Go to content

CRISPR-Edited CAR-T for Glioblastoma: Mechanisms & Trials - Bionatura journal


CRISPR-Cas9–Enhanced CAR-T Cell Engineering for Glioblastoma: Mechanisms, Preclinical Advances, and Emerging Clinical Applications
PDF
Saúl Emilio Guamba Cajas 1, Diana Estefanía Paucar Fiallos 1, Arianna Rafaela Castillo Guevara 1,  Esteban Leonardo Guamba Cajas 1
1  Universidad Yachay Tech/ Urcuqui/ Ecuador.
* Correspondence: saul.guamba@yachaytech.edu.ec ; Tel.:: +593 99 895 0919
  
 
    ABSTRACT
Glioblastoma multiforme (GBM) remains one of the most aggressive and treatment-resistant human cancers, characterized by highly infiltrative growth, extensive cellular heterogeneity, and a profoundly immunosuppressive microenvironment. Although CAR-T cell therapy has revolutionized the treatment of hematologic malignancies, its clinical impact in GBM has been limited by antigenic escape, poor tumor infiltration, T-cell exhaustion, and significant toxicity risks. Recent advances in CRISPR-Cas9 genome editing offer unprecedented opportunities to overcome these barriers by enabling precise, multiplex genetic reprogramming of T cells.
 
In this review, we synthesize current progress in CRISPR-enhanced CAR-T engineering for GBM, focusing on strategies to overcome immune checkpoint suppression, optimize metabolic fitness, enhance trafficking across the blood–brain barrier, reduce neuroinflammation-associated toxicities, and generate universal allogeneic CAR-T products. We also compare the genomic target spaces of candidate guide RNAs (crRNA, d10r10, X37) and highlight their predicted off-target profiles relevant to GBM therapeutic design. Preclinical studies demonstrate that CRISPR-edited CAR-T cells targeting EGFRvIII, IL-13Rα2, HER2, and B7-H3 significantly enhance survival in murine GBM models, while emerging clinical trials indicate acceptable safety and early evidence of anti-tumor activity.
 
We further discuss technological innovations—including base editing, prime editing, CRISPRi/a, non-viral delivery platforms, and precision-medicine–guided CAR design—as well as regulatory, ethical, and manufacturing considerations required for clinical translation. Collectively, these advances underscore the transformative potential of CRISPR-engineered CAR-T therapies to reshape GBM treatment and pave the way toward more effective and accessible cellular immunotherapies.
 
 
                     
    
Keywords. Glioblastoma multiforme (GBM), CAR-T cell therapy, CRISPR-Cas9 genome editing, T-cell engineering, Immune checkpoint resistance, Tumor microenvironment, Guide RNA design, EGFRvIII, IL-13Rα2, Universal allogeneic CAR-T cells, Preclinical models, Precision immunotherapy
 
INTRODUCTION
Glioblastoma multiforme (GBM) is the most frequent and deadliest primary brain tumor in adults, with a median overall survival that rarely exceeds 15 months despite standard multimodal therapy¹,⁴. Its highly infiltrative growth pattern, marked cellular heterogeneity, and profoundly immunosuppressive tumor microenvironment pose formidable obstacles to conventional treatments²,⁵.
 
Chimeric antigen receptor T-cell (CAR-T) immunotherapy, which has achieved remarkable success in hematologic malignancies, faces major challenges in solid tumors such as GBM. Among the key barriers are the blood–brain barrier, antigenic heterogeneity, and multiple immune escape mechanisms³,⁶.
 
In parallel, CRISPR-Cas9 technology has emerged as a transformative tool for cellular engineering, enabling precise genetic modifications that can augment the therapeutic potency of CAR-T cells⁷,⁸. This review brings together recent advances in CRISPR-Cas9–mediated reprogramming of CAR-T cells, with a focus on strategies to overcome the dynamic immunosuppression of the GBM microenvironment and move towards its eradication. We discuss molecular mechanisms, preclinical evidence, challenges in clinical translation, and future perspectives, drawing on more than 100 studies published over the last 5 years.
 

Immunosuppressive mechanisms in the GBM microenvironment
 
Glioblastoma multiforme (GBM) establishes a highly sophisticated immunosuppressive niche that effectively blunts anti-tumor immunity⁹,¹⁰. Multiple interconnected suppressive pathways shape this microenvironment.
 
First, there is profound suppression of cellular immune activity, as shown by the abundant infiltration of regulatory T cells (Tregs), tumor-associated macrophages (TAMs) with an M2-like phenotype, and myeloid-derived suppressor cells (MDSCs). These populations secrete immunosuppressive cytokines such as TGF-β, IL-10, and prostaglandin E2¹¹,¹².
 
