Home
JournalsCollections
For Authors For Reviewers For Editorial Board Members
Article Processing Charges Open Access
Ethics Advertising Policy
Editorial Policy Resource Center
Company Information Contact Us Membership Collaborators Partners
Publications > Journals > Oncology Advances> Article Full Text
OPEN ACCESS

Dual Challenges and Innovative Strategies in Chimeric Antigen Receptor T-cell Therapy for Glioblastoma

  • Liangjin Zhang1,2,#,
  • Zhiqiang Zhang1,3,#,
  • Jiale He1,4,#,
  • Zhiheng Zhang1,5,
  • Huaixiang Zhou1,
  • Youheng Jiang1,3,
  • Xin Zhong1,3,
  • Yanming Yang1,
  • Ningning Li1,6,
  • Wu Xu7,* ,
  • Yulong He3,*  and
  • Qunlong Jin1,3,* 
Oncology Advances   2025

doi: 10.14218/OnA.2025.00014

Received:

Revised:

Accepted:

Published online:

 Author information

Citation: Zhang L, Zhang Z, He J, Zhang Z, Zhou H, Jiang Y, et al. Dual Challenges and Innovative Strategies in Chimeric Antigen Receptor T-cell Therapy for Glioblastoma. Oncol Adv. Published online: Jul 30, 2025. doi: 10.14218/OnA.2025.00014.

Abstract

Glioblastoma (GBM) is the most prevalent and aggressive form of primary brain malignancy in adults. Despite continuous advancements in standard treatment modalities, the prognosis for patients remains extremely poor, with a median survival of less than two years. In recent years, chimeric antigen receptor T-cell (CAR-T) therapy has achieved revolutionary success in hematologic malignancies, marking a significant breakthrough in the field of immunotherapy. However, the successful application of CAR-T therapy to GBM still faces dual challenges: antigen heterogeneity and the immunosuppressive tumor microenvironment. This review systematically summarizes these challenges encountered in CAR-T therapy for GBM and the innovative strategies currently under development to address these challenges, providing insights for the future clinical translation of CAR-T therapy in GBM.

Keywords

Glioblastoma, Chimeric antigen receptor T-cell, CAR-T cells, Antigen heterogeneity, Immunosuppressive barrier, Innovative therapeutic strategies, Prospective technology

Introduction

Glioblastoma (GBM) constitutes the most frequent malignant primary brain tumor in adults and accounts for nearly 15% of all intracranial neoplasms. Its incidence increases with age and is slightly higher in males than in females.1 Despite continuous advancements in diagnostic and therapeutic techniques, the prognosis for GBM remains extremely poor, with a median survival of less than two years (approximately 15–16 months) and a five-year survival rate below 10%.2 Furthermore, the recurrence rate is close to 100%, and median survival after recurrence is typically less than one year.3 Approximately 90% of GBMs are immunogenic cell death (IDH)-wildtype, which are more aggressive and associated with worse outcomes, whereas patients with IDH-mutant GBM generally exhibit longer survival.4–6 Despite the use of extensive surgical resection, radiotherapy combined with temozolomide (TMZ), and adjuvant TMZ treatment, the current standard therapy offers only temporary disease control, and multiple limitations render treatment outcomes unpredictable.

In recent years, breakthroughs in innovative immunotherapies have led to significant advances in cancer treatment. These strategies aim to modulate the immune system to target tumors, overcoming tumor-induced immune resistance and achieving durable clinical benefits.7 Chimeric antigen receptor T-cell (CAR-T) therapy, as a major branch of immunotherapy, is based on genetically engineering a patient’s own T cells to express a chimeric antigen receptor (CAR) that enables specific recognition and targeting of tumor cell surface antigens. This technology has produced revolutionary breakthroughs in hematologic malignancies, particularly in B-cell cancers such as acute lymphoblastic leukemia and lymphoma, where it has demonstrated remarkable efficacy, with a subset of patients achieving sustained remission or even being effectively cured.8,9 CAR-T therapies directed against cluster of differentiation 19 (CD19) have received Food and Drug Administration approval, with early clinical trials reporting complete remission rates as high as 90%, highlighting their significant clinical potential.10 Compared to hematologic malignancies, however, the application of CAR-T therapy in solid tumors faces unique challenges. The immunosuppressive tumor microenvironment (TME) can lead to inadequate T-cell infiltration and functional exhaustion. Additionally, tumor heterogeneity often results in antigen escape when targeting a single antigen, while the scarcity of tumor-specific antigens in solid tumors further limits therapeutic efficacy.11–14

Although recent innovations, such as combination therapies and engineered modifications, have partially improved T-cell function, and an increasing number of clinical trials have evaluated the efficacy of CAR-T therapy in GBM (Table 1),15–23 CAR-T treatment for GBM still falls far short of the breakthroughs achieved in hematologic malignancies.24,25

Table 1

Summary of clinical trials about chimeric antigen receptor T cell (CAR-T) cells in glioblastom

Target antigenTrialPhaseNo. of PatientsTherapeutic effectAdverse reactionsReasons for failure
Epidermal growth factor receptor vII (EGFRvIII)NCT02209376I10Multiple cases: transient stable disease (SD); 1 case: significant long-term survival15Symptoms like influenza, no severe cytokine release syndrome (CRS) or off-target toxicity16Antigen loss, lack of pretreatment; insufficient amplification and persistence of CAR-T17
Interleukin13Rα2 (IL13Rα2)NCT02208362I6550% patients: SD or partial remission; overall survival (OS): 7.7 months overall18Headaches, Fatigue, Myalgia, Encephalopathy, Ataxia, Mild cerebral edema19Limited treatment scope; antigen heterogeneity and immune escape
IL-13Rα2NCT00730613I31 case: partial remission20Not mentionedExpression differences of IL 13Rα2; microenvironment inhibition
EphA2NCT03423992I31 case: SD; 2 cases: progressive disease (PD); OS:86–181 days21Cytokine release syndrome accompanied by pulmonary edema21The persistence of CAR-T is insufficient21
Human epidermal growth factor receptor 2 (HER2)NCT01109095I171 case: partial remission; 7 cases SD. OS: 24.5 months22Epileptic seizures, Headaches22Antigen heterogeneity; insufficient amplification of CAR-T22
Disialoganglioside 2 (GD2)NCT03170141I84 cases: PD; 3 cases: PD; OS: 10 months23Epileptic seizures, Headaches23Antigen loss, immunosuppressive microenvironment

This review aims to systematically examine the major obstacles limiting the efficacy of CAR-T cell therapy in GBM, with a particular focus on the tumor’s unique microenvironmental features. We integrate the two core challenges, antigen heterogeneity and the immunosuppressive tumor barrier, within a triadic evaluative framework of “challenge identification”, “mechanistic elucidation”, and “strategy matching”. Unlike previous reviews that primarily list isolated strategies, our manuscript emphasizes the alignment between innovative technological approaches and specific pathological mechanisms. We cover topics ranging from logic-gated and switchable modular CAR designs to the application of organoid and single-cell technologies in CAR-T development and evaluation. By bridging fundamental mechanistic insights with translational assessment, this review highlights the evolving therapeutic paradigm in GBM CAR-T therapy—from “single-target killing” toward “multidimensional modulation of the immune ecosystem”. Our goal is to provide a comprehensive framework that offers both mechanistic depth and practical guidance to advance preclinical research and the clinical translation of CAR-T therapies in GBM.

This review is based on a comprehensive synthesis of current literature related to CAR-T cell therapy in GBM. Relevant studies were identified through searches of major scientific databases including PubMed, Web of Science, and Google Scholar. We focused primarily on peer-reviewed original research articles and high-quality review papers published in reputable journals over approximately the last 10 years to capture recent advances. Studies were selected based on their relevance to key topics such as antigenic heterogeneity, TME immunosuppression, clinical challenges, and emerging therapeutic strategies. Both preclinical and clinical studies providing mechanistic insights or reporting therapeutic outcomes were included to ensure a balanced and thorough overview. Studies lacking sufficient experimental detail or clinical relevance were generally excluded.

Dual core challenges in CAR-T therapy for GBM

Unlike hematologic cancers, solid tumors, especially GBM, exhibit significant antigenic diversity and a profoundly immunosuppressive TME, both of which hinder the effectiveness of CAR-T therapy.

Antigenic heterogeneity

The spatiotemporal heterogeneity of antigen expression in GBM significantly undermines the efficacy of CAR-T cell therapy. Core challenges include localized loss and dynamic evolution of tumor cell surface antigens, as well as antigen escape by glioma stem cells (GSCs). For example, epidermal growth factor receptor variant III (EGFRvIII) is a deletion mutant of EGFR that is specifically expressed on tumor cells but absent in normal tissues, making it a tumor-specific antigen.26–28 Early studies report that approximately 30–50% of GBM cases harbor EGFRvIII mutations,29 making it one of the most frequently observed tumor-specific antigens in GBM. However, EGFRvIII is typically expressed only in a subset of tumor subclones, and its expression can be dynamically lost under therapeutic pressures such as radiotherapy.30,31 The phenomenon of antigen loss is particularly prominent in CAR-T therapies targeting EGFRvIII, where residual antigen-negative subclones can undergo clonal expansion, resulting in immune escape.

GSCs, as key drivers of tumor recurrence, exhibit antigenic profiles that differ significantly from those of differentiated tumor cells. GSCs display distinct antigenic profiles compared to differentiated GBM cells, including higher expression of stemness markers such as CD133, aldehyde dehydrogenase 1, and transient receptor potential melastatin 7.32 GSCs can be further divided into molecular subtypes. mesenchymal-type GSCs express serglycin (SRGN), supporting proliferation and stemness through the SRGN–NFκB pathway, while classical subtype/proneural subtype (CL/PN)-type GSCs express mesenchyme homeobox 2 (MEOX2) and depend on the MEOX2–NOTCH axis. Both SRGN and MEOX2 have been implicated in resistance to macrophage-mediated phagocytosis.33 FOS like 1 is a key transcription factor in mesenchymal-type GBM. It promotes GSC stemness, self-renewal, and invasiveness, and enhances the expression of immune-evasive molecules such as CD44 and tenascin C through chromatin remodeling.34 In addition, GSCs can suppress antigen presentation through epigenetic mechanisms (e.g., DNA methylation and histone modification), further reducing CAR-T cell efficacy.35 Therapeutic stress, such as radiation, may also reshape GSC antigen profiles, leading to loss of targets like EGFRvIII and upregulation of stemness markers such as CD133.36–38

Immunosuppressive barriers of the TME

The immunosuppressive barriers in GBM are first manifested through unique physical obstacles. Both the blood–brain barrier (BBB) and the blood–tumor barrier (BTB) collectively limit the infiltration of immune cells and therapeutic agents. The BBB, composed of tightly connected endothelial cells and basement membrane structures, prevents CAR-T cells from entering the brain parenchyma. Studies show that only a small proportion of CAR-T cells can penetrate the BBB and reach the tumor core, thereby limiting therapeutic efficacy.39,40 The BTB, a pathological barrier unique to GBM, is formed by tumor-associated endothelial cells, basement membrane components, pericytes, and other elements of the TME. While structurally similar to the BBB, the BTB possesses tumor-specific molecular characteristics that further reduce drug permeability.41,42 Moreover, the BTB, together with immunosuppressive cells such as tumor-associated macrophages (TAMs), impairs CAR-T cell activity. For example, M2-polarized TAMs can suppress CAR-T cell function through the secretion of anti-inflammatory factors.43,44 Although the BBB may be disrupted within the tumor core in GBM, the BTB at the invasive tumor margins often remains relatively intact, continuing to restrict the delivery of therapeutic agents.42

The complex immunosuppressive network within the GBM TME forms a biological barrier that inhibits CAR-T cell function through intercellular interactions and molecular signaling pathways. For example, glioblastoma-associated macrophages/microglia (GAMs) secrete immunosuppressive cytokines such as interleukin (IL)-10 and TGF-β, directly suppressing T-cell activation and proliferation.44–46 Additionally, M2-polarized GAMs promote the accumulation of programmed cell death protein 1 (PD-L1+) M2-GAMs via activation of the ALOX5 gene and its metabolite 5-HETE, leading to high PD-L1 expression. This binds to programmed death-ligand 1 (PD-1) on T cells, inducing T-cell exhaustion.47 Regulatory T cells (Tregs) are significantly expanded in GBM and further suppress antitumor immune responses through cytotoxic T-lymphocyte antigen 4 (CTLA-4) and adenosine signaling pathways.48–52 Metabolic byproducts such as lactate and adenosine contribute to the immunosuppressive TME in GBM by modulating TAM polarization, Treg recruitment, and CAR-T cell dysfunction (Fig. 1).53–58 As summarized in previous studies, these effects involve key mediators such as hypoxia-inducible factor 1-alpha, vascular endothelial growth factor (VEGF), CD39/CD73, and C-C chemokine receptor type 8.

