CAR-T Cell Therapy for Cancer

Authors

DOI:

https://doi.org/10.54133/ajms.v6i2.726

Keywords:

CAR-T cells, Chimeric antigen receptor, Immune cell therapy

Abstract

Objective: To provide a basic overview of the status of CAR-T cell therapy and foresee its future applicability in cancer treatment. Method: The search engines PubMed, Google Scholar, ResearchGate and Web of Science were employed in obtaining peer-reviewed articles using the criteria outlined in the method section. Main points: CAR-T cell therapy has proved a lifesaving option for hematological malignancies despite its huge cost per treatment. Clinical trials are still ongoing to improve the effectiveness of this therapy for solid tumors as well as make it more affordable and easier to set up. Conclusion: CAR-T cell therapy represents a useful addition to the arsenal in the fight against cancer, particularly in lifesaving scenarios in dealing with serious hematological malignancies.

Downloads

Download data is not yet available.

References

Oiseth SJ, Aziz MS. Cancer immunotherapy: a brief review of the history, possibilities, and challenges ahead. J Cancer Metastasis Treat. 2017;3:250-261. doi: 10.20517/2394-4722.2017.41.

Decker WK, Safdar A. Bioimmunoadjuvants for the treatment of neoplastic and infectious disease: Coley's legacy revisited. Cytok Growth Fact Rev. 2009;20(4):271-281. doi: 10.1016/j.cytogfr.2009.07.004.

Decker WK, da Silva RF, Sanabria MH, Angelo LS, Guimarães F, Burt BM, et al. Cancer Immunotherapy: Historical Perspective of a Clinical Revolution and Emerging Preclinical Animal Models. Front Immunol. 2017;8:829. doi: 10.3389/fimmu.2017.00829.

Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20(11):651-668. doi: 10.1038/s41577-020-0306-5.

Wu HC, Chang DK, Huang CT. Targeted therapy for cancer. J Cancer Mol. 2006;2(2):57-66.

Arruebo M, Vilaboa N, Sáez-Gutierrez B, Lambea J, Tres A, Valladares M, et al. Assessment of the evolution of cancer treatment therapies. Cancers (Basel). 2011;3(3):3279-3330. doi: 10.3390/cancers3033279.

Li Y, Ayala-Orozco C, Rauta PR, Krishnan S. The application of nanotechnology in enhancing immunotherapy for cancer treatment: current effects and perspective. Nanoscale. 2019;11(37):17157-17178. doi: 10.1039/c9nr05371a.

Al-Janabi I. Response challenges to cancer immunotherapies. Al-Rafidain J Med Sci. 2022;2:51-80. doi: 10.54133/ajms.v2i.65.

Shafer P, Kelly LM, Hoyos V. Cancer therapy with TCR-engineered T cells: Current strategies, challenges, and prospects. Front Immunol. 2022;13:835762. doi: 10.3389/fimmu.2022.835762.

Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 1986;233(4770):1318-1321. doi: 10.1126/science.3489291.

Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988;319(25):1676-1680. doi: 10.1056/NEJM198812223192527.

Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin Cancer Res. 2011;17(13):4550-4557. doi: 10.1158/1078-0432.CCR-11-0116.

Goff SL, Dudley ME, Citrin DE, Somerville RP, Wunderlich JR, Danforth DN, et al. Randomized, prospective evaluation comparing intensity of lymphodepletion before adoptive transfer of tumor-infiltrating lymphocytes for patients with metastatic melanoma. J Clin Oncol. 2016;34(20):2389-2397. doi: 10.1200/JCO.2016.66.7220.

Kumar BV, Connors TJ, Farber DL. Human T cell development, localization, and function throughout Life. Immunity. 2018;48(2):202-213. doi: 10.1016/j.immuni.2018.01.007.

van den Broek T, Borghans JAM, van Wijk F. The full spectrum of human naive T cells. Nat Rev Immunol. 2018;18(6):363-373. doi: 10.1038/s41577-018-0001-y.