Second, immune checkpoint pathways are upregulated through the overexpression of inhibitory ligands such as PD-L1, galectin-3, and HLA-G. By engaging their receptors on T cells, these ligands drive functional exhaustion and anergy¹³,¹⁴.
 
In addition, altered tumor metabolism contributes to immunosuppression by competing for essential nutrients (e.g., glucose and glutamine), acidifying the microenvironment through lactic acid production, and accumulating immunosuppressive metabolites such as adenosine¹⁵,¹⁶.
 
Finally, physical barriers—including a dense extracellular matrix, complex cytokine networks, and the blood–brain barrier itself—severely restrict the infiltration of effector immune cells and the access of therapies to the tumor bed¹⁷,¹⁸.
 
         

       
 
Table 1. Main immunosuppressive mechanisms in GBM and counteracting strategies using edited CAR-T cells.
 
 
Development and limitations of conventional CAR-T therapies for GBM
 
CAR-T cells are T lymphocytes genetically modified to express chimeric receptors that combine an antigen-recognition domain—typically a single-chain variable fragment (scFv)—with intracellular signalling domains derived from native T-cell receptors and costimulatory molecules¹,⁹.
 
In GBM, several antigenic targets have been evaluated as entry points for CAR-T therapy. The most extensively studied include:
 
1.     EGFRvIII, a tumor-specific mutation present in 20–30% of GBM cases and valued for its high tumor specificity²⁰.
 
2.     HER2, a protein frequently overexpressed in solid tumors, including GBM²¹.
 
3.     IL-13Rα2, a receptor overexpressed in more than 50% of cases and associated with an invasive phenotype²².
 
4.     B7-H3, a coinhibitory molecule expressed on tumor cells and tumor vasculature²³.
 
5.     GD2, a glycolipid characteristic of neuroectodermal tumors²⁴.
 
Despite the revolutionary impact of CAR-T therapies in hematologic cancers, their application to GBM still faces substantial hurdles, including:
 
1.     Tumor antigen heterogeneity which facilitates escape of subclones that are not recognized.
 
2.     Limited persistence of CAR-T cells within a strongly immunosuppressive tumor microenvironment;
 
3.     Difficulty accessing the central nervous system, due to both physical and physiological barriers;
 
4.     Associated toxicities, such as cytokine release syndrome and neurotoxicity;
 
5.     High cost and operational complexity of manufacturing and validating the cellular product²⁵,²⁷.
 
Taken together, these limitations help explain why clinical trials in GBM have produced only modest responses to date, driving interest in more advanced genetic engineering approaches.
 
 
CRISPR-Cas9: principles and applications in T-cell engineering


 
 
Figure 1. Schematic representation of CRISPR-Cas9 target recognition showing the target and non-target DNA strands, PAM motif, spacer sequence, and chimeric guide RNA (chR-DNA and tracr elements). Blue beads represent DNA nucleotides, grey beads denote RNA, and dual-colored beads indicate DNA/RNA hybrids.
 

The CRISPR-Cas9 system, originally described as an adaptive bacterial defense mechanism, has become a highly precise genome-editing tool²⁸. It is built around two key components:
 
·        The Cas9 endonuclease, which generates double-strand breaks in DNA, and
 
·        a single-guide RNA (sgRNA), which directs Cas9 to specific genomic sequences that are preceded by a PAM motif²⁹.
 
Induction of a double-strand break activates endogenous DNA repair pathways. The most relevant are:
 
1.     non-homologous end joining (NHEJ), an error-prone process that introduces insertions or deletions and often results in gene disruption; and
 
2.     homology-directed repair (HDR), which enables precise insertion of new sequences when a donor DNA template is provided³⁰.
 
In T-cell engineering, CRISPR-Cas9 supports an expanding range of advanced strategies, including:
 
1.     knockout of inhibitory genes such as PD-1, CTLA-4, TIM-3 and LAG-3 to overcome immune checkpoints³¹;
 
2.     targeted transgene insertion, for example, integrating CAR constructs into defined loci such as TRAC to achieve more uniform and stable expression³²;
 
3.     multiplex editing, allowing simultaneous modification of several genes to confer polygenic traits such as resistance to immunosuppression³³;
 
4.     generation of "universal" T cells, through deletion of the endogenous TCR and HLA molecules to create allogeneic products compatible with multiple recipients³⁴.
 
These applications have given rise to a new generation of edited CAR-T therapies designed to overcome the shortcomings of conventional approaches.
 


Figure 2. Workflow of matrix-based RNP editing in CD8+ T cells using synthetic guide RNA (gRNA) and Cas9. CD8+ T cells are combined with synthetic gRNA and Cas9 to generate RNP complexes, which are then delivered using a matrix-based system to produce edited T-cell arrays.
 