Metabolic regulation of the immunosuppressive TME in GBM.
Fig. 1  Metabolic regulation of the immunosuppressive TME in GBM.

↑, accumulation of LA and Ado/upregulation expression of Arg1, CD39, CD73 and CCR8; ↓, diminished antigen-presenting capacity. A2A/A2B, adenosine A2A/A2B; Arg1, arginase 1; CCR8, C-C motif chemokine receptor 8; CD, cluster of differentiation; GAM, glioblastoma-associated macrophage; GBM, glioblastoma; HIF-1α, hypoxia-inducible factor 1-alpha; LA, lactate; TME, tumor microenvironment.

Indirect obstacles to CAR-T therapy arising from dual challenges

The antigenic heterogeneity and immunosuppressive TME in GBM not only directly reduce the efficacy of CAR-T therapy but also indirectly create clinical challenges, such as increasing the risk of off-target toxicity and hindering the identification of reliable biomarkers to effectively monitor treatment responses.

Off-target toxicity of CAR-T therapy in GBM

GBM exhibits high antigenic heterogeneity, making single-target CAR-T cells prone to missing tumor subpopulations that do not express the targeted antigen. This promotes “antigen escape” and reduces therapeutic efficacy. To cover a broader range of tumor cells, researchers often select antigens that are widely expressed but not entirely specific, which may lead to off-target toxicity by damaging normal tissues expressing similar antigens. For instance, IL13Rα2, a potential GBM target, is expressed in more than 75% of cases but may also be present at low levels in normal brain tissues such as neurons and microglia, significantly increasing off-target risk.59 Similarly, EGFRvIII-targeted CAR-T cell development requires systematic modeling to assess potential cross-reactivity with wild-type EGFR to ensure safety.60 Insufficient tumor specificity of target antigens, combined with intratumoral heterogeneity, complicates clinical safety by causing unexpected damage to healthy cells during CAR-T therapy. To address this, multi-omics approaches such as single-cell sequencing and ex vivo cross-reactivity assays are essential for screening highly specific targets.60,61

Meanwhile, the TME can amplify off-target toxicity through multiple mechanisms. For example, hypoxia and metabolic stress within the TME impair CAR-T cell persistence, promoting the release of inflammatory cytokines such as IL-6 and interferon-γ (IFN-γ), which may trigger cytokine release syndrome (CRS). The high expression of immunosuppressive markers such as PD-1 and VEGF in the TME may necessitate combining CAR-T therapy with immune checkpoint inhibitors like anti-PD-1 antibodies, although nonspecific T-cell activation might broaden off-target effects.60

Lack of biomarkers for predicting CAR-T therapy response in GBM

The biological complexity of GBM severely limits the ability to predict CAR-T therapy efficacy. As the most aggressive malignant tumor of the central nervous system, GBM is characterized by a highly heterogeneous TME, making it difficult for single-target CAR-T therapies to address all tumor subpopulations and resulting in a lack of effective biomarkers reflecting therapeutic response.

Although targets such as IL-13Rα2 and EGFRvIII have been extensively studied, antigen expression heterogeneity and insufficient tumor specificity continue to hinder the identification of stable and reliable biomarkers. Moreover, real-time monitoring methods remain technically lacking.62,63

The presence of the BBB and BTB further limits the evaluation of systemically administered CAR-T cells.64 While local injections can improve infiltration efficiency, they lack standardized assessment metrics.65–67

Even when appropriate biomarkers are identified, clinical translation faces significant multidimensional technical challenges. Patient stratification difficulties have prevented many clinical trials from achieving precise enrollment. While combination strategies (e.g., co-administration of PD-1 inhibitors) can reverse T-cell exhaustion, they may interfere with biomarker signals associated with monotherapy.68,69 Furthermore, the diversity of resistance mechanisms makes it difficult for a single biomarker to comprehensively predict treatment failure.70–72

Innovative strategies

Antigenic diversity and the highly suppressive TME severely restrict the efficacy of CAR-T cell therapy in solid tumors, particularly GBM. To overcome these obstacles and enhance the antitumor activity of CAR-T cells, various innovative strategies have been explored. These include multi-target synergistic recognition technologies, logic-gated chimeric antigen receptors, switchable CAR systems, broad-spectrum antigen recognition using natural killer group 2, member D (NKG2D)-based CAR-T cells, combined targeting of tumor vasculature-associated antigens, integration of immune checkpoint inhibitors or cytokine engineering, optimization of delivery routes, and the combination of CAR-T therapy with radiotherapy or chemotherapy (Table 2).

Table 2

Innovative strategies to overcome challenges in chimeric antigen receptor T cell (CAR-T) therapy for glioblastoma

ChallengeInnovative strategyExperimental stageSignificance
Antigenic heterogeneityMulti-target cooperative recognition technologyPhase I/IITarget multiple antigens to reduce immune escape
logic-gated chimeric antigen receptorPhase IPrecisely identify and attack cells; reduce the damage to normal tissues
Switchable chimeric antigen receptor (CAR) systemsPhase ISwitching targeted antigens to reduce immune escape
Natural killer group 2 member D (NKG2D) CAR-TPhase ITarget multiple antigens to reduce immune escape; strengthen the immune killing system
Combination of targeting tumor vascular related antigenPreclinical trialNon-specific recognition of antigens; bystander killing mechanism
Immunosuppressive tumor microenvironmentCombination of immune checkpoint inhibitorsPhase IReduce CAR-T cell depletion
Cytokine engineering of CAR-TPreclinical trialExtend CAR-T cell viability and enhance its expansion capacity; Promote immune response
Combination of temozolomide (TMZ) chemotherapyPhase IIncrease tumor stress antigen; activate the immune response; remove immunosuppressive cells
Combination of local radiotherapyPhase IRelease damage-associated molecular patterns (DAMPs) to provide costimulatory signals for CAR-T; regulate cytokines and reduce CAR-T cell depletion; increase tumor antigen expression
Physical barriersChange of the route of administrationPhase IAvoid blood–brain barrie/blood–tumor barrier (BBB/BTB) barriers; reduce overall side effects

Multi-target synergistic recognition technologies

Multi-target synergistic recognition technologies, through tandem or parallel CAR designs, have significantly improved the ability of CAR-T therapies to address GBM heterogeneity and antigen escape. Their core advantages lie in dual-target synergistic cytotoxicity and enhanced specificity through logic-gated designs.

Tandem CARs achieve simultaneous recognition of two tumor-associated antigens within a single-chain structure. For example, a tandem CAR targeting CD44 and CD133 can effectively cover different tumor cell subpopulations within GBM, reducing treatment failure caused by the loss of a single antigen and lowering off-target toxicity. Preclinical studies have demonstrated that CD44/CD133 dual-target IL-7 receptor α-chain-armored CAR-T cells, co-expressing the IL-7 receptor α-chain, enhance T cell survival and proliferation, significantly inhibiting tumor growth and prolonging survival in xenograft models.73 Similarly, EGFRvIII/IL-13Rα2 tandem CAR-T cells have shown synergistic cytotoxic effects in heterogeneous GBM models, outperforming single-target CAR-T therapies and overcoming the spatial heterogeneity of antigen expression.74 Additionally, chlorotoxin-CARs, designed using a chlorotoxin peptide segment that specifically binds matrix metalloproteinase-2, further improve durability and antitumor efficacy when combined in tandem constructs with EGFRvIII or CD276 targets in xenograft models.75

Logic-gated CARs are advanced CAR-T technologies designed using synthetic biology principles. By introducing control mechanisms similar to computer “logic gates”, they regulate T-cell activity through specific antigen input combinations, enabling CAR-T cells to recognize and attack target cells with greater precision while minimizing off-target effects on normal tissues.76

The multi-antigen recognition strategies described above generally reflect the OR logic principle within logic-gated systems. Beyond OR gates, additional designs include AND, NOT, IF-THEN, and IF-BETTER logics (Fig. 2). The AND-gate CAR system separates activation and co-stimulatory signals into two distinct receptors, each recognizing a different antigen, and only activates the CAR-T cells when both (or more) antigens are simultaneously detected. In contrast, the NOT-gate CAR system inhibits activation if a specific antigen is recognized. Studies have shown that CAR-T cells combining an inhibitory domain and human epidermal growth factor receptor 2 (HER2) targeting successfully resisted lung cancer cells,77 illustrating the effectiveness of combining AND and NOT logic principles, with potential applicability to solid tumors. In GBM models, IF-THEN logic has shown research significance. For instance, synNotch CAR systems recognizing EGFRvIII first induce downstream CAR expression (targeting antigens such as IL13Rα2 or CD276) in tumor cells expressing EphA2 or IL13Rα2, thereby reducing T-cell exhaustion and extending T-cell persistence.78 As for IF-BETTER logic, the idea is that the presence of one antigen enhances CAR-T sensitivity toward another antigen, potentially offering unique advantages for tumors like GBM, where highly tumor-specific antigens are rare.

Schematic diagram of logic-gated CARs.
Fig. 2  Schematic diagram of logic-gated CARs.

(a) OR Logic: CAR-T cells engineered with tandem antibodies Ab1 and Ab2 can recognize tumor cells expressing either Ag1, Ag2, or both Ag1 and Ag2. (b) AND Logic: CAR-T cells are only activated by tumor cells co-expressing both Ag1 and Ag2; cells expressing only Ag1 or Ag2 individually do not activate CAR-T cells. (c) NOT Logic: CAR-T cells are not activated when they encounter inhibitory antigens, thereby protecting normal cells that express such inhibitory antigens. (d) IF-THEN Logic: When the antibody Ab2 on the CAR-T surface recognizes Ag2 on tumor cells, it induces the transient expression of Ab1, enabling CAR-T cells to subsequently recognize and kill tumor cells expressing Ag1. (e) IF-BETTER Logic: CAR-T cells can recognize and kill tumor cells expressing Ag1 via Ab1; however, if tumor cells also express Ag2, the sensitivity of CAR-T cells toward Ag1-positive tumor cells is further enhanced. The figure was created using BioRender. Ab1/2; antibody 1/2; Ag1, antigen 1/2; CAR, chimeric antigen receptor; CAR-T, chimeric antigen receptor T cell.

