomide in 11 patients with newly diagnosed GBM (NCT00639639).68 In this small trial, the vaccine demonstrated safety and improved PFS and overall survival (OS) when compared to historical controls. As a follow-up, 2 clinical trials have been recently completed and are awaiting results, including a phase II trial in conjunction with a tetanus-diphtheria toxoid vaccine (NCT02465268) and a phase I/II trial to identify the vaccine efficacy in patients recovering from lymphopenia caused by temozolomide (NCT00639639).

VBI-1901 is a vaccine utilizing an enveloped virus-like particle targeting the CMV antigens gB and pp65.69 In a phase I/IIa trial with 16 patients, the vaccine demonstrated a median OS of 12.9 months and a 12-month OS of 62.5%. In the ongoing part C of the phase I/II trial (NCT03382977), 60 patients across 8 institutions are to be randomized to the active study arm (VBI-1901 + GM-CSF), with the overall survival to be the primary endpoint.70

In a recently published paper focusing on the delivery of mRNA (encoding for pp65 and whole-tumor antigens) in intravenously administered multilamellar lipid particle aggregates, the authors found that the tumor microenvironment could be reprogrammed with increased gene signatures for antigen presentation and cytotoxicity.71 In a feasibility study (NCT04573140), lipid particle aggregates loaded with pp65 mRNA and autologous whole-tumor mRNA, led to periphery immune activation and a proinflammatory reprogramming of the tumor microenvironment in O6-methylguanine-DNA methyl-transferase (MGMT) unmethylated GBM patients.71 Other trials that are currently active and recruiting include pp65 in conjunction with EphA2 and survivin vaccination (NCT05283109), and pp65 in the treatment of medulloblastoma and malignant glioma (NCT03299309). Furthermore, a recent human trial (NCT04573140) has evaluated feasibility, safety, and activity of multilamellar RNA lipid particle aggregates (LPAs) encoding for pp65, a delivery technique aiming to enhance payload packaging and immunogenicity of vaccines.71 Their results demonstrated improved and rapid cytokine/chemokine release and immune activation/trafficking.

VEGFR-Based Vaccines

VXM01 is a DNA vaccine consisting of an attenuated Salmonella Typhi Typ21a carrying a plasmid encoding for vascular endothelial growth factor receptor-2 (VEGFR-2).72 VEGFR-2 is commonly expressed within tumor microenvironments due to its roles in upregulating angiogenesis and cellular proliferation.73 In a phase I/II clinical trial with 28 included patients (NCT03750071), no treatment-related toxicities were observed when VXM01 was administered with avelumab, an immune checkpoint inhibitor. Interim results suggested that adverse events were mostly unrelated to VXM01, and 3 partial responses with tumor reduction of 58%, 81%, and 95% to baseline were reported in the nonresectable patients (out of 25 total).72 Further analysis of the data suggested that 4 specific T cell clones were significantly enriched in the long-term survivor cohort, suggesting that these clones may have predictive value for stratifying patients prior to vaccination.72

Stem Cell-Based Vaccines

Other preclinical approaches have targeted GBM stem cells. This cell population is known for its resistance to chemotherapy and contributes to the aggressive and highly recurrent nature of GBM.74 By targeting the stem cells, theoretically, a greater antitumor response could be elicited. Approaches to extracting RNA from CD133+ glioma stem cells to selectively target them with DC vaccines have led to robust and long-lasting immune responses in humanized mouse models.75 Mice treated with the approach had a median survival of >60 days, whereas control mice had a median survival of ~40 days. The inhibition of stem cell propagation and tumor growth may lead to decreased immune resistance as these cells are most likely to drive immune resistance.

Other developments have focused on utilizing mRNA from cancer stem cells and transfecting monocyte-derived autologous DCs.76 In this approach, harvested tumor cells are grown as tertiary tumor spheres and whole-cell mRNA is then processed and transfected into DCs. This method has been tested clinically, albeit in a small number of patients (n = 7) who had significantly longer PFS compared to a matched control cohort (694 days vs. 236 days).76

Whole Tumor Lysate Approaches

Though many of the vaccine approaches discussed above have attempted to predict or identify antigens associated with GBM, whole-tumor cell approaches allow investigators to take antigens directly from the tumor. Advantages of this approach include the ability to load DCs with a wide range of antigens, which can provide a more comprehensive immune response and lessen the likelihood of tumor escape due to heterogeneity. In addition, these approaches allow DCs to display nonmutated, but nonetheless posttranslationally modified, antigens that are likely to be missed with conventional peptide prediction vaccination strategies.77 Preclinical studies have found these methods to be superior to peptide-pulsed DC approaches.78 Nonetheless, a challenge with this approach is selectivity in purifying, preparing, and quantifying immunogenic antigens from the whole-tumor lysates. In addition, it is difficult to measure the dose of relevant antigens pulsed into DCs, thus making the standardization and safety evaluation in patients obscure.