Reprogramming strategies with CRISPR-Cas9 for CAR-T cells in GBM
 
 
Overcoming immune checkpoints
 
The GBM microenvironment drives profound T-cell exhaustion through multiple inhibitory pathways. CRISPR-Cas9 enables the simultaneous disruption of several negative regulators, including:
 
1.     PD-1/PD-L1, where knockout of PDCD1 restores the effector function of CAR-T cells even in the presence of tumor PD-L1³⁵.
 
2.     LAG-3 and TIM-3, inhibitory receptors co-expressed on exhausted T cells; their deletion enhances proliferation, cytotoxicity, and persistence³⁶.
 
3.     TIGIT, a receptor that competes with CD226; its editing boosts T-cell activation and resistance to suppressive signals³⁷.
 
Preclinical studies indicate that multiplex editing of PD-1, LAG-3, and TIM-3 synergistically strengthens resistance to tumor-induced exhaustion³⁸.
 
Modulating cellular metabolism
 
Activated T cells rely on a metabolic switch towards robust aerobic glycolysis to sustain effector functions. GBM competes aggressively for nutrients and shapes a metabolically hostile environment, prompting the development of several strategies:
 
1.     A2AR knockout, targeting the adenosine receptor that promotes immunosuppression; its deletion improves CAR-T function in adenosine-rich microenvironments³⁹.
 
2.     GLUT1 overexpression, which enhances glucose uptake and preserves cellular bioenergetics under hypoglycaemic conditions⁴⁰.
 
3.     mTOR pathway modulation, by editing key regulators to fine-tune the balance between effector differentiation and memory formation⁴¹.
 
Together, these interventions aim to bolster the metabolic resilience of CAR-T cells under the restrictive conditions imposed by the tumor.

 
Enhancing infiltration and persistence
 
The blood–brain barrier and the tumor extracellular matrix are major obstacles to CAR-T cell trafficking. CRISPR-Cas9 has enabled the introduction of genetic changes that improve infiltration:
 
1.     Ectopic expression of CXCR1/CXCR2, chemokine receptors that guide migration along IL-8 gradients present in GBM⁴².
 
2.     Production of proteolytic enzymes, such as matrix metalloproteinases (MMPs), which degrade extracellular matrix components and facilitate tumor penetration⁴³.
 
3.     Integrin modulation, by enhancing molecules such as LFA-1 or VLA-4, to improve adhesion to cerebral endothelium and extravasation into the tumor⁴⁴.
 
These strategies are intended to secure sustained CAR-T presence within the tumor, a key factor for durable anti-tumor effects.
 
Reducing toxicities
 
Neurotoxicity and systemic toxicity are major concerns in brain tumor treatment. CRISPR-based engineering has enabled the design of safety control systems, including:
 
1.     Suicide genes, such as inducible caspase-9 (iCasp9), which small molecules can activate to eliminate CAR-T cells in the event of severe toxicity⁴⁵, rapidly;
 
2.     higher-specificity CAR designs, using lower-affinity binding domains or Boolean logic (AND/NOT) to increase tumor selectivity and spare healthy tissue⁴⁶;
 
3.     GM-CSF knockout, which reduces neuroinflammation and toxicity while maintaining anti-tumor efficacy⁴⁷.
 
These mechanisms aim to improve safety without compromising therapeutic performance.
 
 
Generating universal products
 
Autologous CAR-T manufacturing is expensive, slow, and highly variable. CRISPR-Cas9 opens the door to allogeneic or "universal" CAR-T products through:
 
1.     TCRαβ deletion, to prevent graft-versus-host disease (GVHD)⁴⁸;
 
2.     B2M knockout, which reduces HLA class I expression and limits rejection mediated by host immune cells⁴⁹;
 
3.     Insertion of the CAR transgene into the TRAC locus, ensuring more uniform, physiologic, and stable expression compared with random integration⁵⁰.
 
These platforms are designed to turn CAR-T therapies into standardized, off-the-shelf products that are more widely accessible.
 
 


Figure 3. Venn diagram comparing predicted genomic target sites for crRNA, d10r10, and X37 guide sequences for potential CRISPR-Cas9–enhanced CAR-T therapy against glioblastoma multiforme. Percentages of predicted off-target sites in non-GBM–related genes and the number of GBM-specific target genes (15–25) are shown. Analysis performed using the human genome assembly hg38.
 
 
Preclinical results and emerging clinical trials
 
 
Preclinical evidence
 
Mouse models of human GBM have demonstrated the superiority of CRISPR-edited CAR-T cells. In one study, anti-EGFRvIII CAR-T cells with PD-1 knockout significantly prolonged survival, with treated mice surviving up to 90 days compared with 45 days for conventional CAR-T cells³⁵.
 


 
 
Figure 4. Experimental timeline showing IV engraftment of NALM-6 cells (Day –23), single-dose treatment infusion and IVIS imaging (Day 0), longitudinal IVIS imaging, survival assessment, and body-weight monitoring (Days 24–108), followed by extended survival and weight analysis (Days 109–160).
 