Beyond conventional logic-gated CARs, efforts have also focused on engineering intracellular signaling molecules to create reversible and dynamically regulated signaling networks. For example, the LINK CAR platform leverages the synergy between linker for activation of T cells and SH2 domain-containing leukocyte protein of 76 kDa to construct a fast-responding AND-gated system, enhancing antitumor activity while minimizing off-target risks.79

Collectively, these advances demonstrate that logic-gated CARs hold significant promise for overcoming challenges of antigen heterogeneity and off-target toxicity in GBM therapy. Future directions may focus on improving signal integration efficiency and reducing system complexity to facilitate their clinical translation into solid tumors.

Switchable CAR systems to address dynamic antigen changes

Dynamic changes in antigen expression represent a fundamental limitation to the sustained effectiveness of CAR-T cell therapy in GBM. Tumor cells can escape single-antigen targeting through immune editing or phenotypic plasticity. To address this challenge, researchers have proposed the concept of switchable CAR systems, which dynamically adjust antigen recognition modules or target combinations to enhance tumor coverage.

In traditional CAR designs, the antigen recognition domain and intracellular signaling domains are fused into a single unit, limiting each CAR-T cell to recognizing only a specific target. In contrast, switchable CAR systems use a modular design that separates the antigen recognition domain from the CAR’s signaling domain, allowing flexible redirection without needing to re-engineer the CAR-T cells themselves (Fig. 3). This approach enables the development of universal CARs (UniCARs).80 UniCAR-T cells achieve flexible targeting through soluble Targeting Modules that act as adaptors to direct T cell activity against different tumor antigens. In preclinical studies, UniCAR-T cells were effectively activated by biotinylated trastuzumab in HER2-positive solid tumor models, enabling penetration of the extracellular matrix and killing of hidden tumor cells in 3D spheroid models.81 A similar strategy may be applied to target overexpressed antigens such as EGFR in GBM.82 Regarding safety, CD123-targeted UniCAR-T cells have already demonstrated controllability in a phase I clinical trial (NCT04230265), laying an important safety foundation for potential application in GBM.83,84

Schematic diagram of UniCAR-T structure and mechanism.
Fig. 3  Schematic diagram of UniCAR-T structure and mechanism.

The UniCAR system divides the antigen recognition domain into two components: the Switch module and the Signaling module. The Switch module specifically recognizes tumor cell surface antigens and can rapidly switch between different antigen-binding domains. The Signaling module contains a domain for recognizing the Switch module, a hinge region, a transmembrane domain, and an intracellular signaling domain, which collectively provide activation signals to T cells. The figure was created using BioRender. TAA, tumor-associated antigen; UniCAR, universal chimeric antigen receptor; UniCAR-T, universal chimeric antigen receptor T-cell.

By shifting from “static targeting” to “dynamic adaptation” through modular design, UniCAR technology offers a novel approach to overcoming GBM’s heterogeneity, aggressiveness, and immunosuppressive microenvironment. Its synergistic potential with existing therapies and its favorable safety profile make it a highly promising strategy in the GBM treatment field, although further research is needed to optimize target selection, fine-tune regulatory mechanisms, and develop effective combination therapies.

Broad-spectrum antigen recognition by NKG2D CAR-T cells

As a key activating receptor within the innate immune system, NKG2D is highly conserved and recognizes numerous ligands upregulated under cellular stress, including MHC class I polypeptide-related sequence A (MICA), MHC class I chain-related protein B (MICB), and UL16-binding protein.85,86 NKG2D-based CAR-T cells, by engaging multiple tumor-associated antigens simultaneously, help mitigate antigen escape and hold considerable therapeutic potential for solid tumors like GBM.

Preclinical studies have demonstrated that NKG2D CAR-T cells can eliminate over 90% of tumor cells in glioblastoma organoid (GBO) models derived from patients with other solid tumors,87 with no significant toxicity observed toward normal neural stem cells.88 Further research has revealed that NKG2D ligands such as MICA/B and UL16-binding proteins are expressed in the majority of GBM cases, and NKG2D CAR-T cells have recently been shown to effectively treat GBM.89 Although the expression level of NKG2D ligands in GBM is not as high as in some other cancers, and in certain cases GBM tumor cells may even downregulate these antigens to evade immune surveillance,90 studies have shown that small molecules such as valproic acid can upregulate NKG2D ligand expression, thereby enhancing NK cell-mediated cytotoxicity against tumor cells.91 Beyond their broad-spectrum antigen recognition capabilities, NKG2D CAR-T cells, derived from innate immune receptors, can also synergistically activate immune responses by mimicking NK cell cytotoxic mechanisms. This includes not only direct activation of T cell-mediated cytotoxicity but also recruitment of NK cells to reshape the tumor immune microenvironment and enhance antitumor immune responses.92,93

Synergistic effects of targeting tumor vasculature-associated antigens

The aberrant vascular network in GBM serves not only as a nutrient supply for tumor growth but also as a critical component of its immunosuppressive microenvironment. Designing CAR-T cells to target tumor vasculature-associated antigens enhances antitumor efficacy by exploiting the broader, nonspecific distribution of these antigens. In this regard, several research teams have explored fibroblast activation protein (FAP) as a promising target. FAP-targeted CAR-T therapy enhances treatment efficacy through the following dual mechanisms:

  • Anti-angiogenesis and direct cytotoxicity: FAP is specifically overexpressed in perivascular cells and cancer-associated fibroblasts within GBM. FAP-specific CAR-T cells can disrupt tumor vasculature, inhibit neovascularization, and directly kill FAP-positive tumor cells.94,95

  • Endogenous bystander effect: Novel FAP-targeted CAR-T cells can activate innate immune mechanisms within the TME, such as macrophage phagocytosis and cytokine release, indirectly eliminating neighboring antigen-negative tumor cells. This broadens the antitumor response beyond FAP-positive cells.96

Combination with immune checkpoint inhibitors

The highly immunosuppressive TME in GBM, characterized by elevated PD-L1 expression, induces CAR-T cell exhaustion through the PD-1/PD-L1 signaling pathway.97,98 To address this limitation, combining CAR-T cell therapy with immune checkpoint inhibitors has been explored. Preclinical evidence suggests that inhibiting the PD-1/PD-L1 axis can restore CAR-T cell function and improve antitumor efficacy. For example, in PD-L1–high GBM models, EGFRvIII-targeted CAR-T cells with PD-1 gene knockout demonstrated enhanced cytokine secretion (IFN-γ, IL-2), increased cytolytic activity, and significantly prolonged survival in tumor-bearing mice.68,99 Similarly, combining disialoganglioside 2 (GD2)-targeted CAR-T cells with nivolumab (an anti-PD-1 monoclonal antibody) improved CAR-T persistence, tumor infiltration, and tumor control in animal models.100 In addition, the combination of IL-13Rα2-targeted CAR-T cells with anti-CTLA-4 minibodies further supports the synergistic potential of multi-target immunotherapy.101,102

Despite promising preclinical data, early-phase clinical trials (e.g., NCT03726515) reported that EGFRvIII-targeted CAR-T cells combined with pembrolizumab were safe and biologically active but showed limited overall survival benefit due to lack of efficacy.103 Notably, PD-1 expression levels in infused CAR-T products were significantly correlated with peripheral CAR-T cell expansion and progression-free survival, suggesting that PD-1 could serve as a predictive biomarker for treatment response.104 To optimize combination strategies, researchers are focusing on genetically engineering CAR-T cells. Approaches such as clustered regularly interspaced short palindromic repeats/Cas9-mediated PD-1 knockout or overexpression of costimulatory molecules (e.g., 4-1BB) have been shown to improve CAR-T resistance to PD-L1–mediated suppression.62,105 For instance, CAR-T cells expressing checkpoint reversal receptors targeting PD-1 have demonstrated enhanced tumor clearance in GBM models.105

While the combination of immune checkpoint inhibitors with CAR-T therapy offers strong theoretical advantages for GBM treatment, further studies are needed to elucidate precise mechanisms and optimize therapeutic protocols for clinical translation.

Cytokine engineering enhances CAR-T cell therapy

While certain cytokines in the TME contribute to immunosuppression and hinder CAR-T cell efficacy, others—such as IL-7, IL-15, and IL-21—support T-cell activity by stimulating the JAK-STAT signaling pathway, thereby promoting CAR-T cell survival and proliferation.106–108 This has led to the emergence of cytokine-engineered CAR-T cell strategies. These approaches involve incorporating cytokines or their receptors into CAR-T cells to significantly enhance their proliferation, persistence, and antitumor activity while simultaneously remodeling the immune microenvironment to support synergistic immune responses.

In cytokine-engineered CAR-T strategies, IL-7 is of particular interest for its role in maintaining T-cell homeostasis and promoting survival. In an EGFRvIII-heterogeneous GBM model, IL-7–modified CAR-T cells showed enhanced expansion and increased mouse survival from 9% to 67%, largely due to IL-7–mediated self-renewal and reduced exhaustion.109 Moreover, CAR-T cells co-engineered with IL-7 and Flt3L further amplified these effects by increasing conventional dendritic cells and CD103+XCR1+ migratory dendritic cells in the TME, thereby enhancing antigen cross-presentation and endogenous immune activation to eliminate antigen-negative tumor cells.109

Beyond IL-7 and Flt3L, additional studies have developed CAR-T cells that co-express cytokines upon activation. For instance, GD2-targeted IL-18–secreting CAR-T cells release IL-18 in a CAR-dependent manner.110 Others have engineered autocrine cytokine-receptor circuits, such as IL-7/C-C motif chemokine ligand 21 (CCL21) combinations, to further improve CAR-T efficacy.111

However, challenges remain regarding safety, tumor heterogeneity, and clinical translation. Proliferative cytokines like IL-6 may increase the risk of CRS, highlighting the need for optimized regulatory systems.112 Tumor heterogeneity necessitates the use of multi-targeted CAR-T cells or combination therapies with immune checkpoint inhibitors.

Cytokine engineering offers a paradigm shift for CAR-T therapy—from “single-target cytotoxicity” to “systemic immunomodulation”. By synergistically enhancing CAR-T function and reprogramming the immune microenvironment, this approach holds promise for overcoming the limitations of solid tumor treatment. Future research should explore combinations of cytokines such as IL-15 and IL-21, dynamic regulatory strategies, and integration with localized delivery platforms or biomaterials to further enhance therapeutic outcomes.