A phase I trial used allogenic human GBM tumor cell lysate in conjunction with mature autologous DCs (NCT01957956). Specifically, GBM cell lines derived from patient biopsies were grown, lysed, and loaded into DCs with their optimized manufacturing strategy.79 Of the 21 patients who were enrolled, the median PFS was 9.7 months, the median OS was 19 months, and the OS was 25% and 10% at 2 and 4 years, respectively.79 In another study, autologous DCs were pulsed with lysate derived from a GBM stem-like cell line.80 In this clinical trial (NCT02010606), 11 patients had a median PFS of 8.75 months and OS of 20.36 months. The cell line was safe and well tolerated, and the results suggest potential efficacy.

Strengthening Tumor Vaccine Responses

A prominent limitation of current vaccines is the lack of robust immune system recruitment and response. GBMs are considered “nonsupportive niches,” where even with DC vaccination, T cell states are often immature or insufficiently activated, leading to an antiproliferative state.81 Furthermore, while DC vaccines have been known to partially correct the tolerogenicity of GBM, this alone still remains insufficient. To this end, multiple preclinical approaches have attempted to strengthen the resultant T cell responses following vaccination, including effective homing and persistence of T cells. The GIFT-7 fusokine tumor vaccine is an engineered fusokine of IL-7 and GM-CSF.82 Application of this vaccine to aged mice led to long-term antitumor immunity and a durable long-term increase in proinflammatory cytokines, resulting in hyperactivation of DCs and increased T cell trafficking to tumor sites.82 In another study, authors found that Tc17-1 cells (CD8+ T cells that produced IL-17 and IFN-γ) combined with poly-ICLC and Pmel-1 peptide vaccine significantly prolonged the survival in animal models.83 Another vaccine platform (ITI-1001) utilized a DNA vaccine that codes for the HCMV protein pp65, gB, and IE-1. In a syngenic orthotopic GBM model, ITI-1001 resulted in strong cellular and humoral immune response, antitumor activity, and enhanced survival of tumor-bearing mice (56%).84 Specifically, the authors observed increased activation of Th1, cytotoxic CD8, reg T cells, and IFN-γ+ CD4 T cells. Finally, an autologous and allogenic mixed glioma cell lysate vaccine in a rat model was developed to reverse tumor-associated immunosuppression.85 The vaccine not only inhibited glioma cell proliferation, it also promoted their apoptosis, leading to a significantly improved survival time for rats. The results were thought to be due to the enhanced secretion of T cell chemokines monocyte chemotactic protein-2 (MCP-2), interferon gamma (IFN-γ), and interleukin 2 (IL-2), which stimulated the proliferation of T cells and NK cells.

Adjuvants

Adjuvant immune response modifiers may potentiate vaccine efficacy and immunogenicity. The addition of Type I interferon and synthetic TLR3 activator poly (I:C) improves DC function, leading to sustained cytokine production postvaccination and a corresponding survival benefit.86 Additional strategies to improve the inflammatory milieu at the tumor site include the administration of granulocyte-macrophage colony-stimulating factor (GM-CSF),68 poly(I:C),86 cytokine IL-12,87 and other TLR agonists.24 Bevacizumab, a monoclonal antibody against VEGF, may lead to improvement in antigen presentation, lymphocyte trafficking, and DC maturation for cell-based vaccines.88 Montanide is an emulsion that has been shown to prolong the release of vaccine antigens. Two trials (NCT02455557, NCT02454634) have shown no associated serious adverse events.51,89 Prevaccination with the tetanus-diphtheria toxoid vaccine has been shown to improve DC migration and efficacy, suppress tumor growth, better lymph node homing, and prolong survival.90 Similarly, synthetic immunomodulatory agents, such as resiquimod and imiquimod, have been shown to enhance cytokine production and skew immunity toward a Th1 response.91,92 Combination with immune checkpoint inhibitors, which aim to reverse the immunosuppressive microenvironment and improve non-exhausting T-cell homing to the glioma site is also currently being investigated in multiple clinical trials.93

An adjuvant that has been explored in the preclinical setting is lenalidomide.94 In a murine GL261 intracranial glioma model, multi-epitope peptides in combination with lenalidomide and anti-PD-1 prolonged the survival of mice by suppressing tumor growth. An increase in the percentage of regulatory T cells was seen, as well as an enhanced cytotoxicity against GL261. Another study investigated the effectiveness of DCs pulsed with protein antigens and anti-PD-1 and found enhanced expression of MHC and costimulatory markers, resulting in increased IFN-γ+ effector T cells and enhanced lysis of GBM target cells.95 Finally, another preclinical study showed that combinatorial treatment with branched multi-peptide and peptide adjuvants such as PADRE and poly-ICLC, anti-PD, and radiation could enhance the antitumor effects of radiotherapy in a GBM mouse model.96