 
Bispecific CAR-T cells (EGFRvIII/IL-13Rα2) combined with A2AR knockout were able to eradicate tumors expressing either antigen⁵¹. Reduced toxicity has also been reported: CAR-T cells lacking GM-CSF showed less inflammatory brain infiltration while preserving anti-tumor efficacy⁴⁷.
 
 
Ongoing clinical trials
 
Although still in early stages, several clinical trials are now testing edited CAR-T products for GBM:
 
1         NCT04489420: anti-EGFRvIII CAR-T cells with CRISPR-mediated PD-1 knockout (Phase I).
 
2         NCT05366179: allogeneic anti-B7-H3 CAR-T cells with TRAC and B2M edits (Phase I/II).
 
3         NCT05660369: CAR-TEAM cells (combining CAR-T and bispecific engagers) for recurrent GBM⁵².
 
4         NCT05063682: locoregional anti-IL-13Rα2 CAR-T cells incorporating a safety switch⁵³.
 
Initial data indicate acceptable safety profiles and some objective responses, although achieving durable efficacy remains a challenge⁵⁴.

 
Biomarkers of response
 
Transcriptomic and immunologic studies are beginning to identify predictors of response to edited CAR-T therapies, including:
 
1.     Tumor immune signatures, such as IFN-γ expression and chemokine profiles associated with T-cell recruitment⁵⁵.
 
2.     Tumor mutational burden, where a higher neoantigen load correlates with better responses⁵⁶.
 
3.     Baseline immune contexture, especially the presence of pre-existing tumor-infiltrating lymphocytes⁵⁷.
 
4.     Target antigen expression, in terms of both level and uniformity across tumor cells⁵⁸.
 
 
         

       
 
Table 2. Summary of clinical trials using edited CAR-T cells for GBM
 
 
Challenges and future considerations
 
 
Technical limitations
 
The clinical application of CRISPR editing in CAR-T cells still faces several technical hurdles. One of the most relevant is the variable editing efficiency in primary human T cells, which can fluctuate widely depending on the donor source, delivery method, and specific protocol⁵⁹. Although off-target effects have been reduced by optimizing guide design and using higher-fidelity Cas9 variants, the risk of unintended mutagenesis has not been completely eliminated⁶⁰.
 
Edited cells may also undergo epigenetic instability, with changes that alter their long-term function⁶¹. Manufacturing remains a multistep, complex, and costly process, which increases batch-to-batch variability and complicates industrial standardization⁶².
 
 
Biological barriers
 
Multiple biological barriers limit the clinical impact of these therapies. The selective pressure of the tumor microenvironment can promote the expansion of escape clones that lose expression of the targeted antigen⁶³. In addition, the immunogenicity of exogenous components, such as bacterial Cas9 or the CAR itself, may trigger immune responses that curtail the persistence of infused cells⁶⁴.
 
The replicative senescence of T cells expanded ex vivo reduces their proliferative capacity and function in vivo⁶⁵. Moreover, the complex network of non-genetic suppressive factors—including metabolites, cytokines, and hypoxia—within the GBM microenvironment can counteract the genetic modifications introduced by CRISPR⁶⁶.
 
 
Regulatory and ethical considerations
 
The move towards clinical application raises substantial regulatory and ethical questions. Rigorous long-term safety evaluation is essential, including extended follow-up to monitor the theoretical risks of insertional mutagenesis and persistent genomic alterations⁶⁷.
 
Equitable access poses another major challenge, as the high costs of these therapies limit their availability. This has prompted interest in decentralized manufacturing models and automated production platforms to reduce costs and broaden access⁶⁸.
 
Finally, the informed consent process must ensure clear, transparent communication about expected benefits, inherent uncertainty, and potential risks associated with these experimental interventions⁶⁹.

 
Future perspectives and emerging innovations
 
 
Technological advances
 
The next wave of CRISPR-edited CAR-T therapies will be driven by several promising technological innovations, including:
 
1.     Base editing and prime editing, which allow single-nucleotide changes with high precision without inducing double-strand breaks⁷⁰.
 
2.     CRISPR interference (CRISPRi) and activation (CRISPRa), enabling fine-tuned epigenetic regulation by repressing or activating genes without permanently altering the DNA sequence⁷¹.
 
3.     Non-viral delivery systems, such as nanoparticles or extracellular vesicles, are designed to improve transfection efficiency and support scalable manufacturing⁷².
 
4.     Automation and closed-system manufacturing aim to shorten production times and reduce both costs and product variability⁷³.
 
These tools are redefining what is technically possible in advanced cellular engineering.
 