Vignali DA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008;8(7):523-532. doi: 10.1038/nri2343.

Levine BL, Miskin J, Wonnacott K, Keir C. Global manufacturing of CAR T cell therapy. Mol Ther Methods Clin Dev. 2016;4:92-101. doi: 10.1016/j.omtm.2016.12.006.

Labanieh L, Majzner RG, Mackall CL. Programming CAR-T cells to kill cancer. Nat Biomed Eng. 2018;2(6):377-391. doi: 10.1038/s41551-018-0235-9.

Ramos CA, Dotti G. Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer therapy. Expert Opin Biol Ther. 2011;11(7):855-873. doi: 10.1517/14712598.2011.573476.

Lenschow DJ, Walunas TL, Bluestone JA. CD28/B7 system of T cell costimulation. Annu Rev Immunol. 1996;14:233-258. doi: 10.1146/annurev.immunol.14.1.233.

Mitra A, Barua A, Huang L, Ganguly S, Feng Q, He B. From bench to bedside: the history and progress of CAR T cell therapy. Front Immunol. 2023;14:1188049. doi: 10.3389/fimmu.2023.1188049.

Stinchcombe JC, Majorovits E, Bossi G, Fuller S, Griffiths GM. Centrosome polarization delivers secretory granules to the immunological synapse. Nature. 2006;443(7110):462-465. doi: 10.1038/nature05071.

Martínez-Lostao L, Anel A, Pardo J. How do cytotoxic lymphocytes kill cancer cells? Clin Cancer Res. 2015;21(22):5047-5056. doi: 10.1158/1078-0432.CCR-15-0685.

Dufva O, Koski J, Maliniemi P, Ianevski A, Klievink J, Leitner J, et al. Integrated drug profiling and CRISPR screening identify essential pathways for CAR T-cell cytotoxicity. Blood. 2020;135(9):597-609. doi: 10.1182/blood.2019002121.

Ivica NA, Young CM. Tracking the CAR-T revolution: Analysis of clinical trials of CAR-T and TCR-T therapies for the treatment of cancer (1997-2020). Healthcare (Basel). 2021;9(8):1062. doi: 10.3390/healthcare9081062.

Zhang C, Liu J, Zhong JF, Zhang X. Engineering CAR-T cells. Biomark Res. 2017;5:22. doi: 10.1186/s40364-017-0102-y.

Kuwana Y, Asakura Y, Utsunomiya N, Nakanishi M, Arata Y, Itoh S, et al. Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions. Biochem Biophys Res Commun. 1987;149(3):960-968. doi: 10.1016/0006-291x(87)90502-x.

Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A. 1993;90(2):720-724. doi: 10.1073/pnas.90.2.720.

Bird RE, Hardman KD, Jacobson JW, Johnson S, Kaufman BM, Lee SM, et al. Single-chain antigen-binding proteins. Science. 1988;242(4877):423-426. doi: 10.1126/science.3140379.

Huston JS, Levinson D, Mudgett-Hunter M, Tai MS, Novotný J, Margolies MN, et al. Protein engineering of antibody binding sites: recovery of specific activity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli. Proc Natl Acad Sci U S A. 1988;85(16):5879-83. doi: 10.1073/pnas.85.16.5879.

Fesnak AD, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer. 2016;16(9):566-581. doi: 10.1038/nrc.2016.97.

Benmebarek MR, Karches CH, Cadilha BL, Lesch S, Endres S, Kobold S. Killing mechanisms of chimeric antigen receptor (CAR) T cells. Int J Mol Sci. 2019;20(6):1283. doi: 10.3390/ijms20061283.

Guest RD, Hawkins RE, Kirillova N, Cheadle EJ, Arnold J, O'Neill A, et al. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J Immunother. 2005;28(3):203-211. doi: 10.1097/01.cji.0000161397.96582.59.

