We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Applications and current challenges of chimeric antigen receptor T cells in treating high-grade gliomas in adult and pediatric populations

    Nicolina Jovanovich

    Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA

    ,
    Ahmed Habib

    Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA

    Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA

    ,
    NU Farrukh Hameed

    Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA

    Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA

    ,
    Lincoln Edwards

    Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA

    Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA

    &
    Pascal O Zinn

    *Author for correspondence: Tel.: +1 412 647 7614;

    E-mail Address: zinnpo@upmc.edu

    Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA

    Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA

    Published Online:https://doi.org/10.2217/imt-2022-0200

    High-grade gliomas (HGGs) continue to be some of the most devastating diseases in the USA. Despite extensive efforts, the survival of HGG patients has remained relatively stagnant. Chimeric antigen receptor (CAR) T-cell immunotherapy has recently been studied in the context of improving these tumors' clinical outcomes. HGG murine models treated with CAR T cells targeting tumor antigens have shown reduced tumor burden and longer overall survival than models without treatment. Subsequent clinical trials investigating the efficacy of CAR T cells have further shown that this therapy could be safe and might reduce tumor burden. However, there are still many challenges that need to be addressed to optimize the safety and efficacy of CAR T-cell therapy in treating HGG patients.

    Plain language summary

    This publication describes the current application of chimeric antigen T-cell (CAR T-cell) therapy in treating high-grade gliomas (HGGs). Treatment of various HGG models with CAR T cells has shown that this therapy is often able to shrink HGG tumors and prolong the survival of these models. Subsequent clinical trials have shown that CAR T-cell therapy can reduce tumor size in some HGG patients. Patients in these clinical trials have tolerated the treatment well, though more robust studies are needed to confirm this treatment's safety. Additionally, other challenges, such as getting CAR T cells into the brain and to the tumor, need to be addressed to improve the effectiveness of this therapy for HGG patients.