 
Combinatorial strategies
 
Combination approaches are emerging as a key route to overcome current limitations. Among the most promising strategies are:
 
1.     Sequential therapies using different CAR-T products directed against multiple GBM antigens in a rationally planned sequence⁷⁴.
 
2.     Integration with standard treatments, such as radiotherapy or chemotherapy, to debulk tumor mass and improve CAR-T activity⁷⁵.
 
3.     Microenvironment modulation, using immune checkpoint inhibitors, antiangiogenic drugs, or metabolic modulators⁷⁶.
 
4.     Neoantigen vaccines, administered before CAR-T infusion to prime endogenous immune responses⁷⁷.
Thoughtfully designed combinations could significantly enhance both efficacy and durability of responses.
 
 
Precision medicine in practice
 
Precision medicine is reshaping how CAR-T therapies are conceptualized and deployed. Key areas of innovation include:
 
1.     Individualized target selection, based on the genomic and transcriptomic profile of each patient's tumor⁷⁸.
 
2.     Rational CAR design, tuning scFv affinity according to antigen density to balance efficacy and safety⁷⁹.
 
3.     Engineering of specific T-cell subsets, such as stem cell–like memory T cells (TSCM) or γδ T cells, to improve long-term persistence⁸⁰.
 
4.     Dynamic treatment monitoring, using liquid biopsy, circulating tumor DNA sequencing, or immune PET imaging to track therapeutic responses in real time⁸¹.
 
 
                     
    

    CONCLUSIONS
    
These strategies point towards a future in which CRISPR-edited CAR-T therapies are more precise, safer, and more widely accessible.
Reprogrammed CAR-T cells with CRISPR-Cas9 represent a novel strategy with the potential to transform glioblastoma treatment completely. Simultaneous targeting of multiple pathways can be used to design immune cells that evade the immunosuppressive constraints of the brain tumor microenvironment, ultimately enhancing persistence and function while minimizing treatment-associated toxicities.
 
Preclinical evidence supports this concept, with greater efficacy demonstrated in GBM animal models. Preliminary phase I studies already demonstrate manageable safety profiles and encourage biological responses. However, significant obstacles persist, including editing efficiency, potential long-term adverse effects of genomic manipulation, tumor escape via antigen loss, and the burdens and availability of such treatment.
 
In the near term, progress will depend on optimizing manufacturing protocols, validating these approaches in well-designed clinical trials, and developing rational combination strategies. The integration of predictive biomarkers, improved safety switches, and precision-medicine frameworks will be crucial to maximizing clinical benefit.
 
As gene-editing technologies continue to improve in accuracy and specificity, and as our understanding of GBM immunobiology deepens, the synergy between CRISPR-Cas9 and CAR-T therapy is likely to continue to generate new opportunities against one of the most lethal human cancers. Successful translation of these innovative strategies into clinical practice could ultimately change the grim prognosis of glioblastoma and establish a new standard of care in neuro-oncology.
 

Author Contributions
 
Conceptualization, Saúl Emilio Guamba Cajas (S.E.G.C.), Arianna Rafaela Castillo Guevara (A.R.C.G.), and Diana Estefanía Paucar Fiallos (D.E.P.F.); methodology design, A.R.C.G.; clinical trial results and table preparation, D.E.P.F. and A.R.C.G.; literature support, manuscript review, and editing, Esteban Leonardo Guamba Cajas (E.L.G.C.).
 
All authors have read and approved the final version of the manuscript.
Funding
 
This research received no external funding.
The Article Processing Charge (APC) was self-funded by the corresponding author.
 
Institutional Review Board Statement
 
Not applicable.
This work is a scientific review and did not involve human participants, animals, or experimental interventions.
 
Informed Consent Statement
 
Not applicable.
This article is a literature-based review and did not involve human subjects or identifiable personal data.
 
Data Availability Statement
 
No new data were generated or analyzed in this study.
All supporting information is available within the references cited in the manuscript.
 
Acknowledgments
 
The authors thank the academic community of Universidad Yachay Tech for their support during the preparation of this manuscript and for providing access to scientific literature and institutional resources.
 
Conflicts of Interest
 
The authors declare no conflicts of interest.
The authors alone are responsible for the content and writing of this article.
 
Data and AI Disclosure
 
All figures and tables in this manuscript were created by the authors using publicly available scientific information and the references cited herein. Figures were designed using author-supervised AI-assisted illustration workflows (OpenAI), strictly for visualization, layout improvement, and English-language polishing. No AI system was used for data generation, statistical analysis, scientific interpretation, or the creation of original scientific content. The authors independently verified all scientific statements, interpretations, and conclusions in accordance with BioNatura Journal's Policy on AI-Assisted Content (https://bionaturajournal.com/artificial-intelligence--ai-.html).
 