Future Directions

Many candidate vaccines failed to impact clinically meaningful endpoints, likely due to nonrobust and nonsustained immunogenicity. Alternative strategies to increase immune activation or increase the potency of vaccines may augment their efficacy. Analysis from personalized vaccine clinical trials has suggested that corticosteroids should be avoided, and additional measures to combat T-cell exhaustion are required.97 The lack of success with immune monotherapies has suggested that combination therapies that target multiple immunological mechanisms in coordination may lead to a stronger immune response characterized by improved APC immunogenicity, leading to increased cytokine production, migration to the tumor, and increased effectiveness at the tumor site.97

Combinatorial Approaches

Due to the lack of success observed with monotherapies, combining multiple different immunological therapies could represent a promising avenue for clinical success. The combination of vaccines with OVs and chimeric antigen receptor T-cell therapy (CAR-T) represents a coordinated approach that comprehensively utilizes multiple immune mechanisms and pathways. CAR-T therapy relies on modified T cells to initiate cytotoxic attacks on targeted antigen-bearing tumor cells, thereby bypassing the MHC presentation pathway.98 Thus, vaccines in conjunction with CAR-T therapy represent tumor treatment via complementary immunologic arms. Multiple clinical trials have been carried out with CAR-T therapy, where although safety and feasibility have been shown, limited efficacy has been demonstrated.99–101 This is likely again in part due to tumor heterogeneity and antigen escape. Targeting multiple epitopes via personalized approaches could, in theory, expand the coverage of GBM and provide notable clinical efficacy. OVs selectively infect and replicate in tumor cells to both lyse tumors and stimulate an antitumor immune response.102 Multiple clinical trials have demonstrated safety and efficacy of these modalities,103 though there remains ongoing exploration in vector and delivery design to improve efficacy of these novel treatment modalities. Combination with vaccine therapies may promote synergy in mechanisms, where OVs could traffic more easily to cells being targeted by vaccines, whereas oncolysis may limit immunological escape against vaccines. Notably, in a recent Nature Cancer publication, Chen et al. showed that OV can be used to induce the expression of bystander T cell epitopes in tumor cells. These are tumor-irrelevant antigens (often viral) that are recognized by bystander T cells, leading to antitumoral cytotoxicity and improved tumor control in preclinical models of numerous malignancies.104 Robust preclinical and clinical investigations are required to assess these possible interactions.

Delivery

The majority of vaccination trials in GBM rely on subcutaneous, intradermal, or intravenous delivery methods. However, there are recent advancements aiming to improve the efficacy of local therapy delivery, including polymeric wafers, nanofibrous scaffolds, and hydrogels.105,106 In one study, a hydrogel vaccine system was designed containing granulocyte-macrophage colony-stimulating factor (GM-CSF) and tumor cell lysates and implanted at the surgical site.107 The personalized hydrogel was able to inhibit tumor recurrence, and coupled with findings from another earlier study implementing polypeptide hydrogels,108 suggests that hydrogels can be used as an effective sustained delivery platform for vaccines in GBM. Another example is mitoxantrone-loaded PEGylated PLGA-based nanoparticles (NP-MTX).109 A study utilizing intratumoral NP-MTX delivery showed that it increased immune cell infiltration, specifically increased frequency of IFN-y secreting CD8 T cells and M1-like macrophages. More importantly, the study was able to demonstrate that vaccines administered via this approach increased the median survival of GL261-bearing mice. Thus, this delivery method may lead to more effective immune cell recruitment, allowing for possibly increased median survival.

Conclusion

Therapeutics for GBM continue to be limited due to tumor heterogeneity, immunosuppressive microenvironment, and the BBB. Vaccine therapeutics employing multiple approaches have been investigated in the preclinical and clinical settings. Whereas monotherapy targeting single epitopes formed the foundation of earlier investigations, the nature of immunologic escape and lack of clinical success is guiding the field away from single-peptide vaccines. Vaccines targeting multiple targets likely form the cornerstone of future advancement, though optimization to improve potency is paramount. Furthermore, these multiple targets are not consistently conserved across patients and tumors, and thus personalized approaches, with hierarchical identification of best target mutations, could improve vaccine efficacy and potency. Finally, adjuvants and combination therapy to improve vaccine delivery and stimulate the immune system are critical to improving possible clinical responses. Though there are encouraging results from preclinical and Phase I clinical investigations, further Phase II and III investigations are required to optimize and realize the clinical potential of vaccines for GBM. Vaccines are a promising treatment for GBM; while significant clinical efficacy has yet to be conclusively demonstrated, new therapeutic strategies to overcome tumor heterogeneity and immune escape may soon bring them into more widespread clinical use.

Contributor Information

Rohan Jha, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.

Lennard Spanehl, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Neurosurgery, Rostock University Medical Center, Rostock, Germany.

Jason A Chen, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.

Florian A Gessler, Department of Neurosurgery, Rostock University Medical Center, Rostock, Germany.

Omar Arnaout, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.

Pablo A Valdes, Department of Neurosurgery, University of Texas at Galveston, Galveston, Texas, USA.