    Tweetable abstract

    Chimeric antigen T-cell therapy has been studied in high-grade glioma models/patients. Studies have confirmed that this therapy may have on-tumor toxicity and an acceptable safety profile in high-grade glioma patients.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Thorbinson C, Kilday JP. Childhood malignant brain tumors: balancing the bench and bedside. Cancers (Basel) 13(23), 6099 (2021).
    • 2. Park JH, De Lomana ALG, Marzese DM et al. A systems approach to brain tumor treatment. Cancers (Basel) 13(13), 3152 (2021).
    • 3. Perrone MG, Ruggiero A, Centonze A, Carrieri A, Ferorelli S, Scilimati A. Diffuse intrinsic pontine glioma (DIPG): breakthrough and clinical perspective. Curr. Med. Chem. 28(17), 3287–3317 (2021).
    • 4. Thomas A, Noël G. Medulloblastoma: optimizing care with a multidisciplinary approach. J. Multidiscip. Healthc. 12, 335–347 (2019).
    • 5. De Vleeschouwer S. Glioblastoma. Codon Publications (2017). https://pubmed.ncbi.nlm.nih.gov/29251853/
    • 6. Johung TB, Monje M. Diffuse intrinsic pontine glioma: new pathophysiological insights and emerging therapeutic targets. Curr. Neuropharmacol. 15(1), 88–97 (2017).
    • 7. Krieger TG, Tirier SM, Park J et al. Modeling glioblastoma invasion using human brain organoids and single-cell transcriptomics. Neuro. Oncol. 22(8), 1138–1149 (2020).
    • 8. Agliardi G, Liuzzi AR, Hotblack A et al. Intratumoral IL-12 delivery empowers CAR-T cell immunotherapy in a pre-clinical model of glioblastoma. Nat. Commun. 12(1), 444 (2021). • This study highlighted the importance of testing therapeutics against more advanced high-grade glioma (HGG) models, as they do not respond to treatment the same as early HGG models, and the exclusive study of early HGG models could lead to efficacy results that cannot be mirrored in patients.
    • 9. Durgin JS, Henderson F, Nasrallah MP et al. Case report: prolonged survival following EGFRvIII CAR T cell treatment for recurrent glioblastoma. Front. Oncol. 11, 669071 (2021).
    • 10. O'Rourke DM, Nasrallah MP, Desai A et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 9(399), eaaa0984 (2017). •• This was the first clinical trial to test EGFRvIII chimeric antigen receptor T cells in patients with residual or recurrent EGFRvIII+ glioblastoma, and the results showed that these chimeric antigen receptor T cells may have some clinical efficacy.
    • 11. Quail DF, Joyce JA. The microenvironmental landscape of brain tumors. Cancer Cell 31(3), 326–341 (2017).
    • 12. Skaga E, Kulesskiy E, Fayzullin A et al. Intertumoral heterogeneity in patient-specific drug sensitivities in treatment-naïve glioblastoma. BMC Cancer 19(1), 628 (2019).
    • 13. Bao Z, Wang Y, Wang Q et al. Intratumor heterogeneity, microenvironment, and mechanisms of drug resistance in glioma recurrence and evolution. Front. Med. 15(4), 551–561 (2021).
    • 14. Petrilli LL, Fuoco C, Palma A et al. Inter and intra-tumor heterogeneity of paediatric type diffuse high-grade gliomas revealed by single-cell mass cytometry. Front. Oncol. 12, 1016343 (2022).
    • 15. Sakthikumar S, Roy A, Haseeb L et al. Whole-genome sequencing of glioblastoma reveals enrichment of non-coding constraint mutations in known and novel genes. Genome Biol. 21(1), 127 (2020).
    • 16. Alharbi M, Mobark N, Bashawri Y et al. Methylation profiling of medulloblastoma in a clinical setting permits sub-classification and reveals new outcome predictions. Front. Neurol. 11, 167 (2020).
    • 17. Kessler T, Berberich A, Casalini B et al. Molecular profiling-based decision for targeted therapies in IDH wild-type glioblastoma. Neurooncol. Adv. 2(1), vdz060 (2020).
    • 18. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc. Natl Acad. Sci. USA 86(24), 10024–10028 (1989).