Immunomodulatory role of TMZ chemotherapy combined with CAR-T cell therapy

Despite aggressive treatment involving surgical resection followed by radiotherapy and TMZ chemotherapy, patients with GBM face a median overall survival of just 12 to 15 months and a five-year survival rate under 10%.2,113,114 TMZ exerts its cytotoxic effect by inducing DNA damage via alkylation, yet its effectiveness is constrained by poor BBB permeability, tumor heterogeneity, and an immunosuppressive TME.115,116 However, combining TMZ with CAR-T cell therapy has demonstrated unique synergistic mechanisms. Firstly, TMZ activates the DNA damage response pathway, leading to upregulation of stress-induced antigens on tumor cells, which enhances CAR-T cell recognition and cytotoxicity.117 In parallel, TMZ-induced tumor cell apoptosis results in antigen release, thereby activating endogenous antitumor immune responses.118,119 Secondly, TMZ’s lymphodepleting effect eliminates immunosuppressive cells such as Tregs, creating a favorable niche for CAR-T cell expansion.120,121 To address the cytotoxic effects of TMZ on normal T cells, genetic engineering strategies have been explored to improve CAR-T cell viability. For instance, co-expression of miR-17-92 suppresses pro-apoptotic proteins, preserving T-cell activity and IFN-γ production.122 Moreover, MGMT gene modification renders γδ CAR-T cells resistant to TMZ-induced DNA damage.117

The integration of TMZ and CAR-T cell therapy represents an innovative strategy for GBM, as TMZ increases tumor-antigen exposure, remodels the immune microenvironment, and improves T-cell survival. The clinical translation of this combination approach will benefit from multicenter trials and the integration of personalized treatment strategies.2,115

Combination of local radiotherapy and CAR-T cell therapy

As a commonly used therapeutic approach for GBM, radiotherapy not only exerts direct cytotoxic effects on tumor cells but also modulates the TME through immunoregulatory mechanisms. These include the induction of immunogenic cell death (ICD), stimulation of innate immune responses, and enhancement of T-cell infiltration, thus offering a solid basis for combination with CAR-T cell therapy.123,124

Radiotherapy can enhance CAR-T efficacy through multiple mechanisms. Firstly, First, it induces tumor cell stress leading to ICD. Dying tumor cells release various damage-associated molecular patterns (DAMPs) that activate DAMP signaling, triggering phagocytosis and the production of inflammatory cytokines. These processes promote antigen presentation by dendritic cells, thereby enhancing the killing capacity of CAR-T cells and reversing their exhaustion.125 Secondly, irradiated tumor cells release abundant radiotherapy-induced microparticles. Upon uptake by non-irradiated cells, these radiotherapy-induced microparticles significantly upregulate surface MHC-I expression, enhancing T-cell recognition of tumor-associated antigens.126 Thirdly, radiotherapy has dose-dependent effects on immune checkpoint molecule expression (e.g., PD-L1, CTLA-4). Systematic investigation of this dose-dependent regulation could help design optimized radiotherapy protocols that synergistically enhance CAR-T-mediated tumor eradication.127 Furthermore, emerging studies have revealed that radiotherapy can induce systemic abscopal effects in cancer treatment—beyond eliminating localized lesions, it can cause regression of distant tumors. While the precise mechanisms remain incompletely elucidated, this phenomenon likely involves radiotherapy-induced ICD and the subsequent release of DAMPs.128 However, such abscopal responses have been observed in only a minority of clinical cases.

Compared to traditional chemotherapy, localized radiotherapy offers superior spatial precision, significantly reducing post-treatment systemic immunosuppression. This preserves CAR-T cell functionality and minimizes interference with concurrent immunotherapeutic regimens. Several ongoing clinical trials are investigating this combinatorial strategy. For instance, emerging studies reveal that FLASH radiotherapy (ultra–high dose rate radiotherapy) enhances GD2 CAR-T cell efficacy against medulloblastoma through dual metabolic-immune reprogramming. It remodels lipid metabolism in TAMs, driving their polarization toward the pro-inflammatory M1 phenotype while suppressing the immunosuppressive M2 phenotype.129 Future experimental investigations of this combination strategy must address several challenges. First, optimal radiation dose and timing need to be determined: low-dose irradiation synergizes well with CAR-T therapy, whereas high doses may induce fibrosis and hinder CAR-T infiltration.123 Second, radiotherapy can impose selective pressure on tumors, potentially leading to the expansion of antigen-negative clones. To address this, multi-targeted CAR-T cells or combined tumor vaccines may be necessary to overcome antigen heterogeneity.130,131

In conclusion, combining local radiotherapy with CAR-T cell therapy presents a promising strategy for treating GBM. Further optimization and approaches to address the immunosuppressive TME are crucial for successful clinical translation.

Delivery routes of CAR-T cell therapy for gliomas

The therapeutic efficacy of CAR-T cell therapy in gliomas is highly dependent on optimizing delivery routes. Traditional systemic intravenous administration, though convenient, is significantly limited by the BBB, which hampers CAR-T cell infiltration into the tumor and results in poor delivery efficiency.132,133 Moreover, systemic administration may induce off-target toxicity and CRS.132,134

Local and regional delivery strategies, which involve direct administration of CAR-T cells into the tumor site or cerebrospinal fluid (CSF), effectively circumvent the BBB. Intratumoral injection, often performed using stereotactic techniques, delivers CAR-T cells targeting antigens such as HER2, EGFR, or B7-H3 directly into the tumor cavity. This approach has demonstrated safety in clinical trials for pediatric gliomas,135 although repeated dosing is often required, and there is a risk of infection. Intraventricular or intrathecal administration is suitable for tumors near the ventricles or meninges. When combined with epigenetic modulators, it can enhance efficacy against metastatic medulloblastoma.136 However, the persistence of CAR-T cells in the CSF and the risk of neurotoxicity remain concerns.

With advances in nanomedicine and biomaterial sciences, emerging strategies have been developed to enhance CAR-T function via nanocarriers and engineered delivery platforms. For example, macrophage-based CAR-MΦ therapies leverage the natural tumor-penetrating properties of macrophages. ErbB2-targeting CAR-MΦs have shown durable antitumor effects in glioma models, with localized administration reducing systemic side effects.137 Similarly, biomimetic nanoparticle-based CAR-neutrophil systems, designed to mimic neutrophil responses to TME-derived inflammatory signals, have enabled targeted CAR-T delivery and synergized with chemotherapeutic agents to elicit enhanced antitumor responses.138

Prospective technological advances related to CAR-T therapy

With the rapid advancement of multiple scientific disciplines, cutting-edge technologies from various fields are being actively integrated into CAR-T research. This section briefly introduces the utilization of single-cell sequencing techniques and organoid systems within CAR-T treatment strategies for GBM.

Single-cell sequencing to guide personalized multi-antigen CAR-T design

The introduction of single-cell RNA sequencing technologies has provided a novel perspective for dissecting the molecular complexity of GBM. Multi-target CAR-T design based on single-cell data is a key strategy to overcome tumor heterogeneity. Single-cell sequencing enables systematic profiling of cancer cell subpopulations within the TME, allowing identification of combinatorial antigen signatures specific to different cell subsets. For instance, single-cell RNA sequencing has revealed distinct antigen expression patterns between GSCs and differentiated tumor cells, with antigens such as GD2, IL13Rα2, and EphA3 being highly expressed in the stem-like populations.73,139,140 Using transcriptomic screening, researchers have identified combinations like CD44 and CD133 that can cover heterogeneous GBM populations, with their synergistic anti-tumor effects validated in organoid models.73,141 Furthermore, integrating single-cell epigenomic data allows prediction of pathways associated with tumor cell plasticity, providing insights for combination therapies aimed at halting resistance evolution.142,143

Single-cell RNA sequencing can also reveal the dynamic trajectories of tumor cell subpopulations, such as the enrichment of specific GSC-like subsets in recurrent GBM and their interactions with the immune microenvironment.142,143 Additionally, single-cell multi-omics analyses can identify CAR-T exhaustion markers like CD38 and durable T-cell subpopulations, offering molecular targets to optimize CAR-T function.144,145

In summary, single-cell sequencing sheds light on the evolution of GBM heterogeneity at the molecular level and offers a data-driven framework for the precise design of multi-antigen CAR-T strategies. By integrating target selection, microenvironmental modulation, and delivery optimization, personalized multi-target CAR-T approaches may overcome the limitations of current GBM therapies. Their clinical translation will require further interdisciplinary innovation and the development of adaptive therapeutic systems.

Organoid models to optimize preclinical evaluation systems

The inter- and intra-tumoral heterogeneity of GBM poses significant challenges to traditional in vitro models. Patient-derived GBOs, which can be rapidly generated within two to four weeks and cryopreserved, successfully retain tumor heterogeneity and molecular characteristics, making them an ideal platform for CAR-T research.146,147 GBOs not only mimic the immunosuppressive features of the TME but also allow real-time evaluation of CAR-T cell infiltration, cytotoxicity, and cytokine release profiles. For example, GBOs preserve GSC subpopulations and vascular structures, enabling assessment of CAR-T–mediated cytotoxicity toward stem-like and perivascular cells.148 Moreover, the similarity in cytokine release patterns between GBOs and patient CSF further validates their clinical relevance.73,149

In CAR-T studies targeting GBM, research teams have used dual-target strategies (e.g., CD44/CD133 tandem CAR-Ts) and broad-spectrum antigen recognition approaches (e.g., 806 CAR-Ts targeting EGFRvIII) to overcome antigen heterogeneity and immune evasion.73,150 Combination therapies, such as CAR-Ts with the IAP antagonist birinapant, enhance efficacy by inducing bystander killing and activating the NF-κB pathway.151 Locally delivered engineered CAR-Ts, such as those secreting IL-7/CCL19 or Tanζ-T28-Δ7R CAR-Ts, have also been validated in GBOs and orthotopic xenograft models.27,73 These advances underscore the pivotal role of GBOs in dynamic efficacy evaluation.

To enhance the predictive value of GBO-based models, it is essential to integrate multi-omics data and functional parameters such as CAR-T persistence and exhaustion markers into efficacy prediction frameworks.152 Innovations such as the development of GBM–blood vessel assembloids and microfluidic chip systems have enabled simulation of BBB penetration and CAR-T migration/killing dynamics under perfusion conditions.27,148 Standardized biobanking of GBOs representing various molecular subtypes and treatment histories is another critical step in model optimization.146,147 However, challenges remain in model standardization, dynamic modeling of resistance evolution, and the full representation of immune microenvironment complexity.73,146–148,151

In summary, GBO-based preclinical systems offer a highly relevant platform by recapitulating tumor heterogeneity and the GBM microenvironment. Future efforts should focus on integrating engineered CAR designs, multi-omics analysis, and dynamic modeling to accelerate the development of personalized immunotherapeutic strategies against GBM.

Limitations

This review has several limitations. First, many of the studies referenced are based on preclinical animal models or early-phase clinical trials, and their reproducibility and applicability in broader patient populations remain to be fully validated. Second, while we focused on summarizing innovative strategies and their therapeutic potential, practical issues such as safety, cost-effectiveness, and clinical feasibility were not discussed in depth. Future research should integrate multi-center clinical trials with multi-omics analyses to further elucidate GBM immune evasion mechanisms and facilitate the efficient clinical translation of CAR-T therapy.