Hombach AA, Schildgen V, Heuser C, Finnern R, Gilham DE, Abken H. T cell activation by antibody-like immunoreceptors: the position of the binding epitope within the target molecule determines the efficiency of activation of redirected T cells. J Immunol. 2007;178(7):4650-4657. doi: 10.4049/jimmunol.178.7.4650.

James SE, Greenberg PD, Jensen MC, Lin Y, Wang J, Till BG, et al. Antigen sensitivity of CD22-specific chimeric TCR is modulated by target epitope distance from the cell membrane. J Immunol. 2008;180(10):7028-7038. doi: 10.4049/jimmunol.180.10.7028.

Wilkie S, Picco G, Foster J, Davies DM, Julien S, Cooper L, et al. Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor. J Immunol. 2008;180(7):4901-4909. doi: 10.4049/jimmunol.180.7.4901.

De Marco RC, Monzo HJ, Ojala PM. CAR T cell therapy: A versatile living drug. Int J Mol Sci. 2023;24(7):6300. doi: 10.3390/ijms24076300.

Cappell KM, Kochenderfer JN. Long-term outcomes following CAR T cell therapy: what we know so far. Nat Rev Clin Oncol. 2023;20(6):359-371. doi: 10.1038/s41571-023-00754-1.

Drake CG, Jaffee E, Pardoll DM. Mechanisms of immune evasion by tumors. Adv Immunol. 2006;90:51-81. doi: 10.1016/S0065-2776(06)90002-9.

Finney HM, Lawson AD, Bebbington CR, Weir AN. Chimeric receptors providing both primary and costimulatory signaling in T cells from a single gene product. J Immunol. 1998;161(6):2791-2797.

Krause A, Guo HF, Latouche JB, Tan C, Cheung NK, Sadelain M. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J Exp Med. 1998;188(4):619-626. doi: 10.1084/jem.188.4.619.

Hombach A, Wieczarkowiecz A, Marquardt T, Heuser C, Usai L, Pohl C, et al. Tumor-specific T cell activation by recombinant immunoreceptors: CD3 zeta signaling and CD28 costimulation are simultaneously required for efficient IL-2 secretion and can be integrated into one combined CD28/CD3 zeta signaling receptor molecule. J Immunol. 2001;167(11):6123-6131. doi: 10.4049/jimmunol.167.11.6123.

Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Biotechnol. 2002;20(1):70-75. doi: 10.1038/nbt0102-70.

Imai C, Mihara K, Andreansky M, Nicholson IC, Pui CH, Geiger TL, et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia. 2004;18(4):676-684. doi: 10.1038/sj.leu.2403302.

Wang J, Jensen M, Lin Y, Sui X, Chen E, Lindgren CG, et al. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains. Hum Gene Ther. 2007;18(8):712-725. doi: 10.1089/hum.2007.028.

Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, Suhoski MM, et al. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci U S A. 2009;106(9):3360-3365. doi: 10.1073/pnas.0813101106.

Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther. 2009;17(8):1453-1464. doi: 10.1038/mt.2009.83.

Yvon E, Del Vecchio M, Savoldo B, Hoyos V, Dutour A, Anichini A, et al. Immunotherapy of metastatic melanoma using genetically engineered GD2-specific T cells. Clin Cancer Res. 2009;15(18):5852-5860. doi: 10.1158/1078-0432.CCR-08-3163.

Zhao Y, Wang QJ, Yang S, Kochenderfer JN, Zheng Z, Zhong X, et al. A herceptin-based chimeric antigen receptor with modified signaling domains leads to enhanced survival of transduced T lymphocytes and antitumor activity. J Immunol. 2009;183(9):5563-5574. doi: 10.4049/jimmunol.0900447.

Finney HM, Akbar AN, Lawson AD. Activation of resting human primary T cells with chimeric receptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series with signals from the TCR zeta chain. J Immunol. 2004;172(1):104-113. doi: 10.4049/jimmunol.172.1.104.