    • 19. Mohanty R, Chowdhury CR, Arega S, Sen P, Ganguly P, Ganguly N. CAR T cell therapy: a new era for cancer treatment (review). Oncol. Rep. 42(6), 2183–2195 (2019).
    • 20. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 11(4), 69 (2021).
    • 21. Vitale C, Strati P. CAR T-cell therapy for B-cell non-Hodgkin lymphoma and chronic lymphocytic leukemia: clinical trials and real-world experiences. Front. Oncol. 10, 849 (2020).
    • 22. Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat. Rev. Cancer 8(4), 299–308 (2008).
    • 23. Chavez JC, Bachmeier C, Kharfan-Dabaja MA. CAR T-cell therapy for B-cell lymphomas: clinical trial results of available products. Ther. Adv. Hematol. 10, 2040620719841581 (2019). • This clinical trial followed one patient whose tumors became undetectable after IL-13Rα2 chimeric antigen receptor T-cell therapy, highlighting the potential of this treatment.
    • 24. Brown CE, Alizadeh D, Starr R et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N. Engl. J. Med. 375(26), 2561–2569 (2016).
    • 25. Akhavan D, Alizadeh D, Wang D, Weist MR, Shepphird JK, Brown CE. CAR T cells for brain tumors: lessons learned and road ahead. Immunol. Rev. 290(1), 60–84 (2019).
    • 26. Patterson JD, Henson JC, Breese RO, Bielamowicz KJ, Rodriguez A. CAR T cell therapy for pediatric brain tumors. Front. Oncol. 10, 1582 (2020).
    • 27. Stylli SS. Novel treatment strategies for glioblastoma. Cancers (Basel) 12(10), 2883 (2020).
    • 28. Li Y, Wu H, Chen G et al. Arming anti-EGFRvIII CAR-T With TGFβ trap improves antitumor efficacy in glioma mouse models. Front. Oncol. 10, 1117 (2020).
    • 29. Nehama D, Di Ianni N, Musio S et al. B7-H3-redirected chimeric antigen receptor T cells target glioblastoma and neurospheres. EBioMedicine 47, 33–43 (2019).
    • 30. An Z, Aksoy O, Zheng T, Fan QW, Weiss WA. Epidermal growth factor receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies. Oncogene 37(12), 1561–1575 (2018).
    • 31. Keller S, Schmidt MHH. EGFR and EGFRvIII promote angiogenesis and cell invasion in glioblastoma: combination therapies for an effective treatment. Int. J. Mol. Sci. 18(6), 1295 (2017).
    • 32. Van Meter TE, Broaddus WC, Rooprai HK, Pilkington GJ, Fillmore HL. Induction of membrane-type-1 matrix metalloproteinase by epidermal growth factor-mediated signaling in gliomas. Neuro. Oncol. 6(3), 188–199 (2004).
    • 33. Chen XC, Wei XT, Guan JH, Shu H, Chen D. EGF stimulates glioblastoma metastasis by induction of matrix metalloproteinase-9 in an EGFR-dependent mechanism. Oncotarget 8(39), 65969–65982 (2017).
    • 34. Johnson LA, Scholler J, Ohkuri T et al. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci. Transl. Med. 7(275), 275ra222 (2015).
    • 35. Han J, Alvarez-Breckenridge CA, Wang QE, Yu J. TGF-β signaling and its targeting for glioma treatment. Am. J. Cancer Res. 5(3), 945–955 (2015).
    • 36. Thomas DA, Massagué J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell 8(5), 369–380 (2005).
    • 37. 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 7(1), 304 (2019).
    • 38. Xia L, Liu JY, Zheng ZZ et al. BRD4 inhibition boosts the therapeutic effects of epidermal growth factor receptor-targeted chimeric antigen receptor T cells in glioblastoma. Mol. Ther. doi: 10.1016/j.ymthe.2021.05.019 (2021) (Epub ahead of print).
    • 39. Hsi LC, Kundu S, Palomo J et al. Silencing IL-13Rα2 promotes glioblastoma cell death via endogenous signaling. Mol. Cancer Ther. 10(7), 1149–1160 (2011).
    • 40. Arima K, Sato K, Tanaka G et al. Characterization of the interaction between interleukin-13 and interleukin-13 receptors. J. Biol. Chem. 280(26), 24915–24922 (2005).
    • 41. Thaci B, Brown CE, Binello E, Werbaneth K, Sampath P, Sengupta S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro. Oncol. 16(10), 1304–1312 (2014).