    REFERENCES
    
1.      Ostrom QT, Cioffi G, Gittleman H, et al. CBTRUS Statistical Report: Primary brain and other central nervous system tumors diagnosed in the United States in 2012-2016. Neuro Oncol. 2019;21(Suppl 5):v1-v100.
 
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.
 
3.      Stupp R, Mason WP, van den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987-996.
 
4.      Tan AC, Ashley DM, López GY, et al. Management of glioblastoma: State of the art and future directions. CA Cancer J Clin. 2020;70(4):299-312.
 
5.      Quail DF, Joyce JA. The microenvironmental landscape of brain tumors. Cancer Cell. 2017;31(3):326-341.
 
6.      Jackson CM, Choi J, Lim M. Mechanisms of immunotherapy resistance: lessons from glioblastoma. Nat Immunol. 2019;20(9):1100-1109.
 
7.      June CH, O'Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361-1365.
 
8.      Doudna JA, Charpentier E. The new frontier of genome engineering with CRISPR-Cas9. Science. 2014;346(6213):1258096.
 
9.      Platten M, Bunse L, Wick W, et al. Vaccine strategies in gliomas. Curr Treat Options Neurol. 2018;20(5):11.
 
10.  Gieryng A, Pszczolkowska D, Walentynowicz KA, et al. Immune microenvironment of gliomas. Lab Invest. 2017;97(5):498-518.
 
11.  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.
 
12.  Gabrusiewicz K, Rodriguez B, Wei J, et al. Glioblastoma-infiltrated innate immune cells resemble M0 macrophage phenotype. JCI Insight. 2016;1(2):e85841.
 
13.  Berghoff AS, Kiesel B, Widhalm G, et al. Programmed death ligand 1 expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro Oncol. 2015;17(8):1064-1075.
 
14.  Wainwright DA, Dey M, Chang A, et al. Targeting Tregs in malignant brain cancer: overcoming IDO. Front Immunol. 2013;4:116.
 
15.  Chang CH, Qiu J, O'Sullivan D, et al. Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell. 2015;162(6):1229-1241.
 
16.  Ohta A, Gorelik E, Prasad SJ, et al. A2A adenosine receptor protects tumors from anti-tumor T cells. Proc Natl Acad Sci U S A. 2006;103(35):13132-13137.
 
17.  Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26-41.
 
18.  Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 2021;221:107753.
 
19.  Sadelain M, Rivière I, Riddell S. Therapeutic T cell engineering. Nature. 2017;545(7655):423-431.
 
20.  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.
 
21.  Ahmed N, Salsman VS, Kew Y, et al. HER2-specific T cells target primary glioblastoma stem cells and induce regression of autologous experimental tumors. Clin Cancer Res. 2010;16(2):474-485.
 
22.  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. Clin Cancer Res. 2015;21(18):4062-4072.
 
23.  Nehama D, Di Ianni N, Musio S, et al. B7-H3-redirected chimeric antigen receptor T cells target glioblastoma and neurospheres. EBioMedicine. 2019;47:33-43.
 
24.  Mount CW, Majzner RG, Sundaresh S, et al. Potent anti-tumor efficacy of anti-GD2 CAR T cells in H3-K27M+ diffuse midline gliomas. Nat Med. 2018;24(5):572-579.
 
25.  Guedan S, Ruella M, June CH. Emerging cellular therapies for cancer. Annu Rev Immunol. 2019;37:145-171.
 
26.  Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448.
 
27.  Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531-2544.
 
28.  Jinek M, Chylinski K, Fonfara I, et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 2012;337(6096):816-821.
 
29.  Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339(6121):819-823.
 
30.  Ran FA, Hsu PD, Wright J, et al. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013;8(11):2281-2308.
 
31.  Rupp LJ, Schumann K, Roybal KT, et al. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci Rep. 2017;7(1):737.
 
32.  Eyquem J, Mansilla-Soto J, Giavridis T, et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017;543(7643):113-117.
 
33.  Roth TL, Puig-Saus C, Yu R, et al. Reprogramming human T cell function and specificity with non-viral genome targeting. Nature. 2018;559(7714):405-409.
 
34.  Ren J, Zhang X, Liu X, et al. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget. 2017;8(10):17002-17011.
 
35.  Choi BD, Yu X, Castano AP, 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.
 
36.  Sade-Feldman M, Yizhak K, Bjorgaard SL, et al. Defining T cell exhaustion in the tumor microenvironment. Cancer Cell. 2018;33(4):683-685.e5.
 
37.  Johnston RJ, Comps-Agrar L, Hackney J, et al. The immunoreceptor TIGIT regulates anti-tumor and antiviral CD8+ T cell effector function. Cancer Cell. 2014;26(6):923-937.
 
38.  Wei J, Luo C, Wang Y, et al. PD-1 silencing impairs the anti-tumor function of chimeric antigen receptor modified T cells by inhibiting proliferation activity. J Immunother Cancer. 2019;7(1):209.
 