Bryan D Choi, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.

Pier Paolo Peruzzi, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.

Joshua D Bernstock, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.

Ennio A Chiocca, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA.

Supplement sponsorship

This article appears as part of the supplement “Immunotherapy for Brain Tumors,” sponsored by the Wilkins Family Chair in Neurosurgical Brain Tumor Research.

Conflict of interest statement

The authors have no personal, financial, or institutional interest in any of the drugs, materials, or devices described in this article. J.D.B. has an equity position in Treovir Inc. and UpFront Diagnostics. J.D.B. is also a cofounder of Centile Bioscience and on the NeuroX1 and QV Bioelectronics scientific advisory boards. J.A.C. is a cofounder and holds equity in Verge Genomics. J.A.C. is also an advisor and holds equity in Gravity Medical Technology. F.A.G. received honoraria from Signus, BBraun, Aesculap, and AstraZeneca.

References

  • 1. Ostrom QT, Price M, Neff C, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2015–2019. Neuro Oncol. 2022;24(Suppl_5):v1–v95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23(8):1231–1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Chien LN, Gittleman H, Ostrom QT, et al. Comparative brain and central nervous system tumor incidence and survival between the United States and Taiwan based on population-based registry. Front Public Health. 2016;4(151):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Westphal M, Hilt DC, Bortey E, et al. A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma. Neuro Oncol. 2003;5(2):79–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Wen PY, Weller M, Lee EQ, et al. Glioblastoma in adults: a Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) consensus review on current management and future directions. Neuro Oncol. 2020;22(8):1073–1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Perry J, Chambers A, Spithoff K, Laperriere N.. Gliadel wafers in the treatment of malignant glioma: a systematic review. Curr Oncol. 2007;14(5):189–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Fisher JP, Adamson DC.. Current FDA-approved therapies for high-grade malignant gliomas. Biomedicines. 2021;9(3):324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Stupp R, Taillibert S, Kanner AA, et al. Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial. JAMA. 2015;314(23):2535–2543. [DOI] [PubMed] [Google Scholar]
  • 9. Korja M, Raj R, Seppä K, et al. Glioblastoma survival is improving despite increasing incidence rates: a nationwide study between 2000 and 2013 in Finland. Neuro Oncol. 2019;21(3):370–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Goenka A, Tiek D, Song X, et al. The many facets of therapy resistance and tumor recurrence in glioblastoma. Cells. 2021;10(3):484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Ou A, Yung WKA, Majd N.. Molecular mechanisms of treatment resistance in glioblastoma. Int J Mol Sci. 2020;22(1):351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Perrin SL, Samuel MS, Koszyca B, et al. Glioblastoma heterogeneity and the tumour microenvironment: implications for preclinical research and development of new treatments. Biochem Soc Trans. 2019;47(2):625–638. [DOI] [PubMed] [Google Scholar]
  • 13. Bernstock JD, Mooney JH, Ilyas A, et al. Molecular and cellular intratumoral heterogeneity in primary glioblastoma: clinical and translational implications. J Neurosurg. 2020;133(3):655–663. [DOI] [PubMed] [Google Scholar]
  • 14. Wang L, Jung J, Babikir H, et al. A single-cell atlas of glioblastoma evolution under therapy reveals cell-intrinsic and cell-extrinsic therapeutic targets. Nat Cancer. 2022;3(12):1534–1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Woroniecka K, Chongsathidkiet P, Rhodin K, et al. T-cell exhaustion signatures vary with tumor type and are severe in glioblastoma. Clin Cancer Res. 2018;24(17):4175–4186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Jackson CM, Choi J, Lim M.. Mechanisms of immunotherapy resistance: lessons from glioblastoma. Nat Immunol. 2019;20(9):1100–1109. [DOI] [PubMed] [Google Scholar]
  • 17. Friebel E, Kapolou K, Unger S, et al. Single-cell mapping of human brain cancer reveals tumor-specific instruction of tissue-invading leukocytes. Cell. 2020;181(7):1626–1642.e20. [DOI] [PubMed] [Google Scholar]
  • 18. Martin-Liberal J, Ochoa De Olza M, Hierro C, et al. The expanding role of immunotherapy. Cancer Treat Rev. 2017;54(2):74–86. [DOI] [PubMed] [Google Scholar]
  • 19. Sener U, Ruff MW, Campian JL.. Immunotherapy in glioblastoma: current approaches and future perspectives. Int J Mol Sci. 2022;23(13):7046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Esfahani K, Roudaia L, Buhlaiga N, et al. A review of cancer immunotherapy: from the past, to the present, to the future. Curr Oncol. 2020;27(12):87–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Swartz AM, Batich KA, Fecci PE, Sampson JH.. Peptide vaccines for the treatment of glioblastoma. J Neurooncol. 2015;123(3):433–440. [DOI] [PubMed] [Google Scholar]