    • 42. Brown CE, Warden CD, Starr R et al. Glioma IL13Rα2 is associated with mesenchymal signature gene expression and poor patient prognosis. PLoS One 8(10), e77769 (2013).
    • 43. 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. 21(18), 4062–4072 (2015).
    • 44. Brown CE, Starr R, Aguilar B et al. Stem-like tumor-initiating cells isolated from IL13Rα2 expressing gliomas are targeted and killed by IL13-zetakine-redirected T Cells. Clin. Cancer Res. 18(8), 2199–2209 (2012).
    • 45. Kahlon KS, Brown C, Cooper LJ, Raubitschek A, Forman SJ, Jensen MC. Specific recognition and killing of glioblastoma multiforme by interleukin 13-zetakine redirected cytolytic T cells. Cancer Res. 64(24), 9160–9166 (2004).
    • 46. Krenciute G, Prinzing BL, Yi Z et al. Transgenic expression of IL15 improves antiglioma activity of IL13Rα2-CAR T cells but results in antigen loss variants. Cancer Immunol. Res. 5(7), 571–581 (2017).
    • 47. Santana Carrero RM, Beceren-Braun F, Rivas SC et al. IL-15 is a component of the inflammatory milieu in the tumor microenvironment promoting antitumor responses. Proc. Natl Acad. Sci. U S A 116(2), 599–608 (2019).
    • 48. Nazha B, Inal C, Owonikoko TK. Disialoganglioside GD2 expression in solid tumors and role as a target for cancer therapy. Front. Oncol. 10, 1000 (2020).
    • 49. Golinelli G, Grisendi G, Prapa M et al. Targeting GD2-positive glioblastoma by chimeric antigen receptor empowered mesenchymal progenitors. Cancer Gene Ther. 27(7–8), 558–570 (2020).
    • 50. Murty S, Haile ST, Beinat C et al. Intravital imaging reveals synergistic effect of CAR T-cells and radiation therapy in a preclinical immunocompetent glioblastoma model. Oncoimmunology 9(1), 1757360 (2020).
    • 51. Lemke D, Pfenning PN, Sahm F et al. Costimulatory protein 4IgB7H3 drives the malignant phenotype of glioblastoma by mediating immune escape and invasiveness. Clin. Cancer Res. 18(1), 105–117 (2012).
    • 52. Digregorio M, Coppieters N, Lombard A, Lumapat PN, Scholtes F, Rogister B. The expression of B7-H3 isoforms in newly diagnosed glioblastoma and recurrence and their functional role. Acta Neuropathol. Commun. 9(1), 59 (2021).
    • 53. Zhou Z, Luther N, Ibrahim GM et al. B7-H3, a potential therapeutic target, is expressed in diffuse intrinsic pontine glioma. J. Neurooncol. 111(3), 257–264 (2013).
    • 54. Theruvath J. Checkpoint molecule B7-H3 is highly expressed on medulloblastoma and proves to be a promising candidate for CAR T cell immunotherapy. 19, Iv28–iv29 (2017).
    • 55. Tang X, Zhao S, Zhang Y et al. B7-H3 as a novel CAR-T therapeutic target for glioblastoma. Mol. Ther. Oncolytics 14, 279–287 (2019).
    • 56. Tokunaga R, Zhang W, Naseem M et al. CXCL9, CXCL10, CXCL11/CXCR3 axis for immune activation – a target for novel cancer therapy. Cancer Treat. Rev. 63, 40–47 (2018).
    • 57. Wang G, Zhang Z, Zhong K et al. CXCL11-armed oncolytic adenoviruses enhance CAR-T cell therapeutic efficacy and reprogram tumor microenvironment in glioblastoma. Mol. Ther. 31(1), 134–153 (2023).
    • 58. Debin JA, Maggio JE, Strichartz GR. Purification and characterization of chlorotoxin, a chloride channel ligand from the venom of the scorpion. Am. J. Physiol. 264(2 Pt 1), C361–C369 (1993).
    • 59. Lyons SA, O'Neal J, Sontheimer H. Chlorotoxin, a scorpion-derived peptide, specifically binds to gliomas and tumors of neuroectodermal origin. Glia 39(2), 162–173 (2002).
    • 60. Wang D, Starr R, Chang WC et al. Chlorotoxin-directed CAR T cells for specific and effective targeting of glioblastoma. Sci. Transl. Med. 12(533), (2020).
    • 61. Bielamowicz K, Fousek K, Byrd TT et al. Trivalent CAR T cells overcome interpatient antigenic variability in glioblastoma. Neuro. Oncol. 20(4), 506–518 (2018).