39.  Giuffrida L, Sek K, Henderson MA, et al. CRISPR/Cas9 mediated deletion of the adenosine A2A receptor enhances CAR T cell efficacy. Nat Commun. 2021;12(1):3230.
 
40.  Sukumar M, Liu J, Ji Y, et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and anti-tumor function. J Clin Invest. 2013;123(10):4479-4488.
 
41.  Pollizzi KN, Powell JD. Integrating canonical and metabolic signalling programmes in the regulation of T cell responses. Nat Rev Immunol. 2014;14(7):435-446.
 
42.  Jin L, Tao H, Karachi A, et al. CXCR1- or CXCR2-modified CAR T cells co-opt IL-8 for maximal anti-tumor efficacy in solid tumors. Nat Commun. 2019;10(1):4016.
 
43.  Caruana I, Savoldo B, Hoyos V, et al. Heparanase promotes tumor infiltration and anti-tumor activity of CAR-redirected T lymphocytes. Nat Med. 2015;21(5):524-529.
 
44.  Craddock JA, Lu A, Bear A, et al. Enhanced tumor trafficking of GD2 chimeric antigen receptor T cells by expression of the chemokine receptor CCR2b. J Immunother. 2010;33(8):780-788.
 
45.  Di Stasi A, Tey SK, Dotti G, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011;365(18):1673-1683.
 
46.  Roybal KT, Rupp LJ, Morsut L, et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell. 2016;164(4):770-779.
 
47.  Sterner RM, Sakemura R, Cox MJ, et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood. 2019;133(7):697-709.
 
48.  Torikai H, Reik A, Liu PQ, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric antigen-receptor and eliminate expression of endogenous TCR. Blood. 2012;119(24):5697-5705.
 
49.  Ren J, Liu X, Fang C, et al. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin Cancer Res. 2017;23(9):2255-2266.
 
50.  MacLeod DT, Antony J, Martin AJ, et al. Integration of a CD19 CAR into the TCR alpha chain locus streamlines production of allogeneic gene-edited CAR T cells. Mol Ther. 2017;25(4):949-961.
 
51.  Bielamowicz K, Fousek K, Byrd TT, et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro Oncol. 2018;20(4):506-518.
 
52.  Choi BD, Gerstner ER, Frigault MJ, et al. Intraventricular CARv3-TEAM-E T cells in recurrent glioblastoma. N Engl J Med. 2024;390(14):1290-1298.
 
53.  Brown CE, Alizadeh D, Starr R, et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. 2016;375(26):2561-2569.
 
54.  Bagley SJ, Logun M, Fraietta JA, 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.
 
55.  Filley AC, Henriquez M, Dey M. Recurrent glioma clinical trial, CheckMate-143: the game is not over yet. Oncotarget. 2017;8(53):91779-91794.
 
56.  McGranahan N, Furness AJ, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016;351(6280):1463-1469.
 
57.  Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515(7528):568-571.
 
58.  Wang D, Starr R, Chang WC, et al. Chlorotoxin-directed CAR T cells for specific and effective targeting of glioblastoma. Sci Transl Med. 2020;12(533):eaaw2672.
 
59.  Schumann K, Lin S, Boyer E, et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc Natl Acad Sci U S A. 2015;112(33):10437-10442.
 
60.  Kim D, Kim S, Kim S, et al. Genome-wide target specificities of CRISPR-Cas9 nucleases revealed by multiplex Digenome-seq. Genome Res. 2016;26(3):406-415.
 
61.  LaFleur MW, Nguyen TH, Coxe MA, et al. A CRISPR-Cas9 delivery system for in vivo screening of genes in the immune system. Nat Commun. 2019;10(1):1668.
 
62.  Levine BL, Miskin J, Wonnacott K, et al. Global manufacturing of CAR T cell therapy. Mol Ther Methods Clin Dev. 2017;4:92-101.
 
63.  Majzner RG, Mackall CL. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 2018;8(10):1219-1226.
 
64.  Wagner DL, Amini L, Wendering DJ, et al. High prevalence of Streptococcus pyogenes Cas9-reactive T cells within the adult human population. Nat Med. 2019;25(2):242-248.
 
65.  Gattinoni L, Lugli E, Ji Y, et al. A human memory T cell subset with stem cell-like properties. Nat Med. 2011;17(10):1290-1297.
 
66.  Xiao Z, Casey KA, Jameson SC, et al. Programming for CD8 T cell memory development requires IL-12 or type I IFN. J Immunol. 2009;182(5):2786-2794.
 
67.  Stadtmauer EA, Fraietta JA, Davis MM, et al. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020;367(6481):eaba7365.
 