  • 22. Kanaly CW, Ding D, Heimberger AB, Sampson JH.. Clinical applications of a peptide-based vaccine for glioblastoma. Neurosurg Clin N Am. 2010;21(1):95–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Saxena M, Van Der Burg SH, Melief CJM, Bhardwaj N.. Therapeutic cancer vaccines. Nat Rev Cancer. 2021;21(6):360–378. [DOI] [PubMed] [Google Scholar]
  • 24. Prins RM, Soto H, Konkankit V, et al. Gene expression profile correlates with T-cell infiltration and relative survival in glioblastoma patients vaccinated with dendritic cell immunotherapy. Clin Cancer Res. 2011;17(6):1603–1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Schlitzer A, Zhang W, Song M, Ma X.. Recent advances in understanding dendritic cell development, classification, and phenotype. F1000Res. 2018;7:F1000 Faculty Rev-1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Reinhard G, Märten A, Kiske SM, et al. Generation of dendritic cell-based vaccines for cancer therapy. Br J Cancer. 2002;86(10):1529–1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Frederico SC, Hancock JC, Brettschneider EES, et al. Making a cold tumor hot: the role of vaccines in the treatment of glioblastoma. Front Oncol. 2021;11:672508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18(1):767–811. [DOI] [PubMed] [Google Scholar]
  • 29. Nair S, Archer GE, Tedder TF.. Isolation and generation of human dendritic cells. Curr Protoc Immunol. 2012;Chapter 7:7.32.1–7.32.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Palucka K, Banchereau J.. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12(4):265–277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Wilde S, Sommermeyer D, Frankenberger B, et al. Dendritic cells pulsed with RNA encoding allogeneic MHC and antigen induce T cells with superior antitumor activity and higher TCR functional avidity. Blood. 2009;114(10):2131–2139. [DOI] [PubMed] [Google Scholar]
  • 32. Smith C, Lineburg KE, Martins JP, et al. Autologous CMV-specific T cells are a safe adjuvant immunotherapy for primary glioblastoma multiforme. J Clin Invest. 2020;130(11):6041–6053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Liau LM, Ashkan K, Tran DD, et al. First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J Transl Med. 2018;16(1):142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. De Godoy LL, Chawla S, Brem S, et al. Assessment of treatment response to dendritic cell vaccine in patients with glioblastoma using a multiparametric MRI-based prediction model. J Neurooncol. 2023;163(1):173–183. [DOI] [PubMed] [Google Scholar]
  • 35. Liau LM, Ashkan K, Brem S, et al. Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma: a phase 3 prospective externally controlled cohort trial. JAMA Oncol. 2023;9(1):112–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Preusser M, van den Bent MJ.. Autologous tumor lysate-loaded dendritic cell vaccination (DCVax-L) in glioblastoma: breakthrough or fata morgana? Neuro Oncol. 2023;25(4):631–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Lynn GM, Sedlik C, Baharom F, et al. Peptide–TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigens. Nat Biotechnol. 2020;38(3):320–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Liu W, Tang H, Li L, et al. Peptide‐based therapeutic cancer vaccine: current trends in clinical application. Cell Prolif. 2021;54(5):e13025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Sampson JH, Heimberger AB, Archer GE, 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(31):4722–4729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Weller M, Kaulich K, Hentschel B, et al. ; German Glioma Network. Assessment and prognostic significance of the epidermal growth factor receptor vIII mutation in glioblastoma patients treated with concurrent and adjuvant temozolomide radiochemotherapy: EGFRvIII mutation and prognosis of glioblastoma. Int J Cancer. 2014;134(10):2437–2447. [DOI] [PubMed] [Google Scholar]
  • 41. Swartz AM, Li QJ, Sampson JH.. Rindopepimut: a promising immunotherapeutic for the treatment of glioblastoma multiforme. Immunotherapy. 2014;6(6):679–690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Sampson JH, Archer GE, Mitchell DA, et al. An epidermal growth factor receptor variant III-targeted vaccine is safe and immunogenic in patients with glioblastoma multiforme. Mol Cancer Ther. 2009;8(10):2773–2779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Iurlaro R, Waldhauer I, Planas-Rigol E, et al. A novel EGFRvIII T-cell bispecific antibody for the treatment of glioblastoma. Mol Cancer Ther. 2022;21(10):1499–1509. [DOI] [PubMed] [Google Scholar]