    • 62. Rashed WM, Maher E, Adel M, Saber O, Zaghloul MS. Pediatric diffuse intrinsic pontine glioma: where do we stand? Cancer Metastasis Rev. 38(4), 759–770 (2019).
    • 63. Northcott PA, Robinson GW, Kratz CP et al. Medulloblastoma. Nat. Rev. Dis. Primers 5(1), 11 (2019).
    • 64. Gilbertson RJ, Pearson AD, Perry RH, Jaros E, Kelly PJ. Prognostic significance of the c-erbB-2 oncogene product in childhood medulloblastoma. Br. J. Cancer 71(3), 473–477 (1995).
    • 65. Maachani UB, Tosi U, Pisapia DJ et al. B7-H3 as a prognostic biomarker and therapeutic target in pediatric central nervous system tumors. Transl. Oncol. 13(2), 365–371 (2020).
    • 66. Mount CW, Majzner RG, Sundaresh S et al. Potent antitumor efficacy of anti-GD2 CAR T cells in H3-K27M. Nat. Med. 24(5), 572–579 (2018).
    • 67. De Billy E, Pellegrino M, Orlando D et al. Dual IGF1R/IR inhibitors in combination with GD2-CAR T-cells display a potent anti-tumor activity in diffuse midline glioma H3K27M-mutant. Neuro. Oncol. 24(7), 1150–1163 (2022). •• This study showed that combinatorial therapy of chimeric antigen receptor T cells with an immune lanscape-altering adjuvant increases the efficacy of chimeric antigen receptor T cells at controlling high-grade glioma growth.
    • 68. Majzner RG, Ramakrishna S, Yeom KW et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature 603(7903), 934–941 (2022).
    • 69. Majzner R. GD2 CAR T cells mediate clinical activity and manageable toxicity in children and young adults with DIPG and H3K27M-mutated diffuse midline gliomas. 23(Suppl. 1), i49–i50 (2021).
    • 70. Ahmed N, Ratnayake M, Savoldo B et al. Regression of experimental medulloblastoma following transfer of HER2-specific T cells. Cancer Res. 67(12), 5957–5964 (2007).
    • 71. Nellan A, Rota C, Majzner R et al. Durable regression of medulloblastoma after regional and intravenous delivery of anti-HER2 chimeric antigen receptor T cells. J. Immunother. Cancer 6(1), 30 (2018).
    • 72. Vitanza NA, Johnson AJ, Wilson AL et al. Locoregional infusion of HER2-specific CAR T cells in children and young adults with recurrent or refractory CNS tumors: an interim analysis. Nat. Med. doi: 10.1038/s41591-021-01404-8 (2021) (Epub ahead of print).
    • 73. Majzner RG, Theruvath JL, Nellan A et al. CAR T cells targeting B7-H3, a pan-cancer antigen, demonstrate potent preclinical activity against pediatric solid tumors and brain tumors. Clin. Cancer Res. 25(8), 2560–2574 (2019).
    • 74. Donovan LK, Delaidelli A, Joseph SK et al. Locoregional delivery of CAR T cells to the cerebrospinal fluid for treatment of metastatic medulloblastoma and ependymoma. Nat. Med. 26(5), 720–731 (2020). • This study showed that unlike in glioblastoma, trivalent chimeric antigen receptor T cell (TRI-CAR T-cell) therapy was not as effective as singular chimeric antigen receptor T-cell (mono-CAR T-cell) therapy in shrinking pediatric high-grade gliomas, highlighting the heterogeneous nature of treatment responses between tumors.
    • 75. Brantley-Sieders DM, Fang WB, Hwang Y, Hicks D, Chen J. Ephrin-A1 facilitates mammary tumor metastasis through an angiogenesis-dependent mechanism mediated by EphA receptor and vascular endothelial growth factor in mice. Cancer Res. 66(21), 10315–10324 (2006).
    • 76. Ogawa K, Pasqualini R, Lindberg RA, Kain R, Freeman AL, Pasquale EB. The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene 19(52), 6043–6052 (2000).
    • 77. Miao H, Li DQ, Mukherjee A et al. EphA2 mediates ligand-dependent inhibition and ligand-independent promotion of cell migration and invasion via a reciprocal regulatory loop with Akt. Cancer Cell 16(1), 9–20 (2009).