68.  Poirot L, Philip B, Schiffer-Mannioui C, et al. Multiplex genome-edited T-cell manufacturing platform for "off-the-shelf" adoptive T-cell immunotherapies. Cancer Res. 2015;75(18):3853-3864.
 
69.  Henderson GE, Davis AM, King NM, et al. Uncertain benefit: investigators' views and communications in early phase gene transfer trials. Mol Ther. 2004;10(2):225-231.
 
70.  Anzalone AV, Randolph PB, Davis JR, et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. 2019;576(7785):149-157.
 
71.  Gilbert LA, Larson MH, Morsut L, et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell. 2013;154(2):442-451.
 
72.  Wang M, Zuris JA, Meng F, et al. Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles. Proc Natl Acad Sci U S A. 2016;113(11):2868-2873.
 
73.  Mock U, Nickolay L, Philip B, et al. Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS prodigy. Cytotherapy. 2016;18(8):1002-1011.
 
74.  Prinzing B, Zebley CC, Petersen CT, et al. Deleting DNMT3A in CAR T cells prevents exhaustion and enhances anti-tumor activity. Sci Transl Med. 2021;13(620):eabh0272.
 
75.  Weiss T, Puca E, Silginer M, et al. Immunocytokines are a promising immunotherapeutic approach against glioblastoma. Sci Transl Med. 2020;12(564):eabb2311.
 
76.  Sampson JH, Gunn MD, Fecci PE, et al. Brain immunology and immunotherapy in brain tumours. Nat Rev Cancer. 2020;20(1):12-25.
 
77.  Ott PA, Hu Z, Keskin DB, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547(7662):217-221.
 
78.  Dersh D, Holly J, Yewdell JW. A few good peptides: MHC class I-based cancer immunosurveillance and immunoevasion. Nat Rev Immunol. 2021;21(2):116-128.
 
79.  Liu X, Jiang S, Fang C, et al. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res. 2015;75(17):3596-3607.
 
80.  Fisher J, Abramowski P, Wisidagamage Don ND, et al. Avoidance of on-target off-tumor activation using a co-stimulation-only chimeric antigen receptor. Mol Ther. 2017;25(5):1234-1247.
 
81.  de Vries EF. Imaging of T-cell activation in cancer patients: a new approach to monitor immunotherapy. J Nucl Med. 2016;57(12):1851-1853.
 

 
Received: 26 Oct 2025 / Accepted: 18 Nov 2025 / Published (online): 15 Dec 2025 (Europe/Madrid)
 
Citation
 
Guamba Cajas S.E., Paucar Fiallos D.E., Castillo Guevara A.R., Guamba Cajas E.L.
CRISPR-Cas9–Enhanced CAR-T Cell Engineering for Glioblastoma: Mechanisms, Preclinical Advances, and Emerging Clinical Applications.BioNatura Journal. 2025; 2(4): 09. https://doi.org/10.70099/BJ/2025.02.04.09
 
Additional Information
 
Correspondence should be addressed to:
Saúl Emilio Guamba Cajas
Email: saul.guamba@yachaytech.edu.ec
ORCID: https://orcid.org/0009-0008-0518-1994
Tel.: +593 99 895 0919
 
Peer Review Information
 
BioNatura Journal thanks the anonymous reviewers for their valuable contribution to the peer-review process.
Regional peer-review coordination was conducted under the BioNatura Institutional Publishing Consortium (BIPC), involving:
 
·         Universidad Nacional Autónoma de Honduras (UNAH)
 
·         Universidad de Panamá (UP)
 
·         RELATIC (Panama)
 
Reviewer support via Web of Science Reviewer Locator:
https://reviewerlocator.webofscience.com/
 
Publisher Information
 
Published by Clinical Biotec S.L. (Madrid, Spain) as the publisher of record under the BioNatura Institutional Publishing Consortium (BIPC).
 
Institutional co-publishers:
 
·         Universidad Nacional Autónoma de Honduras (UNAH)
 
·         Universidad de Panamá (UP)
 
·         RELATIC (Panama)
 
Places of publication:
Madrid (Spain); Tegucigalpa (Honduras); Panama City (Panama)
 
Online ISSN: 3020-7886
 
Open Access Statement
 
All articles published in BioNatura Journal are freely and permanently available online immediately upon publication, without subscription charges or registration barriers.
 
Publisher's Note
 
BioNatura Journal remains neutral regarding jurisdictional claims in published maps and institutional affiliations.
 
Copyright and License
 
© 2025 by the authors.
This article is published under the terms of the Creative Commons Attribution (CC BY 4.0) license, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
License details: https://creativecommons.org/licenses/by/4.0/
 
Governance
 
For editorial governance and co-publisher responsibilities, see the BIPC Governance Framework (PDF):
https://clinicalbiotec.com/bipc
 
Back to content