  • 44. Schmidts A, Srivastava AA, Ramapriyan R, et al. Tandem chimeric antigen receptor (CAR) T cells targeting EGFRvIII and IL-13Rα2 are effective against heterogeneous glioblastoma. Neurooncol. Adv. 2023;5(1):vdac185vdac185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Rosenthal MA, Balana C, Van Linde ME, et al. ATIM-49 (LTBK-01). AMG 596, a novel anti-EGFRVIII bispecific T cell engager (BITE®) molecule for the treatment of glioblastoma (GBM): planned interim analysis in recurrent GBM (RGBM). Neuro Oncol. 2019;21(Suppl_6):vi283–vi283. [Google Scholar]
  • 46. Spira A, Hansen AR, Harb WA, et al. Multicenter, open-label, phase I study of DSP-7888 dosing emulsion in patients with advanced malignancies. Target Oncol. 2021;16(4):461–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Fu S, Piccioni DE, Liu H, et al. A phase I study of the WT2725 dosing emulsion in patients with advanced malignancies. Sci Rep. 2021;11(1):22355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Sumitomo Pharma America, Inc. A randomized, multicenter, adaptive phase 3 study of DSP-7888 dosing emulsion in combination with bevacizumab versus bevacizumab alone in patients with recurrent or progressive glioblastoma following initial therapy (WIZARD 201G). clinicaltrials.gov; 2023. https://clinicaltrials.gov/study/NCT03149003. Accessed December 8, 2024. [Google Scholar]
  • 49. Yokota C, Kagawa N, Takano K, et al. Maintenance of WT1 expression in tumor cells is associated with a good prognosis in malignant glioma patients treated with WT1 peptide vaccine immunotherapy. Cancer Immunol Immunother. 2022;71(1):189–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Wheatley SP, Altieri DC.. Survivin at a glance. J Cell Sci. 2019;132(7):jcs223826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Ahluwalia MS, Reardon DA, Abad AP, et al. Phase IIa study of SurVaxM plus adjuvant temozolomide for newly diagnosed glioblastoma. J Clin Oncol. 2023;41(7):1453–1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Peereboom D, Nabors LB, Kumthekar P, et al. Results of phase II trial of SL-701, a novel immunotherapy targeting IL-13Ra2, EphA2, and survivin, in adults with second-line recurrent glioblastoma (GBM). Ann Oncol. 2018;29(Suppl 8):viii122–viii123. [Google Scholar]
  • 53. Khasraw M. ETAPA I: Evaluation of tumor associated P30-peptide antigen I; a pilot trial of peptide-based tumor associated antigen vaccines in newly diagnosed, unmethylated, and untreated glioblastoma (GBM). clinicaltrials.gov; 2024. https://clinicaltrials.gov/study/NCT05283109. Accessed February 17, 2025. [Google Scholar]
  • 54. Hu C, Yang J, Qi Z, et al. Heat shock proteins: biological functions, pathological roles, and therapeutic opportunities. MedComm (2020). 2022;3(3):e161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Bloch O, Lim M, Sughrue ME, et al. Autologous heat shock protein peptide vaccination for newly diagnosed glioblastoma: impact of peripheral PD-L1 expression on response to therapy. Clin Cancer Res. 2017;23(14):3575–3584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Rampling R, Peoples S, Mulholland PJ, et al. A cancer research UK first time in human phase I trial of IMA950 (Novel Multipeptide Therapeutic Vaccine) in patients with newly diagnosed glioblastoma. Clin Cancer Res. 2016;22(19):4776–4785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Migliorini D, Dutoit V, Allard M, et al. 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(7):923–933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. National Brain Tumor Society | Community here. Breakthroughs ahead. National Brain Tumor Society. https://braintumor.org/. Accessed February 17, 2025. [Google Scholar]
  • 59.Glioblastoma multiforme multipeptide vaccine IMA950 and poly ICLC with or without Varlilumab in treating patients with newly diagnosed or recurrent grade ii glioma - NCI. June 23, 2016. https://www.cancer.gov/about-cancer/treatment/clinical-trials/search/v?id=NCI-2017-01369. Accessed February 17, 2025. [Google Scholar]
  • 60. Wen PY, Reardon DA, Armstrong TS, et al. 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(19):5799–5807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Wick W, Idbaih A, Tabatabai G, et al. EO2401, a novel microbiome-derived therapeutic vaccine for patients with recurrent glioblastoma: ROSALIE study. J Clin Oncol. 2022;40(16_suppl):2034–2034. [Google Scholar]
  • 62. Hilf N, Kuttruff-Coqui S, Frenzel K, et al. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature. 2019;565(7738):240–245. [DOI] [PubMed] [Google Scholar]
  • 63. Wick W, Dietrich PY, Kuttruff S, et al. ; GAPVAC Consortium. GAPVAC-101: first-in-human trial of a highly personalized peptide vaccination approach for patients with newly diagnosed glioblastoma. J Clin Oncol. 2018;36(15_suppl):2000–2000. [Google Scholar]