    • 78. Sikkema AH, Den Dunnen WF, Hulleman E et al. EphB2 activity plays a pivotal role in pediatric medulloblastoma cell adhesion and invasion. Neuro. Oncol. 14(9), 1125–1135 (2012).
    • 79. Parker NR, Khong P, Parkinson JF, Howell VM, Wheeler HR. Molecular heterogeneity in glioblastoma: potential clinical implications. Front. Oncol. 5, 55 (2015).
    • 80. Daneman R, Prat A. The blood–brain barrier. Cold Spring Harb. Perspect. Biol. 7(1), a020412 (2015).
    • 81. Chuntova P, Downey KM, Hegde B, Almeida ND, Okada H. Genetically engineered T-cells for malignant glioma: overcoming the barriers to effective immunotherapy. Front. Immunol. 9, 3062 (2018).
    • 82. Truffaux N, Philippe C, Paulsson J et al. Preclinical evaluation of dasatinib alone and in combination with cabozantinib for the treatment of diffuse intrinsic pontine glioma. Neuro. Oncol. 17(7), 953–964 (2015).
    • 83. Lin YJ, Mashouf LA, Lim M. CAR T cell therapy in primary brain tumors: current investigations and the future. Front. Immunol. 13, 817296 (2022).
    • 84. Marin BM, Porath KA, Jain S et al. Heterogeneous delivery across the blood–brain barrier limits the efficacy of an EGFR-targeting antibody drug conjugate in glioblastoma. Neuro. Oncol. 23(12), 2042–2053 (2021).
    • 85. Abramson JS, McGree B, Noyes S et al. Anti-CD19 CAR T cells in CNS diffuse large-B-cell lymphoma. N. Engl. J. Med. 377(8), 783–784 (2017).
    • 86. Sarkaria JN, Hu LS, Parney IF et al. Is the blood–brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro. Oncol. 20(2), 184–191 (2018).
    • 87. Ung C, Upton DH, Venkat P et al. DIPG-21. DIPG cells alter the permeability of the blood–brain barrier in the brainstem leading to treatment failure. Neuro. Oncol. 24(Suppl. 1), i22 (2022).
    • 88. Amankulor NM, Kim Y, Arora S et al. Mutant IDH1 regulates the tumor-associated immune system in gliomas. Genes Dev. 31(8), 774–786 (2017).
    • 89. Mehta AM, Sonabend AM, Bruce JN. Convection-enhanced delivery. Neurotherapeutics 14(2), 358–371 (2017).
    • 90. Souweidane MM, Kramer K, Pandit-Taskar N et al. Convection-enhanced delivery for diffuse intrinsic pontine glioma: a single-centre, dose-escalation, phase 1 trial. Lancet Oncol. 19(8), 1040–1050 (2018).
    • 91. Ishida J, Alli S, Bondoc A et al. MRI-guided focused ultrasound enhances drug delivery in experimental diffuse intrinsic pontine glioma. J. Control. Rel. 330, 1034–1045 (2021).
    • 92. Chen KT, Chai WY, Lin YJ et al. Neuronavigation-guided focused ultrasound for transcranial blood–brain barrier opening and immunostimulation in brain tumors. Sci. Adv. 7(6), eabd0772 (2021).
    • 93. Decordova S, Shastri A, Tsolaki AG et al. Molecular heterogeneity and immunosuppressive microenvironment in glioblastoma. Front. Immunol. 11, 1402 (2020).
    • 94. Gargett T, Ebert LM, Truong NTH et al. GD2-targeting CAR-T cells enhanced by transgenic IL-15 expression are an effective and clinically feasible therapy for glioblastoma. J. Immunother. Cancer 10(9), e005187 (2022).
    • 95. Ahmed N, Gandhi D, Melhem ER, Frenkel V. MRI guided focused ultrasound-mediated delivery of therapeutic cells to the brain: a review of the state-of-the-art methodology and future applications. Front. Neurol. 12, 669449 (2021).
    • 96. Atik AF, Suryadevara CM, Schweller RM et al. Hyaluronic acid based low viscosity hydrogel as a novel carrier for convection enhanced delivery of CAR T cells. J. Clin. Neurosci. 56, 163–168 (2018).
    • 97. Du M, Hari P, Hu Y, Mei H. Biomarkers in individualized management of chimeric antigen receptor T cell therapy. Biomark. Res. 8, 13 (2020).
    • 98. Mirzaei HR, Mirzaei H, Namdar A, Rahmati M, Till BG, Hadjati J. Predictive and therapeutic biomarkers in chimeric antigen receptor T-cell therapy: a clinical perspective. J. Cell. Physiol. 234(5), 5827–5841 (2019).