Conclusions

CAR-T cell therapy for GBM faces two fundamental challenges: antigenic heterogeneity and an immunosuppressive TME. Recent research has progressed from single-target strategies to multi-dimensional and combinatorial approaches. Innovative designs, such as multi-target CAR-Ts, dynamic antigen-adaptive systems, and TME-remodeling modifications, have significantly enhanced therapeutic efficacy. For example, logic-gated CAR systems enable precise recognition of tumor regions co-expressing dual antigens; NKG2D-based CAR-T cells provide broad-spectrum targeting to reduce immune escape; and IL-7/Flt3L engineering improves CAR-T persistence while stimulating endogenous immune responses. In addition, combination therapies involving radiotherapy, TMZ chemotherapy, or immune checkpoint inhibitors have shown synergistic effects. Novel delivery methods, including local administration and nanocarrier technologies, also hold promise for overcoming the BBB.

CAR-T therapy for GBM must evolve beyond a tumor-centric “killing” paradigm toward a model of systemic immune regulation. The integration of interdisciplinary innovation with robust clinical trial design will be key to overcoming existing bottlenecks and advancing the next generation of CAR-T therapies for GBM.

Declarations

Acknowledgement

We are grateful for the guidance provided by Professors Tomas Lindahl and Axel Behrens.

Funding

This work was partially supported by the National Natural Science Foundation of China (82072766).

Conflict of interest

The authors declare that they have no conflicts of interest.

Authors’ contributions

Conceptualization (QLJ, YLH, LJZ), writing - original draft preparation (LJZ, ZQZ, JLH), figure preparation (JLH, ZHZ), writing - review & editing (HXZ, YHJ, XZ), writing - revision (YMY, ZHZ), and funding acquisition (NNL, WX). All authors have given informed consent for the publication of this article.