Brentjens RJ, Santos E, Nikhamin Y, Yeh R, Matsushita M, La Perle K, et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin Cancer Res. 2007;13(18 Pt 1):5426-5435. doi: 10.1158/1078-0432.CCR-07-0674.

Wang J, Press OW, Lindgren CG, Greenberg P, Riddell S, Qian X, et al. Cellular immunotherapy for follicular lymphoma using genetically modified CD20-specific CD8+ cytotoxic T lymphocytes. Mol Ther. 2004;9(4):577-586. doi: 10.1016/j.ymthe.2003.12.011.

Zhong XS, Matsushita M, Plotkin J, Riviere I, Sadelain M. Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+ T cell-mediated tumor eradication. Mol Ther. 2010;18(2):413-420. doi: 10.1038/mt.2009.210.

Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18(4):843-851. doi: 10.1038/mt.2010.24.

Chmielewski M, Abken H. TRUCKs: the fourth generation of CARs. Expert Opin Biol Ther. 2015;15(8):1145-1154. doi: 10.1517/14712598.2015.1046430.

Maus MV, June CH. Making better chimeric antigen receptors for adoptive T-cell therapy. Clin Cancer Res. 2016;22(8):1875-1884. doi: 10.1158/1078-0432.CCR-15-1433.

Yu S, Li A, Liu Q, Li T, Yuan X, Han X, et al. Chimeric antigen receptor T cells: a novel therapy for solid tumors. J Hematol Oncol. 2017;10(1):78. doi: 10.1186/s13045-017-0444-9.

Kagoya Y, Tanaka S, Guo T, Anczurowski M, Wang CH, Saso K, et al. A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects. Nat Med. 2018;24(3):352-359. doi: 10.1038/nm.4478.

Hosseinkhani N, Derakhshani A, Kooshkaki O, Abdoli Shadbad M, Hajiasgharzadeh K, Baghbanzadeh A, et al. Immune Checkpoints and CAR-T Cells: The Pioneers in Future Cancer Therapies? Int J Mol Sci. 2020;21(21):8305. doi: 10.3390/ijms21218305.

Pang Y, Hou X, Yang C, Liu Y, Jiang G. Advances on chimeric antigen receptor-modified T-cell therapy for oncotherapy. Mol Cancer. 2018;17(1):91. doi: 10.1186/s12943-018-0840-y.

Dabas P, Danda A. Revolutionizing cancer treatment: a comprehensive review of CAR-T cell therapy. Med Oncol. 2023;40(9):275. doi: 10.1007/s12032-023-02146-y.

Schwella N, Braun A, Ahrens N, Rick O, Salama A. Leukapheresis after high-dose chemotherapy and autologous peripheral blood progenitor cell transplantation: a novel approach to harvest a second autograft. Transfusion. 2003;43(2):259-264. doi: 10.1046/j.1537-2995.2003.00306.x.

Alcoforado M. Leukapheresis: A Therapeutic Procedure for Blood Disorders. Med Case Rep. 2023;9(6):308.

Casati A, Varghaei-Nahvi A, Feldman SA, Assenmacher M, Rosenberg SA, Dudley ME, et al. Clinical-scale selection and viral transduction of human naïve and central memory CD8+ T cells for adoptive cell therapy of cancer patients. Cancer Immunol Immunother. 2013;62(10):1563-1573. doi: 10.1007/s00262-013-1459-x.

Watanabe N, Mo F, McKenna MK. Impact of manufacturing procedures on CAR T cell functionality. Front Immunol. 2022;13:876339. doi: 10.3389/fimmu.2022.876339.

Riet T, Holzinger A, Dörrie J, Schaft N, Schuler G, Abken H. Nonviral RNA transfection to transiently modify T cells with chimeric antigen receptors for adoptive therapy. Methods Mol Biol. 2013;969:187-201. doi: 10.1007/978-1-62703-260-5_12.