  • 64. Keskin DB, Anandappa AJ, Sun J, et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature. 2019;565(7738):234–239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Yang B, Jeang J, Yang A, Wu TC, Hung CF.. DNA vaccine for cancer immunotherapy. Human Vaccines Immunother. 2014;10(11):3153–3164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Ashley DM, Faiola B, Nair S, et al. Bone marrow-generated dendritic cells pulsed with tumor extracts or tumor RNA induce antitumor immunity against central nervous system tumors. J Exp Med. 1997;186(7):1177–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Ranganathan P, Clark PA, Kuo JS, Salamat MS, Kalejta RF.. Significant association of multiple human cytomegalovirus genomic Loci with glioblastoma multiforme samples. J Virol. 2012;86(2):854–864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Batich KA, Reap EA, Archer GE, et al. Long-term survival in glioblastoma with cytomegalovirus pp65-targeted vaccination. Clin Cancer Res. 2017;23(8):1898–1909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.By. VBI Vaccines Presents Encouraging Early Tumor Response Data From Randomized Controlled Phase 2b Study of VBI-1901 in Recurrent Glioblastoma. VBI Vaccines. April 3, 2024. https://www.vbivaccines.com/press-releases/vbi-vaccines-presents-encouraging-early-tumor-response-data-from-randomized-controlled-phase-2b-study-of-vbi-1901-in-recurrent-glioblastoma/. Accessed October 14, 2024.
  • 70. Merrell RT, Wen PY, Forst DA, et al. Randomized phase IIb trial of a CMV vaccine immunotherapeutic candidate (VBI-1901) in recurrent glioblastomas. J Clin Oncol. 2024;42(16_suppl):TPS2100–TPS2100. [Google Scholar]
  • 71. Mendez-Gomez HR, DeVries A, Castillo P, et al. RNA aggregates harness the danger response for potent cancer immunotherapy. Cell. 2024;187(10):2521–2535.e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Wick W, Wick A, Chinot O, et al. KS05.6.A Oral DNA vaccination targeting VEGFR2 combined with the anti-PD-L1 antibody avelumab in patients with progressive glioblastoma—final results. NCT03750071. Neuro-Oncology. 2022;24(Suppl_2):ii6–ii6. [Google Scholar]
  • 73. Zhang Y, Huang H, Coleman M, et al. VEGFR2 activity on myeloid cells mediates immune suppression in the tumor microenvironment. JCI Insight. 2021;6(23):e150735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Fidoamore A, Cristiano L, Antonosante A, et al. Glioblastoma stem cells microenvironment: the paracrine roles of the Niche in drug and radioresistance. Stem Cells Int. 2016;2016:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Do ASMS, Amano T, Edwards LA, et al. CD133 mRNA-loaded dendritic cell vaccination abrogates glioma stem cell propagation in humanized glioblastoma mouse model. Mol Ther Oncolytics. 2020;18:295–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Vik-Mo EO, Nyakas M, Mikkelsen BV, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013;62(9):1499–1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Kacen A, Javitt A, Kramer MP, et al. Post-translational modifications reshape the antigenic landscape of the MHC I immunopeptidome in tumors. Nat Biotechnol. 2023;41(2):239–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Grauer OM, Sutmuller RPM, Van Maren W, et al. Elimination of regulatory T cells is essential for an effective vaccination with tumor lysate‐pulsed dendritic cells in a murine glioma model. Int J Cancer. 2008;122(8):1794–1802. [DOI] [PubMed] [Google Scholar]
  • 79. Parney IF, Anderson SK, Gustafson MP, et al. Phase I trial of adjuvant mature autologous dendritic cell/allogeneic tumor lysate vaccines in combination with temozolomide in newly diagnosed glioblastoma. Neurooncol. Adv. 2022;4(1):vdac089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Hu JL, Omofoye OA, Rudnick JD, et al. 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. Clinical Cancer Res. 2022;28(4):689–696. [DOI] [PubMed] [Google Scholar]
  • 81. Naulaerts S, Datsi A, Borras DM, et al. Multiomics and spatial mapping characterizes human CD8+ T cell states in cancer. Sci Transl Med. 2023;15(691):eadd1016. [DOI] [PubMed] [Google Scholar]
  • 82. Shireman JM, Gonugunta N, Zhao L, et al. GM-CSF and IL-7 fusion cytokine engineered tumor vaccine generates long-term Th-17 memory cells and increases overall survival in aged syngeneic mouse models of glioblastoma. Aging Cell. 2023;22(7):e13864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Ohkuri T, Kosaka A, Ikeura M, Salazar AM, Okada H.. IFN-γ- and IL-17-producing CD8+ T (Tc17-1) cells in combination with poly-ICLC and peptide vaccine exhibit antiglioma activity. J ImmunoTher Cancer. 2021;9(6):e002426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Adhikari AS, Macauley J, Johnson Y, et al. Development and characterization of an HCMV multi-antigen therapeutic vaccine for glioblastoma using the UNITE platform. Front Oncol. 2022;12:850546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. He H, Cen Y, Wang P, et al. The therapeutic effect of an autologous and allogenic mixed glioma cell lysate vaccine in a rat model. J Cancer Res Clin Oncol. 2023;149(2):609–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Okada H, Kalinski P, Ueda R, 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(3):330–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Teng MWL, Bowman EP, McElwee JJ, et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med. 2015;21(7):719–729. [DOI] [PubMed] [Google Scholar]