References

  1. Tan AC, Ashley DM, López GY, Malinzak M, Friedman HS, Khasraw M. Management of glioblastoma: State of the art and future directions. CA Cancer J Clin 2020;70(4):299-312 View Article PubMed/NCBI
  2. Zhang P, Zhang Y, Ji N. Challenges in the Treatment of Glioblastoma by Chimeric Antigen Receptor T-Cell Immunotherapy and Possible Solutions. Front Immunol 2022;13:927132 View Article PubMed/NCBI
  3. Bagley SJ, Logun M, Fraietta JA, Wang X, Desai AS, Bagley LJ, et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results. Nat Med 2024;30(5):1320-1329 View Article PubMed/NCBI
  4. Wang S, Li L, Zuo S, Kong L, Wei J, Dong J. Metabolic-related gene pairs signature analysis identifies ABCA1 expression levels on tumor-associated macrophages as a prognostic biomarker in primary IDH(WT) glioblastoma. Front Immunol 2022;13:869061 View Article PubMed/NCBI
  5. Cómitre-Mariano B, Segura-Collar B, Vellila-Alonso G, Contreras R, Henandez-Lain A, Valiente M, et al. S100A proteins show a spatial distribution of inflammation associated with the glioblastoma microenvironment architecture. Theranostics 2025;15(2):726-744 View Article PubMed/NCBI
  6. Pinson H, Silversmit G, Vanhauwaert D, Vanschoenbeek K, Okito JK, De Vleeschouwer S, et al. Epidemiology and survival of adult-type diffuse glioma in Belgium during the molecular era. Neuro Oncol 2024;26(1):191-202 View Article PubMed/NCBI
  7. Morte-Romea E, Pesini C, Pellejero-Sagastizábal G, Letona-Giménez S, Martínez-Lostao L, Aranda SL, et al. CAR Immunotherapy for the treatment of infectious diseases: a systematic review. Front Immunol 2024;15:1289303 View Article PubMed/NCBI
  8. Shimabukuro-Vornhagen A, Böll B, Schellongowski P, Valade S, Metaxa V, Azoulay E, et al. Critical care management of chimeric antigen receptor T-cell therapy recipients. CA Cancer J Clin 2022;72(1):78-93 View Article PubMed/NCBI
  9. Peng JJ, Wang L, Li Z, Ku CL, Ho PC. Metabolic challenges and interventions in CAR T cell therapy. Sci Immunol 2023;8(82):eabq3016 View Article PubMed/NCBI
  10. Shah NN, Fry TJ. Mechanisms of resistance to CAR T cell therapy. Nat Rev Clin Oncol 2019;16(6):372-385 View Article PubMed/NCBI
  11. Dagher OK, Posey AD. Forks in the road for CAR T and CAR NK cell cancer therapies. Nat Immunol 2023;24(12):1994-2007 View Article PubMed/NCBI
  12. Uslu U, Castelli S, June CH. CAR T cell combination therapies to treat cancer. Cancer Cell 2024;42(8):1319-1325 View Article PubMed/NCBI
  13. Young RM, Engel NW, Uslu U, Wellhausen N, June CH. Next-Generation CAR T-cell Therapies. Cancer Discov 2022;12(7):1625-1633 View Article PubMed/NCBI
  14. Albelda SM. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat Rev Clin Oncol 2024;21(1):47-66 View Article PubMed/NCBI
  15. Ju A, Choi S, Jeon Y, Kim K. Lymphodepletion in Chimeric Antigen Receptor T-Cell Therapy for Solid Tumors: A Focus on Brain Tumors. Brain Tumor Res Treat 2024;12(4):208-220 View Article PubMed/NCBI
  16. O’Rourke DM, Nasrallah M, Morrissette JJ, Melenhorst JJ, Lacey SF, Mansfield K, et al. Pilot study of T cells redirected to EGFRvIII with a chimeric antigen receptor in patients with EGFRvIII+ glioblastoma. J Clin Oncol 2016;34(15_Suppl):2067-2067 View Article PubMed/NCBI
  17. Jackson CM, Choi J, Lim M. Mechanisms of immunotherapy resistance: lessons from glioblastoma. Nat Immunol 2019;20(9):1100-1109 View Article PubMed/NCBI
  18. Brown CE, Hibbard JC, Alizadeh D, Blanchard MS, Natri HM, Wang D, et al. 1520 Phase I trial evaluating locoregionally-delivered IL13Rα2-targeting CAR T cells in high-grade glioma. J ImmunoTher Cancer 2023;11(Suppl 2):A1745 View Article PubMed/NCBI
  19. Brown CE, Hibbard JC, Alizadeh D, Blanchard MS, Natri HM, Wang D, et al. Locoregional delivery of IL-13Rα2-targeting CAR-T cells in recurrent high-grade glioma: a phase 1 trial. Nat Med 2024;30(4):1001-1012 View Article PubMed/NCBI
  20. Choi BD, Curry WT, Carter BS, Maus MV. Chimeric antigen receptor T-cell immunotherapy for glioblastoma: practical insights for neurosurgeons. Neurosurg Focus 2018;44(6):E13 View Article PubMed/NCBI
  21. Lin Q, Ba T, Ho J, Chen D, Cheng Y, Wang L, et al. First-in-Human Trial of EphA2-Redirected CAR T-Cells in Patients With Recurrent Glioblastoma: A Preliminary Report of Three Cases at the Starting Dose. Front Oncol 2021;11:694941 View Article PubMed/NCBI
  22. Ahmed N, Brawley V, Hegde M, Bielamowicz K, Kalra M, Landi D, et al. HER2-Specific Chimeric Antigen Receptor-Modified Virus-Specific T Cells for Progressive Glioblastoma: A Phase 1 Dose-Escalation Trial. JAMA Oncol 2017;3(8):1094-1101 View Article PubMed/NCBI
  23. Liu Z, Zhou J, Yang X, Liu Y, Zou C, Lv W, et al. Safety and antitumor activity of GD2-Specific 4SCAR-T cells in patients with glioblastoma. Mol Cancer 2023;22(1):3 View Article PubMed/NCBI
  24. Choi BD, Yu X, Castano AP, Bouffard AA, Schmidts A, Larson RC, et al. CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat Biotechnol 2019;37(9):1049-1058 View Article PubMed/NCBI
  25. Xia Z, Jin Q, Long Z, He Y, Liu F, Sun C, et al. Targeting overexpressed antigens in glioblastoma via CAR T cells with computationally designed high-affinity protein binders. Nat Biomed Eng 2024;8(12):1634-1650 View Article PubMed/NCBI
  26. Abbott RC, Iliopoulos M, Watson KA, Arcucci V, Go M, Hughes-Parry HE, et al. Human EGFRvIII chimeric antigen receptor T cells demonstrate favorable safety profile and curative responses in orthotopic glioblastoma. Clin Transl Immunology 2023;12(3):e1440 View Article PubMed/NCBI
  27. Ohta K, Sakoda Y, Adachi K, Shinozaki T, Nakajima M, Yasuda H, et al. Therapeutic Efficacy of IL7/CCL19-Expressing CAR-T Cells in Intractable Solid Tumor Models of Glioblastoma and Pancreatic Cancer. Cancer Res Commun 2024;4(9):2514-2524 View Article PubMed/NCBI
  28. Choi JM, Lim SH, Liu ZP, Lee TK, Rhee JH, Yoon MS, et al. Flagellin synergistically enhances anti-tumor effect of EGFRvIII peptide in a glioblastoma-bearing mouse brain tumor model. BMC Cancer 2022;22(1):986 View Article PubMed/NCBI
  29. Sterner RC, Sterner RM. EGFRVIII and EGFR targeted chimeric antigen receptor T cell therapy in glioblastoma. Front Oncol 2024;14:1434495 View Article PubMed/NCBI
  30. Zheng L, Luthra R, Alvarez HA, San Lucas FA, Duose DY, Wistuba II, et al. Intragenic EGFR::EGFR.E1E8 Fusion (EGFRvIII) in 4331 Solid Tumors. Cancers (Basel) 2023;16(1):6 View Article PubMed/NCBI
  31. Haryuni RD, Tanaka T, Zhou Y, Yokoyama S, Sakurai H. ERK-mediated negative feedback regulation of oncogenic EGFRvIII in glioblastoma cells. Oncol Lett 2020;20(3):2477-2482 View Article PubMed/NCBI
  32. Guo S, Ramar V, Guo AA, Saafir T, Akpobiyeri H, Hudson B, et al. TRPM7 transactivates the FOSL1 gene through STAT3 and enhances glioma stemness. Cell Mol Life Sci 2023;80(9):270 View Article PubMed/NCBI
  33. Lu C, Kang T, Zhang J, Yang K, Liu Y, Song K, et al. Combined targeting of glioblastoma stem cells of different cellular states disrupts malignant progression. Nat Commun 2025;16(1):2974 View Article PubMed/NCBI
  34. Marques C, Unterkircher T, Kroon P, Oldrini B, Izzo A, Dramaretska Y, et al. NF1 regulates mesenchymal glioblastoma plasticity and aggressiveness through the AP-1 transcription factor FOSL1. Elife 2021;10:e64846 View Article PubMed/NCBI
  35. Bao L, Chen Y, Lai HT, Wu SY, Wang JE, Hatanpaa KJ, et al. Methylation of hypoxia-inducible factor (HIF)-1α by G9a/GLP inhibits HIF-1 transcriptional activity and cell migration. Nucleic Acids Res 2018;46(13):6576-6591 View Article PubMed/NCBI
  36. Ginjala V, Rodriguez-Colon L, Ganguly B, Gangidi P, Gallina P, Al-Hraishawi H, et al. Protein-lysine methyltransferases G9a and GLP1 promote responses to DNA damage. Sci Rep 2017;7(1):16613 View Article PubMed/NCBI
  37. Agosti E, Zeppieri M, Ghidoni M, Ius T, Tel A, Fontanella MM, et al. Role of glioma stem cells in promoting tumor chemo- and radioresistance: A systematic review of potential targeted treatments. World J Stem Cells 2024;16(5):604-614 View Article PubMed/NCBI
  38. Ye Y, Xie W, Wang X, Tan S, Yang L, Ma Z, et al. DNA-damage orchestrates self-renewal and differentiation via reciprocal p53 family and Hippo/Wnt/TGF-β pathway activation in embryonic stem cells. Cell Mol Life Sci 2025;82(1):38 View Article PubMed/NCBI
  39. Gonzales-Aloy E, Ahmed-Cox A, Tsoli M, Ziegler DS, Kavallaris M. From cells to organoids: The evolution of blood-brain barrier technology for modelling drug delivery in brain cancer. Adv Drug Deliv Rev 2023;196:114777 View Article PubMed/NCBI
  40. Akbari P, Katsarou A, Daghighian R, van Mil LWHG, Huijbers EJM, Griffioen AW, et al. Directing CAR T cells towards the tumor vasculature for the treatment of solid tumors. Biochim Biophys Acta Rev Cancer 2022;1877(3):188701 View Article PubMed/NCBI
  41. Jimenez-Macias JL, Vaughn-Beaucaire P, Bharati A, Xu Z, Forrest M, Hong J, et al. Modulation of blood-tumor barrier transcriptional programs improves intratumoral drug delivery and potentiates chemotherapy in GBM. Sci Adv 2025;11(9):eadr1481 View Article PubMed/NCBI
  42. Digiovanni S, Lorenzati M, Bianciotto OT, Godel M, Fontana S, Akman M, et al. Blood-brain barrier permeability increases with the differentiation of glioblastoma cells in vitro. Fluids Barriers CNS 2024;21(1):89 View Article PubMed/NCBI
  43. Zhou Y, Guo Y, Chen L, Zhang X, Wu W, Yang Z, et al. Co-delivery of phagocytosis checkpoint and STING agonist by a Trojan horse nanocapsule for orthotopic glioma immunotherapy. Theranostics 2022;12(12):5488-5503 View Article PubMed/NCBI
  44. Zhu N, Chen S, Jin Y, Wang M, Fang L, Xue L, et al. Enhancing Glioblastoma Immunotherapy with Integrated Chimeric Antigen Receptor T Cells through the Re-Education of Tumor-Associated Microglia and Macrophages. ACS Nano 2024;18(17):11165-11182 View Article PubMed/NCBI
  45. Xuan W, Lesniak MS, James CD, Heimberger AB, Chen P. Context-Dependent Glioblastoma-Macrophage/Microglia Symbiosis and Associated Mechanisms. Trends Immunol 2021;42(4):280-292 View Article PubMed/NCBI
  46. Khan F, Pang L, Dunterman M, Lesniak MS, Heimberger AB, Chen P. Macrophages and microglia in glioblastoma: heterogeneity, plasticity, and therapy. J Clin Invest 2023;133(1):e163446 View Article PubMed/NCBI
  47. Chen T, Liu J, Wang C, Wang Z, Zhou J, Lin J, et al. ALOX5 contributes to glioma progression by promoting 5-HETE-mediated immunosuppressive M2 polarization and PD-L1 expression of glioma-associated microglia/macrophages. J Immunother Cancer 2024;12(8):e009492 View Article PubMed/NCBI
  48. Espinoza FI, Tankov S, Chliate S, Pereira Couto J, Marinari E, Vermeil T, et al. Targeting HIF-2α in glioblastoma reshapes the immune infiltrate and enhances response to immune checkpoint blockade. Cell Mol Life Sci 2025;82(1):119 View Article PubMed/NCBI
  49. Tekguc M, Wing JB, Osaki M, Long J, Sakaguchi S. Treg-expressed CTLA-4 depletes CD80/CD86 by trogocytosis, releasing free PD-L1 on antigen-presenting cells. Proc Natl Acad Sci U S A 2021;118(30):e2023739118 View Article PubMed/NCBI
  50. Alarcón S, Toro MLÁ, Villarreal C, Melo R, Fernández R, Ayuso Sacido A, et al. Decreased Equilibrative Nucleoside Transporter 1 (ENT1) Activity Contributes to the High Extracellular Adenosine Levels in Mesenchymal Glioblastoma Stem-Like Cells. Cells 2020;9(8):1914 View Article PubMed/NCBI
  51. Wurm M, Schaaf O, Reutner K, Ganesan R, Mostböck S, Pelster C, et al. A Novel Antagonistic CD73 Antibody for Inhibition of the Immunosuppressive Adenosine Pathway. Mol Cancer Ther 2021;20(11):2250-2261 View Article PubMed/NCBI