Hollingsworth RE, Jansen K. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines. 2019;4:7. doi: 10.1038/s41541-019-0103-y.

Kraus MH, Popescu NC, Amsbaugh SC, King CR. Overexpression of the EGF receptor-related proto-oncogene erbB-2 in human mammary tumor cell lines by different molecular mechanisms. EMBO J. 1987;6(3):605-610. doi: 10.1002/j.1460-2075.1987.tb04797.x.

Pils D, Pinter A, Reibenwein J, Alfanz A, Horak P, Schmid BC, et al. In ovarian cancer the prognostic influence of HER2/neu is not dependent on the CXCR4/SDF-1 signalling pathway. Br J Cancer. 2007;96(3):485-4891. doi: 10.1038/sj.bjc.6603581.

Feola S, Chiaro J, Martins B, Cerullo V. Uncovering the tumor antigen landscape: What to know about the discovery process. Cancers (Basel). 2020;12(6):1660. doi: 10.3390/cancers12061660.

Fratta E, Coral S, Covre A, Parisi G, Colizzi F, Danielli R, et al. The biology of cancer testis antigens: putative function, regulation and therapeutic potential. Mol Oncol. 2011;5(2):164-182. doi: 10.1016/j.molonc.2011.02.001.

Nin DS, Deng LW. Biology of cancer-testis antigens and their therapeutic implications in cancer. Cells. 2023;12(6):926. doi: 10.3390/cells12060926.

Van den Eynde BJ, van der Bruggen P. T cell defined tumor antigens. Curr Opin Immunol. 1997;9(5):684-693. doi: 10.1016/s0952-7915(97)80050-7.

Robbins PF, El-Gamil M, Li YF, Kawakami Y, Loftus D, Appella E, et al. A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med. 1996;183(3):1185-1192. doi: 10.1084/jem.183.3.1185.

Vigneron N. Human tumor antigens and cancer immunotherapy. Biomed Res Int. 2015;2015:948501. doi: 10.1155/2015/948501.

Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348(6230):69-74. doi: 10.1126/science.aaa4971.

Cerezo-Wallis D, Soengas MS. Understanding tumor-antigen presentation in the new era of cancer immunotherapy. Curr Pharm Des. 2016;22(41):6234-6250. doi: 10.2174/1381612822666160826111041.

Miller BC, Maus MV. CD19-targeted CAR T cells: A new tool in the fight against B cell malignancies. Oncol Res Treat. 2015;38(12):683-690. doi: 10.1159/000442170.

June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361-1365. doi: 10.1126/science.aar6711.

Holling TM, Schooten E, van Den Elsen PJ. Function and regulation of MHC class II molecules in T-lymphocytes: of mice and men. Hum Immunol. 2004;65(4):282-290. doi: 10.1016/j.humimm.2004.01.005.

Hewitt EW. The MHC class I antigen presentation pathway: strategies for viral immune evasion. Immunology. 2003;110(2):163-169. doi: 10.1046/j.1365-2567.2003.01738.x.

Neefjes J, Jongsma ML, Paul P, Bakke O. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol. 2011;11(12):823-836. doi: 10.1038/nri3084.

Hicklin DJ, Marincola FM, Ferrone S. HLA class I antigen downregulation in human cancers: T-cell immunotherapy revives an old story. Mol Med Today. 1999;5(4):178-186. doi: 10.1016/s1357-4310(99)01451-3.

Brady MS, Eckels DD, Ree SY, Schultheiss KE, Lee JS. MHC class II-mediated antigen presentation by melanoma cells. J Immunother Emphasis Tumor Immunol. 1996;19(6):387-397. doi: 10.1097/00002371-199611000-00001. PMID: 9041456.

Akahori Y, Wang L, Yoneyama M, Seo N, Okumura S, Miyahara Y, et al. Antitumor activity of CAR-T cells targeting the intracellular oncoprotein WT1 can be enhanced by vaccination. Blood. 2018;132(11):1134-1145. doi: 10.1182/blood-2017-08-802926.