  • 88. Brown NF, Carter TJ, Ottaviani D, Mulholland P.. Harnessing the immune system in glioblastoma. Br J Cancer. 2018;119(10):1171–1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Platten M, Bunse L, Wick A, et al. A vaccine targeting mutant IDH1 in newly diagnosed glioma. Nature. 2021;592(7854):463–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Mitchell DA, Batich KA, Gunn MD, et al. Tetanus toxoid and CCL3 improve dendritic cell vaccines in mice and glioblastoma patients. Nature. 2015;519(7543):366–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Rozenblit M, Hendrickx W, Heguy A, et al. Transcriptomic profiles conducive to immune-mediated tumor rejection in human breast cancer skin metastases treated with Imiquimod. Sci Rep. 2019;9(1):8572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Jansen MHE, Mosterd K, Arits AHMM, et al. Five-year results of a randomized controlled trial comparing effectiveness of photodynamic therapy, topical imiquimod, and topical 5-fluorouracil in patients with superficial basal cell carcinoma. J Invest Dermatol. 2018;138(3):527–533. [DOI] [PubMed] [Google Scholar]
  • 93. Ser MH, Webb MJ, Sener U, Campian JL.. Immune checkpoint inhibitors and glioblastoma: a review on current state and future directions. J Immunother Precis Oncol. 2024;7(2):97–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Tran TAT, Kim YH, Kim GE, et al. The long multi-epitope peptide vaccine combined with adjuvants improved the therapeutic effects in a glioblastoma mouse model. Front Immunol. 2022;13:1007285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Kim YH, Tran TAT, Duong THO, et al. Feasibility of dendritic cell-based vaccine against glioblastoma by using cytoplasmic transduction peptide (CTP)-fused protein antigens combined with anti-PD1. Human Vaccines Immunother. 2020;16(11):2840–2848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Tran TAT, Kim YH, Duong THO, et al. Peptide vaccine combined adjuvants modulate anti-tumor effects of radiation in glioblastoma mouse model. Front Immunol. 2020;11:1165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Sampson JH, Gunn MD, Fecci PE, Ashley DM.. Brain immunology and immunotherapy in brain tumours. Nat Rev Cancer. 2020;20(1):12–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Sadelain M, Rivière I, Riddell S.. Therapeutic T cell engineering. Nature. 2017;545(7655):423–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. O’Rourke DM, Nasrallah MP, Desai 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(399):eaaa0984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Brown CE, Badie B, Barish ME, et al. Bioactivity and safety of IL13Rα2-redirected chimeric antigen receptor CD8+ T cells in patients with recurrent glioblastoma. Clinical Cancer Res. 2015;21(18):4062–4072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Luksik AS, Yazigi E, Shah P, Jackson CM.. CAR T cell therapy in glioblastoma: overcoming challenges related to antigen expression. Cancers (Basel). 2023;15(5):1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Kang KD, Bernstock JD, Totsch SK, et al. Safety and efficacy of intraventricular immunovirotherapy with oncolytic HSV-1 for CNS cancers. Clin Cancer Res. 2022;28(24):5419–5430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Bernstock JD, Blitz SE, Hoffman SE, et al. Recent oncolytic virotherapy clinical trials outline a roadmap for the treatment of high-grade glioma. Neurooncol Adv. 2023;5(1):vdad081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Chen X, Zhao J, Yue S, et al. An oncolytic virus delivering tumor-irrelevant bystander T cell epitopes induces anti-tumor immunity and potentiates cancer immunotherapy. Nat Cancer. 2024;5(July 2024):1063–1081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Bu LL, Yan J, Wang Z, et al. Advances in drug delivery for post-surgical cancer treatment. Biomaterials. 2019;219(2019):119182. [DOI] [PubMed] [Google Scholar]
  • 106. Cha GD, Kang T, Baik S, et al. Advances in drug delivery technology for the treatment of glioblastoma multiforme. J Control Release. 2020;328(2020):350–367. [DOI] [PubMed] [Google Scholar]
  • 107. Lu Y, Wu C, Yang Y, et al. Inhibition of tumor recurrence and metastasis via a surgical tumor-derived personalized hydrogel vaccine. Biomater Sci. 2022;10(5):1352–1363. [DOI] [PubMed] [Google Scholar]
  • 108. Song H, Yang P, Huang P, et al. Injectable polypeptide hydrogel-based co-delivery of vaccine and immune checkpoint inhibitors improves tumor immunotherapy. Theranostics. 2019;9(8):2299–2314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Bausart M, Rodella G, Dumont M, et al. Combination of local immunogenic cell death-inducing chemotherapy and DNA vaccine increases the survival of glioblastoma-bearing mice. Nanomed Nanotechnol Biol Med. 2023;50(2023):102681. [DOI] [PubMed] [Google Scholar]

Articles from Neuro-Oncology Advances are provided here courtesy of Oxford University Press

RESOURCES