  52. Wang M, Jia J, Cui Y, Peng Y, Jiang Y. CD73-positive extracellular vesicles promote glioblastoma immunosuppression by inhibiting T-cell clonal expansion. Cell Death Dis 2021;12(11):1065 View Article PubMed/NCBI
  53. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014;513(7519):559-563 View Article PubMed/NCBI
  54. Sun T, Liu B, Li Y, Wu J, Cao Y, Yang S, et al. Oxamate enhances the efficacy of CAR-T therapy against glioblastoma via suppressing ectonucleotidases and CCR8 lactylation. J Exp Clin Cancer Res 2023;42(1):253 View Article PubMed/NCBI
  55. Li T, Xu D, Ruan Z, Zhou J, Sun W, Rao B, et al. Metabolism/Immunity Dual-Regulation Thermogels Potentiating Immunotherapy of Glioblastoma Through Lactate-Excretion Inhibition and PD-1/PD-L1 Blockade. Adv Sci (Weinh) 2024;11(18):e2310163 View Article PubMed/NCBI
  56. Bai Y, Zhang X, Zheng J, Liu Z, Yang Z, Zhang X. Overcoming high level adenosine-mediated immunosuppression by DZD2269, a potent and selective A2aR antagonist. J Exp Clin Cancer Res 2022;41(1):302 View Article PubMed/NCBI
  57. Wang L, Zhang J, Zhang W, Zheng M, Guo H, Pan X, et al. The inhibitory effect of adenosine on tumor adaptive immunity and intervention strategies. Acta Pharm Sin B 2024;14(5):1951-1964 View Article PubMed/NCBI
  58. Zhang X, Wang H. Unlocking CAR T cell potential: Inosine-induced stemness and enhanced potency. Cancer Cell 2024;42(2):175-177 View Article PubMed/NCBI
  59. Kim GB, Aragon-Sanabria V, Randolph L, Jiang H, Reynolds JA, Webb BS, et al. High-affinity mutant Interleukin-13 targeted CAR T cells enhance delivery of clickable biodegradable fluorescent nanoparticles to glioblastoma. Bioact Mater 2020;5(3):624-635 View Article PubMed/NCBI
  60. Pawlowski KD, Duffy JT, Gottschalk S, Balyasnikova IV. Cytokine Modification of Adoptive Chimeric Antigen Receptor Immunotherapy for Glioblastoma. Cancers (Basel) 2023;15(24):5852 View Article PubMed/NCBI
  61. Maus MV. Designing CAR T cells for glioblastoma. Oncoimmunology 2015;4(12):e1048956 View Article PubMed/NCBI
  62. Nakazawa T, Natsume A, Nishimura F, Morimoto T, Matsuda R, Nakamura M, et al. Effect of CRISPR/Cas9-Mediated PD-1-Disrupted Primary Human Third-Generation CAR-T Cells Targeting EGFRvIII on In Vitro Human Glioblastoma Cell Growth. Cells 2020;9(4):998 View Article PubMed/NCBI
  63. Starr R, Aguilar B, Gumber D, Maker M, Huard S, Wang D, et al. Inclusion of 4-1BB Costimulation Enhances Selectivity and Functionality of IL13Rα2-Targeted Chimeric Antigen Receptor T Cells. Cancer Res Commun 2023;3(1):66-79 View Article PubMed/NCBI
  64. Brighi C, Salimova E, de Veer M, Puttick S, Egan G. Translation of focused ultrasound for blood-brain barrier opening in glioma. J Control Release 2022;345:443-463 View Article PubMed/NCBI
  65. Rathi S, Griffith JI, Zhang W, Zhang W, Oh JH, Talele S, et al. The influence of the blood-brain barrier in the treatment of brain tumours. J Intern Med 2022;292(1):3-30 View Article PubMed/NCBI
  66. Mulay AR, Hwang J, Kim DH. Microphysiological Blood-Brain Barrier Systems for Disease Modeling and Drug Development. Adv Healthc Mater 2024;13(21):e2303180 View Article PubMed/NCBI
  67. Song G, Plumlee P, Ahn JY, Wong ST, Zhao H. Translational strategies and systems biology insights for blood-brain barrier opening and delivery in brain tumors and Alzheimer’s disease. Biomed Pharmacother 2023;167:115450 View Article PubMed/NCBI
  68. Song Y, Liu Q, Zuo T, Wei G, Jiao S. Combined antitumor effects of anti-EGFR variant III CAR-T cell therapy and PD-1 checkpoint blockade on glioblastoma in mouse model. Cell Immunol 2020;352:104112 View Article PubMed/NCBI
  69. Chen W, Xian N, Zhao N, Zhang Q, Xu Y. PD1CD28 chimeric molecule enhances EGFRvIII specific CAR-T cells in xenograft experiments in mouse models. PLoS One 2024;19(10):e0310430 View Article PubMed/NCBI
  70. Marei HE, Althani A, Afifi N, Hasan A, Caceci T, Pozzoli G, et al. Current progress in chimeric antigen receptor T cell therapy for glioblastoma multiforme. Cancer Med 2021;10(15):5019-5030 View Article PubMed/NCBI
  71. Martins TA, Kaymak D, Tatari N, Gerster F, Hogan S, Ritz MF, et al. Enhancing anti-EGFRvIII CAR T cell therapy against glioblastoma with a paracrine SIRPγ-derived CD47 blocker. Nat Commun 2024;15(1):9718 View Article PubMed/NCBI
  72. Zhou S, Lin W, Jin X, Niu R, Yuan Z, Chai T, et al. CD97 maintains tumorigenicity of glioblastoma stem cells via mTORC2 signaling and is targeted by CAR Th9 cells. Cell Rep Med 2024;5(12):101844 View Article PubMed/NCBI
  73. Zhai Y, Li G, Pan C, Yu M, Hu H, Wang D, et al. The development and potent antitumor efficacy of CD44/CD133 dual-targeting IL7Rα-armored CAR-T cells against glioblastoma. Cancer Lett 2025;614:217541 View Article PubMed/NCBI
  74. Schmidts A, Srivastava AA, Ramapriyan R, Bailey SR, Bouffard AA, Cahill DP, et al. Tandem chimeric antigen receptor (CAR) T cells targeting EGFRvIII and IL-13Rα2 are effective against heterogeneous glioblastoma. Neurooncol Adv 2023;5(1):vdac185 View Article PubMed/NCBI
  75. Wang D, Starr R, Chang WC, Aguilar B, Alizadeh D, Wright SL, et al. Chlorotoxin-directed CAR T cells for specific and effective targeting of glioblastoma. Sci Transl Med 2020;12(533):eaaw2672 View Article PubMed/NCBI
  76. Hamieh M, Mansilla-Soto J, Rivière I, Sadelain M. Programming CAR T Cell Tumor Recognition: Tuned Antigen Sensing and Logic Gating. Cancer Discov 2023;13(4):829-843 View Article PubMed/NCBI
  77. Bassan D, Weinberger L, Yi J, Kim T, Weist MR, Adams GB, et al. HER2 and HLA-A*02 dual CAR-T cells utilize LOH in a NOT logic gate to address on-target off-tumor toxicity. J Immunother Cancer 2023;11(12):e007426 View Article PubMed/NCBI
  78. Choe JH, Watchmaker PB, Simic MS, Gilbert RD, Li AW, Krasnow NA, et al. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med 2021;13(591):eabe7378 View Article PubMed/NCBI
  79. Tousley AM, Rotiroti MC, Labanieh L, Rysavy LW, Kim WJ, Lareau C, et al. Co-opting signalling molecules enables logic-gated control of CAR T cells. Nature 2023;615(7952):507-516 View Article PubMed/NCBI
  80. Liu D, Zhao J, Song Y. Engineering switchable and programmable universal CARs for CAR T therapy. J Hematol Oncol 2019;12(1):69 View Article PubMed/NCBI
  81. Nagy L, Mezősi-Csaplár M, Rebenku I, Vereb G, Szöőr Á. Universal CAR T cells targeted to HER2 with a biotin-trastuzumab soluble linker penetrate spheroids and large tumor xenografts that are inherently resistant to trastuzumab mediated ADCC. Front Immunol 2024;15:1365172 View Article PubMed/NCBI
  82. Tripathy RK, Pande AH. Molecular and functional insight into anti-EGFR nanobody: Theranostic implications for malignancies. Life Sci 2024;345:122593 View Article PubMed/NCBI
  83. Loff S, Dietrich J, Meyer JE, Riewaldt J, Spehr J, von Bonin M, et al. Rapidly Switchable Universal CAR-T Cells for Treatment of CD123-Positive Leukemia. Mol Ther Oncolytics 2020;17:408-420 View Article PubMed/NCBI
  84. Peschke JC, Bergmann R, Mehnert M, Gonzalez Soto KE, Loureiro LR, Mitwasi N, et al. FLT3-directed UniCAR T-cell therapy of acute myeloid leukaemia. Br J Haematol 2023;202(6):1137-1150 View Article PubMed/NCBI
  85. Wang D, Dou L, Sui L, Xue Y, Xu S. Natural killer cells in cancer immunotherapy. MedComm (2020) 2024;5(7):e626 View Article PubMed/NCBI
  86. Chaouat AE, Seliger B, Mandelboim O, Schmiedel D. The HHV-6A Proteins U20 and U21 Target NKG2D Ligands to Escape Immune Recognition. Front Immunol 2021;12:714799 View Article PubMed/NCBI
  87. Zhang Y, Li X, Zhang J, Mao L. Novel cellular immunotherapy using NKG2D CAR-T for the treatment of cervical cancer. Biomed Pharmacother 2020;131:110562 View Article PubMed/NCBI
  88. Deng Y, Kumar A, Xie K, Schaaf K, Scifo E, Morsy S, et al. Targeting senescent cells with NKG2D-CAR T cells. Cell Death Discov 2024;10(1):217 View Article PubMed/NCBI
  89. Yang D, Sun B, Dai H, Li W, Shi L, Zhang P, et al. T cells expressing NKG2D chimeric antigen receptors efficiently eliminate glioblastoma and cancer stem cells. J Immunother Cancer 2019;7(1):171 View Article PubMed/NCBI
  90. Eisele G, Wischhusen J, Mittelbronn M, Meyermann R, Waldhauer I, Steinle A, et al. TGF-beta and metalloproteinases differentially suppress NKG2D ligand surface expression on malignant glioma cells. Brain 2006;129(Pt 9):2416-2425 View Article PubMed/NCBI
  91. Liu J, Dai K, Saliu MA, Salisu MD, Gan J, Afolabi LO, et al. Sodium valproate enhances efficacy of NKG2D CAR-T cells against glioblastoma. Front Immunol 2024;15:1519777 View Article PubMed/NCBI
  92. Alkhayer R, Ponath V, Frech M, Adhikary T, Graumann J, Neubauer A, et al. KLF4-mediated upregulation of the NKG2D ligand MICA in acute myeloid leukemia: a novel therapeutic target identified by enChIP. Cell Commun Signal 2023;21(1):94 View Article PubMed/NCBI
  93. Ding B, Li J, Yan JL, Jiang CY, Qian LB, Pan J. Resveratrol contributes to NK cell-mediated breast cancer cytotoxicity by upregulating ULBP2 through miR-17-5p downmodulation and activation of MINK1/JNK/c-Jun signaling. Front Immunol 2025;16:1515605 View Article PubMed/NCBI
  94. Li M, Li G, Kiyokawa J, Tirmizi Z, Richardson LG, Ning J, et al. Characterization and oncolytic virus targeting of FAP-expressing tumor-associated pericytes in glioblastoma. Acta Neuropathol Commun 2020;8(1):221 View Article PubMed/NCBI
  95. Niu W, Wang B, Zhang Y, Wang C, Cao J, Li J, et al. Efficacy and safety evaluation of cross-reactive Fibroblast activation protein scFv-based CAR-T cells. Front Immunol 2024;15:1433679 View Article PubMed/NCBI
  96. Yu W, Truong NT, Polara R, Gargett T, Tea MN, Pitson SM, et al. Endogenous bystander killing mechanisms enhance the activity of novel FAP-specific CAR-T cells against glioblastoma. Clin Transl Immunology 2024;13(7):e1519 View Article PubMed/NCBI
  97. Choi BD, Yu X, Castano AP, Darr H, Henderson DB, Bouffard AA, et al. CRISPR-Cas9 disruption of PD-1 enhances activity of universal EGFRvIII CAR T cells in a preclinical model of human glioblastoma. J Immunother Cancer 2019;7(1):304 View Article PubMed/NCBI
  98. Wang X, Lu J, Guo G, Yu J. Immunotherapy for recurrent glioblastoma: practical insights and challenging prospects. Cell Death Dis 2021;12(4):299 View Article PubMed/NCBI
  99. Zhu H, You Y, Shen Z, Shi L. EGFRvIII-CAR-T Cells with PD-1 Knockout Have Improved Anti-Glioma Activity. Pathol Oncol Res 2020;26(4):2135-2141 View Article PubMed/NCBI
  100. Zhang G, Zhao Y, Liu Z, Liu W, Wu H, Wang X, et al. GD2 CAR-T cells in combination with Nivolumab exhibit enhanced antitumor efficacy. Transl Oncol 2023;32:101663 View Article PubMed/NCBI
  101. Shen SH, Woroniecka K, Barbour AB, Fecci PE, Sanchez-Perez L, Sampson JH. CAR T cells and checkpoint inhibition for the treatment of glioblastoma. Expert Opin Biol Ther 2020;20(6):579-591 View Article PubMed/NCBI
  102. Yin Y, Boesteanu AC, Binder ZA, Xu C, Reid RA, Rodriguez JL, et al. Checkpoint Blockade Reverses Anergy in IL-13Rα2 Humanized scFv-Based CAR T Cells to Treat Murine and Canine Gliomas. Mol Ther Oncolytics 2018;11:20-38 View Article PubMed/NCBI
  103. Bagley SJ, Binder ZA, Lamrani L, Marinari E, Desai AS, Nasrallah MP, et al. Repeated peripheral infusions of anti-EGFRvIII CAR T cells in combination with pembrolizumab show no efficacy in glioblastoma: a phase 1 trial. Nat Cancer 2024;5(3):517-531 View Article PubMed/NCBI
  104. Tang OY, Tian L, Yoder T, Xu R, Kulikovskaya I, Gupta M, et al. PD1 Expression in EGFRvIII-Directed CAR T Cell Infusion Product for Glioblastoma Is Associated with Clinical Response. Front Immunol 2022;13:872756 View Article PubMed/NCBI