Klebanoff CA, Rosenberg SA, Restifo NP. Prospects for gene-engineered T cell immunotherapy for solid cancers. Nat Med. 2016 ;22(1):26-36. doi: 10.1038/nm.4015.

Li J, Li W, Huang K, Zhang Y, Kupfer G, Zhao Q. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: lessons learned and strategies for moving forward. J Hematol Oncol. 2018;11(1):22. doi: 10.1186/s13045-018-0568-6.

Metzinger MN, Verghese C, Hamouda DM, Lenhard A, Choucair K, Senzer N, et al. Chimeric antigen receptor T-Cell therapy: Reach to solid tumor experience. Oncology. 2019;97(2):59-74. doi: 10.1159/000500488.

Katz SC, Burga RA, McCormack E, Wang LJ, Mooring W, Point GR, et al. Phase I hepatic immunotherapy for metastases study of intra-arterial chimeric antigen receptor-modified T-cell therapy for CEA+ liver metastases. Clin Cancer Res. 2015;21(14):3149-3159. doi: 10.1158/1078-0432.CCR-14-1421.

Thistlethwaite FC, Gilham DE, Guest RD, Rothwell DG, Pillai M, Burt DJ, et al. The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer Immunol Immunother. 2017;66(11):1425-1436. doi: 10.1007/s00262-017-2034-7.

Zhang C, Wang Z, Yang Z, Wang M, Li S, Li Y, et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA+ metastatic colorectal cancers. Mol Ther. 2017;25(5):1248-1258. doi: 10.1016/j.ymthe.2017.03.010.

Ahmed N, Brawley V, Hegde M, Bielamowicz K, Wakefield A, Ghazi A, et al. Autologous HER2 CMV bispecific CAR T cells are safe and demonstrate clinical benefit for glioblastoma in a Phase I trial. J Immunother Cancer. 2015;3(Suppl 2):O11. doi: 10.1186/2051-1426-3-S2-O11.

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. doi: 10.1001/jamaoncol.2017.0184.

Feng KC, Guo YL, Liu Y, Dai HR, Wang Y, Lv HY, et al. Cocktail treatment with EGFR-specific and CD133-specific chimeric antigen receptor-modified T cells in a patient with advanced cholangiocarcinoma. J Hematol Oncol. 2017;10(1):4. doi: 10.1186/s13045-016-0378-7.

Brown CE, Badie B, Barish ME, Weng L, Ostberg JR, Chang WC, 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. doi: 10.1158/1078-0432.CCR-15-0428.

O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 2017;9(399):eaaa0984. doi: 10.1126/scitranslmed.aaa0984.

Junghans RP, Ma Q, Rathore R, Gomes EM, Bais AJ, Lo AS, et al. Phase I trial of anti-PSMA designer CAR-T cells in prostate cancer: Possible role for interacting interleukin 2-T cell pharmacodynamics as a determinant of clinical response. Prostate. 2016;76(14):1257-1270. doi: 10.1002/pros.23214.

Stroncek DF, Lee DW, Ren J, Sabatino M, Highfill S, Khuu H, et al. Elutriated lymphocytes for manufacturing chimeric antigen receptor T cells. J Transl Med. 2017;15(1):59. doi: 10.1186/s12967-017-1160-5.

Beatty GL, O'Hara MH, Nelson AM, McGarvey M, Torigian DA, Lacey SF, et al. Safety and antitumor activity of chimeric antigen receptor modified T cells in patients with chemotherapy refractory metastatic pancreatic cancer. J Clin Oncol. 2015;33(15):3007. doi: 10.1200/jco.2015.33.15_suppl.3007.

Ye L, Lou Y, Lu L, Fan X. Mesothelin-targeted second generation CAR-T cells inhibit growth of mesothelin-expressing tumors in vivo. Exp Ther Med. 2019;17(1):739-747. doi: 10.3892/etm.2018.7015.