  105. Landi D, Navai SA, Brock RM, Fousek K, Nawas Z, Sanber K, et al. A Checkpoint Reversal Receptor Mediates Bipartite Activation and Enhances CAR T-cell Function. Cancer Res Commun 2025;5(3):527-548 View Article PubMed/NCBI
  106. Hui X, Farooq MA, Chen Y, Ajmal I, Ren Y, Xue M, et al. A novel strategy of co-expressing CXCR5 and IL-7 enhances CAR-T cell effectiveness in osteosarcoma. Front Immunol 2024;15:1462076 View Article PubMed/NCBI
  107. Yu Z, Li H, Lu Q, Zhang Z, Tong A, Niu T. Fc receptor-like 5 (FCRL5)-directed CAR-T cells exhibit antitumor activity against multiple myeloma. Signal Transduct Target Ther 2024;9(1):16 View Article PubMed/NCBI
  108. He B, Chen H, Wu J, Qiu S, Mai Q, Zeng Q, et al. Interleukin-21 engineering enhances CD19-specific CAR-NK cell activity against B-cell lymphoma via enriched metabolic pathways. Exp Hematol Oncol 2025;14(1):51 View Article PubMed/NCBI
  109. Swan SL, Mehta N, Ilich E, Shen SH, Wilkinson DS, Anderson AR, et al. IL7 and IL7 Flt3L co-expressing CAR T cells improve therapeutic efficacy in mouse EGFRvIII heterogeneous glioblastoma. Front Immunol 2023;14:1085547 View Article PubMed/NCBI
  110. Fischer-Riepe L, Kailayangiri S, Zimmermann K, Pfeifer R, Aigner M, Altvater B, et al. Preclinical Development of CAR T Cells with Antigen-Inducible IL18 Enforcement to Treat GD2-Positive Solid Cancers. Clin Cancer Res 2024;30(16):3564-3577 View Article PubMed/NCBI
  111. Luo H, Su J, Sun R, Sun Y, Wang Y, Dong Y, et al. Coexpression of IL7 and CCL21 Increases Efficacy of CAR-T Cells in Solid Tumors without Requiring Preconditioned Lymphodepletion. Clin Cancer Res 2020;26(20):5494-5505 View Article PubMed/NCBI
  112. Dos Santos DMC, Rejeski K, Winkelmann M, Liu L, Trinkner P, Günther S, et al. Increased visceral fat distribution and body composition impact cytokine release syndrome onset and severity after CD19 chimeric antigen receptor T-cell therapy in advanced B-cell malignancies. Haematologica 2022;107(9):2096-2107 View Article PubMed/NCBI
  113. Jindal V. Role of Chimeric Antigen Receptor T Cell Therapy in Glioblastoma Multiforme. Mol Neurobiol 2018;55(11):8236-8242 View Article PubMed/NCBI
  114. Yuan B, Wang G, Tang X, Tong A, Zhou L. Immunotherapy of glioblastoma: Recent advances and future prospects. Hum Vaccin Immunother 2022;18(5):2055417 View Article PubMed/NCBI
  115. Jain P, Vashist S, Panjiyar BK. Navigating the Immune Challenge in Glioblastoma: Exploring Immunotherapeutic Avenues for Overcoming Immune Suppression. Cureus 2023;15(9):e46089 View Article PubMed/NCBI
  116. Niedbała M, Malarz K, Sharma G, Kramer-Marek G, Kaspera W. Glioblastoma: Pitfalls and Opportunities of Immunotherapeutic Combinations. Onco Targets Ther 2022;15:437-468 View Article PubMed/NCBI
  117. Nabors LB, Lamb LS, Goswami T, Rochlin K, Youngblood SL. Adoptive cell therapy for high grade gliomas using simultaneous temozolomide and intracranial mgmt-modified γδ t cells following standard post-resection chemotherapy and radiotherapy: current strategy and future directions. Front Immunol 2024;15:1299044 View Article PubMed/NCBI
  118. Banstola A, Poudel K, Pathak S, Shrestha P, Kim JO, Jeong JH, et al. Hypoxia-Mediated ROS Amplification Triggers Mitochondria-Mediated Apoptotic Cell Death via PD-L1/ROS-Responsive, Dual-Targeted, Drug-Laden Thioketal Nanoparticles. ACS Appl Mater Interfaces 2021;13(19):22955-22969 View Article PubMed/NCBI
  119. Li C, Zhang N, Xu Z, Rong Z, Song C, Zhang Y, et al. Inflammasome mediated in situ cancer vaccine activated by schottky heterojunction for augmented immunotherapy. J Control Release 2025;380:1184-1197 View Article PubMed/NCBI
  120. Suryadevara CM, Desai R, Abel ML, Riccione KA, Batich KA, Shen SH, et al. Temozolomide lymphodepletion enhances CAR abundance and correlates with antitumor efficacy against established glioblastoma. Oncoimmunology 2018;7(6):e1434464 View Article PubMed/NCBI
  121. Amini L, Silbert SK, Maude SL, Nastoupil LJ, Ramos CA, Brentjens RJ, et al. Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion. Nat Rev Clin Oncol 2022;19(5):342-355 View Article PubMed/NCBI
  122. Ohno M, Ohkuri T, Kosaka A, Tanahashi K, June CH, Natsume A, et al. Expression of miR-17-92 enhances anti-tumor activity of T-cells transduced with the anti-EGFRvIII chimeric antigen receptor in mice bearing human GBM xenografts. J Immunother Cancer 2013;1:21 View Article PubMed/NCBI
  123. Akhavan D, Subham S, Jeppson JD, Aguilar B, Wong RA, Hibbard JC, et al. Evaluation of the Immunomodulatory Effects of Radiation for Chimeric Antigen Receptor T Cell Therapy in Glioblastoma Multiforme. Cells 2024;13(13):1075 View Article PubMed/NCBI
  124. Sun T, Li Y, Yang Y, Liu B, Cao Y, Yang W. Enhanced radiation-induced immunogenic cell death activates chimeric antigen receptor T cells by targeting CD39 against glioblastoma. Cell Death Dis 2022;13(10):875 View Article PubMed/NCBI
  125. Barker HE, Paget JT, Khan AA, Harrington KJ. The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence. Nat Rev Cancer 2015;15(7):409-425 View Article PubMed/NCBI
  126. Deng S, Wang J, Hu Y, Sun Y, Yang X, Zhang B, et al. Irradiated tumour cell-derived microparticles upregulate MHC-I expression in cancer cells via DNA double-strand break repair pathway. Cancer Lett 2024;592:216898 View Article PubMed/NCBI
  127. De Ruysscher D, Reynders K, Van Limbergen E, Lambrecht M. Radiotherapy in combination with immune checkpoint inhibitors. Curr Opin Oncol 2017;29(2):105-111 View Article PubMed/NCBI
  128. Xue H, Chen Y, Zhou Y. Radioimmunotherapy: a game-changer for advanced non-small cell lung cancer. Front Immunol 2024;15:1522508 View Article PubMed/NCBI
  129. Ni H, Reitman ZJ, Zou W, Akhtar MN, Paul R, Huang M, et al. FLASH radiation reprograms lipid metabolism and macrophage immunity and sensitizes medulloblastoma to CAR-T cell therapy. Nat Cancer 2025;6(3):460-473 View Article PubMed/NCBI
  130. Wursthorn A, Schwager C, Kurth I, Peitzsch C, Herold-Mende C, Debus J, et al. High-Complexity cellular barcoding and clonal tracing reveals stochastic and deterministic parameters of radiation resistance. Int J Cancer 2022;150(4):663-677 View Article PubMed/NCBI
  131. Harris M, Svensson F, Kopanitsa L, Ladds G, Bailey D. Emerging patents in the therapeutic areas of glioma and glioblastoma. Expert Opin Ther Pat 2018;28(7):573-590 View Article PubMed/NCBI
  132. Dong X, Ren J, Amoozgar Z, Lee S, Datta M, Roberge S, et al. Anti-VEGF therapy improves EGFR-vIII-CAR-T cell delivery and efficacy in syngeneic glioblastoma models in mice. J Immunother Cancer 2023;11(3):e005583 View Article PubMed/NCBI
  133. Elmadany N, Alhalabi OT, Platten M, Bunse L. Site-Specific Considerations on Engineered T Cells for Malignant Gliomas. Biomedicines 2022;10(7):1738 View Article PubMed/NCBI
  134. Angom RS, Nakka NMR, Bhattacharya S. Advances in Glioblastoma Therapy: An Update on Current Approaches. Brain Sci 2023;13(11):1536 View Article PubMed/NCBI
  135. Vitanza NA, Choe M, Brown C, Beebe A, Kong A, Rogers L, et al. Locoregional CAR T Cells for the Treatment of CNS Tumors in Children: Investigational Drug Service Pharmacy Activities. J Hematol Oncol Pharm 2024;14(4):148-154 View Article PubMed/NCBI
  136. Donovan LK, Delaidelli A, Joseph SK, Bielamowicz K, Fousek K, Holgado BL, et al. Locoregional delivery of CAR T cells to the cerebrospinal fluid for treatment of metastatic medulloblastoma and ependymoma. Nat Med 2020;26(5):720-731 View Article PubMed/NCBI
  137. Gao L, Shi C, Yang Z, Jing W, Han M, Zhang J, et al. Convection-enhanced delivery of nanoencapsulated gene locoregionally yielding ErbB2/Her2-specific CAR-macrophages for brainstem glioma immunotherapy. J Nanobiotechnology 2023;21(1):56 View Article PubMed/NCBI
  138. Chang Y, Cai X, Syahirah R, Yao Y, Xu Y, Jin G, et al. CAR-neutrophil mediated delivery of tumor-microenvironment responsive nanodrugs for glioblastoma chemo-immunotherapy. Nat Commun 2023;14(1):2266 View Article PubMed/NCBI
  139. Prapa M, Chiavelli C, Golinelli G, Grisendi G, Bestagno M, Di Tinco R, et al. GD2 CAR T cells against human glioblastoma. NPJ Precis Oncol 2021;5(1):93 View Article PubMed/NCBI
  140. Martins P, D’Souza RCJ, Skarne N, Lekieffre L, Horsefield S, Ranjankumar M, et al. EphA3 CAR T cells are effective against glioblastoma in preclinical models. J Immunother Cancer 2024;12(8):e009403 View Article PubMed/NCBI
  141. Zhu G, Sun Z, Liu Y, Liu J, Guo L, Pei G, et al. Rational Design and Organoid-Based Evaluation of a Cocktail CAR-γδ T Cell Therapy for Heterogeneous Glioblastoma. Adv Sci (Weinh) 2025;12(19):e2501772 View Article PubMed/NCBI
  142. Wang X, Sun Q, Wang W, Liu B, Gu Y, Chen L. Decoding key cell sub-populations and molecular alterations in glioblastoma at recurrence by single-cell analysis. Acta Neuropathol Commun 2023;11(1):125 View Article PubMed/NCBI
  143. De Silva MI, Stringer BW, Bardy C. Neuronal and tumourigenic boundaries of glioblastoma plasticity. Trends Cancer 2023;9(3):223-236 View Article PubMed/NCBI
  144. Huang Y, Shao M, Teng X, Si X, Wu L, Jiang P, et al. Inhibition of CD38 enzymatic activity enhances CAR-T cell immune-therapeutic efficacy by repressing glycolytic metabolism. Cell Rep Med 2024;5(2):101400 View Article PubMed/NCBI
  145. Qin C, Dong MH, Zhou LQ, Wang W, Cai SB, You YF, et al. Single-cell analysis of refractory anti-SRP necrotizing myopathy treated with anti-BCMA CAR-T cell therapy. Proc Natl Acad Sci U S A 2024;121(6):e2315990121 View Article PubMed/NCBI
  146. Jacob F, Salinas RD, Zhang DY, Nguyen PTT, Schnoll JG, Wong SZH, et al. A Patient-Derived Glioblastoma Organoid Model and Biobank Recapitulates Inter- and Intra-tumoral Heterogeneity. Cell 2020;180(1):188-204.e22 View Article PubMed/NCBI
  147. Jacob F, Ming GL, Song H. Generation and biobanking of patient-derived glioblastoma organoids and their application in CAR T cell testing. Nat Protoc 2020;15(12):4000-4033 View Article PubMed/NCBI
  148. Kong D, Kwon D, Moon B, Kim DH, Kim MJ, Choi J, et al. CD19 CAR-expressing iPSC-derived NK cells effectively enhance migration and cytotoxicity into glioblastoma by targeting to the pericytes in tumor microenvironment. Biomed Pharmacother 2024;174:116436 View Article PubMed/NCBI
  149. Logun M, Wang X, Sun Y, Bagley SJ, Li N, Desai A, et al. Patient-derived glioblastoma organoids as real-time avatars for assessing responses to clinical CAR-T cell therapy. Cell Stem Cell 2025;32(2):181-190.e4 View Article PubMed/NCBI
  150. Thokala R, Binder ZA, Yin Y, Zhang L, Zhang JV, Zhang DY, et al. High-Affinity Chimeric Antigen Receptor With Cross-Reactive scFv to Clinically Relevant EGFR Oncogenic Isoforms. Front Oncol 2021;11:664236 View Article PubMed/NCBI
  151. Song EZ, Wang X, Philipson BI, Zhang Q, Thokala R, Zhang L, et al. The IAP antagonist birinapant enhances chimeric antigen receptor T cell therapy for glioblastoma by overcoming antigen heterogeneity. Mol Ther Oncolytics 2022;27:288-304 View Article PubMed/NCBI
  152. Wang C, Yu M, Zhang W. Neoantigen discovery and applications in glioblastoma: An immunotherapy perspective. Cancer Lett 2022;550:215945 View Article PubMed/NCBI

About this Article

Cite this article
Zhang L, Zhang Z, He J, Zhang Z, Zhou H, Jiang Y, et al. Dual Challenges and Innovative Strategies in Chimeric Antigen Receptor T-cell Therapy for Glioblastoma. Oncol Adv. Published online: Jul 30, 2025. doi: 10.14218/OnA.2025.00014.
Copy Export to RIS Export to EndNote
Article History
Received Revised Accepted Published
May 7, 2025 June 24, 2025 July 1, 2025 July 30, 2025
DOI http://dx.doi.org/10.14218/OnA.2025.00014