Jirapongwattana N, Thongchot S, Chiraphapphaiboon W, Chieochansin T, Sa-Nguanraksa D, Warnnissorn M, et al. Mesothelin‑specific T cell cytotoxicity against triple negative breast cancer is enhanced by 40s ribosomal protein subunit 3‑treated self‑differentiated dendritic cells. Oncol Rep. 2022;48(1):127. doi: 10.3892/or.2022.8338.

Wang K, Wei G, Liu D. CD19: a biomarker for B cell development, lymphoma diagnosis and therapy. Exp Hematol Oncol. 2012;1(1):36. doi: 10.1186/2162-3619-1-36.

Yang J, Zhou W, Li D, Niu T, Wang W. BCMA-targeting chimeric antigen receptor T-cell therapy for multiple myeloma. Cancer Lett. 2023;553:215949. doi: 10.1016/j.canlet.2022.215949.

Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448. doi: 10.1056/NEJMoa1709866.

Locke FL, Ghobadi A, Jacobson CA, Miklos DB, Lekakis LJ, Oluwole OO, et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol. 2019;20(1):31-42. doi: 10.1016/S1470-2045(18)30864-7.

Abramson JS, Palomba ML, Gordon LI, Lunning MA, Wang M, Arnason J, et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet. 2020;396(10254):839-852. doi: 10.1016/S0140-6736(20)31366-0.

Wang M, Munoz J, Goy A, Locke FL, Jacobson CA, Hill BT, et al. KTE-X19 CAR T-cell therapy in relapsed or refractory Mantle-cell lymphoma. N Engl J Med. 2020;382(14):1331-1342. doi: 10.1056/NEJMoa1914347.

Munshi NC, Anderson LD, Shah N, Madduri D, Berdeja J, Lonial S, et al. Idecabtagene Vicleucel in relapsed and refractory multiple myeloma. N Engl J Med. 2021;384(8):705-716. doi: 10.1056/NEJMoa2024850.

Martin T, Usmani SZ, Berdeja JG, Agha M, Cohen AD, Hari P, et al. Ciltacabtagene autoleucel, an anti-B-cell maturation antigen chimeric antigen receptor T-cell therapy, for relapsed/refractory multiple myeloma: CARTITUDE-1 2-year follow-up. J Clin Oncol. 2023;41(6):1265-1274. doi: 10.1200/JCO.22.00842.

Cooper ML, Choi J, Staser K, Ritchey JK, Devenport JM, Eckardt K, et al. An "off-the-shelf" fratricide-resistant CAR-T for the treatment of T cell hematologic malignancies. Leukemia. 2018;32(9):1970-1983. doi: 10.1038/s41375-018-0065-5.

Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. 'Off-the-shelf' allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov. 2020;19(3):185-199. doi: 10.1038/s41573-019-0051-2.

Morgan MA, Büning H, Sauer M, Schambach A. Use of cell and genome modification technologies to generate improved "off-the-shelf" CAR T and CAR NK cells. Front Immunol. 2020;11:1965. doi: 10.3389/fimmu.2020.01965.

Kagoya Y, Guo T, Yeung B, Saso K, Anczurowski M, Wang CH, et al. Genetic ablation of HLA class I, class II, and the T-cell receptor enables allogeneic T cells to be used for adoptive T-cell therapy. Cancer Immunol Res. 2020;8(7):926-936. doi: 10.1158/2326-6066.CIR-18-0508.

Mailankody S, Matous JV, Chhabra S, Liedtke M, Sidana S, Oluwole OO, et al. Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: phase 1 UNIVERSAL trial interim results. Nat Med. 2023;29(2):422-429. doi: 10.1038/s41591-022-02182-7.

Rurik JG, Tombácz I, Yadegari A, Méndez Fernández PO, Shewale SV, Li L, et al. CAR T cells produced in vivo to treat cardiac injury. Science. 2022;375(6576):91-96. doi: 10.1126/science.abm0594.

Mei H, Li C, Jiang H, Zhao X, Huang Z, Jin D, et al. A bispecific CAR-T cell therapy targeting BCMA and CD38 in relapsed or refractory multiple myeloma. J Hematol Oncol. 2021;14(1):161. doi: 10.1186/s13045-021-01170-7.

Zeng W, Zhang Q, Zhu Y, Ou R, Peng L, Wang B, et al. Engineering novel CD19/CD22 dual-target CAR-T cells for improved anti-tumor activity. Cancer Invest. 2022;40(3):282-292. doi: 10.1080/07357907.2021.2005798.

van der Schans JJ, van de Donk NWCJ, Mutis T. Dual targeting to overcome current challenges in multiple myeloma CAR T-cell treatment. Front Oncol. 2020;10:1362. doi: 10.3389/fonc.2020.01362.

Faust JR, Hamill D, Kolb EA, Gopalakrishnapillai A, Barwe SP. Mesothelin: An immunotherapeutic target beyond solid tumors. Cancers (Basel). 2022;14(6):1550. doi: 10.3390/cancers14061550.

Patel U, Abernathy J, Savani BN, Oluwole O, Sengsayadeth S, Dholaria B. CAR T cell therapy in solid tumors: A review of current clinical trials. EJHaem. 2021;3(Suppl 1):24-31. doi: 10.1002/jha2.356.

Adusumilli PS, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J, et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med. 2014;6(261):261ra151. doi: 10.1126/scitranslmed.3010162.

Rupp LJ, Schumann K, Roybal KT, Gate RE, Ye CJ, Lim WA, 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. doi: 10.1038/s41598-017-00462-8.

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. doi: 10.1186/s40425-019-0806-7.

Ajina A, Maher J. Prospects for combined use of oncolytic viruses and CAR T-cells. J Immunother Cancer. 2017;5(1):90. doi: 10.1186/s40425-017-0294-6.

Grosser R, Cherkassky L, Chintala N, Adusumilli PS. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell. 2019;36(5):471-482. doi: 10.1016/j.ccell.2019.09.006.

Donnadieu E, Dupré L, Pinho LG, Cotta-de-Almeida V. Surmounting the obstacles that impede effective CAR T cell trafficking to solid tumors. J Leukoc Biol. 2020;108(4):1067-1079. doi: 10.1002/JLB.1MR0520-746R.

Eyquem J, Mansilla-Soto J, Giavridis T, van der Stegen SJ, Hamieh M, Cunanan KM et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017;543(7643):113-117. doi: 10.1038/nature21405.

Straathof KC, Pulè MA, Yotnda P, Dotti G, Vanin EF, Brenner MK, et al. An inducible caspase 9 safety switch for T-cell therapy. Blood. 2005;105(11):4247-4254. doi: 10.1182/blood-2004-11-4564.

Stein MN, Teply BA, Gergis U, Strickland D, Senesac J, Bayle H, et al. Early results from a phase 1, multicenter trial of PSCA-specific GoCAR T cells (BPX-601) in patients with metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol. 2023;41(6). doi: 10.1200/JCO.2023.41.6_suppl.1.

Adkins S. CAR T-cell therapy: Adverse events and management. J Adv Pract Oncol. 2019;10(Suppl 3):21-28. doi: 10.6004/jadpro.2019.10.4.11.

Downloads

Published

2024-04-15

How to Cite

Al-Janabi, I. I. (2024). CAR-T Cell Therapy for Cancer. Al-Rafidain Journal of Medical Sciences ( ISSN 2789-3219 ), 6(2), 21–31. https://doi.org/10.54133/ajms.v6i2.726

Issue

Section

Review article

Similar Articles

1 2 3 4 5 > >> 

You may also start an advanced similarity search for this article.