Journal Home > Volume 1 , Issue 1

Chimeric antigen receptor (CAR) T cell therapy is a relatively new form of targeted therapy that has demonstrated impressive success in treating hematological malignancies. It has been challenging to translate this success to solid tumors. Reasons for this include barriers to delivery, tumor heterogeneity, cancer cells' ability to evade the immune system as well as identifying the optimal target. Most CAR T clinical trials have targeted well‐characterized cancer targets with significant preclinical and in some cases clinical validation. Published results from some of these trials show signs of anti‐cancer activity that warrant encouragement, but also caution, given instances of unacceptable toxicity. The narrow therapeutic window is complicated by the ability of CAR T cells to expand in patients regardless of dose. Here, we review those trials showing encouraging results in the context of target selection. It is clear that more specific tumor targeting is required, either by affinity tuning to avoid low‐level target expression in healthy cells, logic gating, or the identification of new targets that are more cancer specific.


menu
Abstract
Full text
Outline
About this article

Target selection and clinical chimeric antigen receptor T cell activity against solid tumors

Show Author's information Eric von Hofe1 ( )Yanping Yang2Moonsoo M. Jin2
AffyImmune Therapeutics, Inc., Natick, Massachusetts, USA
Department of Radiology, Molecular Imaging Innovations Institute, Weill Cornell Medicine, New York, New York, USA

Abstract

Chimeric antigen receptor (CAR) T cell therapy is a relatively new form of targeted therapy that has demonstrated impressive success in treating hematological malignancies. It has been challenging to translate this success to solid tumors. Reasons for this include barriers to delivery, tumor heterogeneity, cancer cells' ability to evade the immune system as well as identifying the optimal target. Most CAR T clinical trials have targeted well‐characterized cancer targets with significant preclinical and in some cases clinical validation. Published results from some of these trials show signs of anti‐cancer activity that warrant encouragement, but also caution, given instances of unacceptable toxicity. The narrow therapeutic window is complicated by the ability of CAR T cells to expand in patients regardless of dose. Here, we review those trials showing encouraging results in the context of target selection. It is clear that more specific tumor targeting is required, either by affinity tuning to avoid low‐level target expression in healthy cells, logic gating, or the identification of new targets that are more cancer specific.

Keywords: targeted therapy, clinical trials, CAR T cell, solid tumors

References(87)

[1]

MacKay M, Afshinnekoo E, Rub J, Hassan C, Khunte M, Baskaran N, et al. The therapeutic landscape for cells engineered with chimeric antigen receptors. Nat Biotechnol. 2020;38(2): 233–44. https://doi.org/10.1038/s41587‐019‐0329‐2

[2]

Monsivais D, Vasquez YM, Chen F, Zhang Y, Chandrashekar DS, Faver JC, et al. Mass‐spectrometry‐based proteomic correlates of grade and stage reveal pathways and kinases associated with aggressive human cancers. Oncogene. 2021;40(11): 2081–95. https://doi.org/10.1038/s41388‐021‐01681‐0

[3]

Allen GM, Lim WA. Rethinking cancer targeting strategies in the era of smart cell therapeutics. Nat Rev Cancer. 2022;22(12): 693–702. https://doi.org/10.1038/s41568‐022‐00505‐x

[4]

Chakradhar S. Driving CARs: as ‘living drugs’, T cell therapies face dose standardization woes. Nat Med. 2015;21(11): 1236–8. https://www.nature.com/articles/nm1115‐1236

[5]

O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell‐based immunotherapy. Nat Rev Clin Oncol. 2019;16(3): 151–67. https://doi.org/10.1038/s41571‐018‐0142‐8

[6]

Cheever MA, Allison JP, Ferris AS, Finn OJ, Hastings BM, Hecht TT, et al. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15(17): 5323–37. https://doi.org/10.1158/1078‐0432.ccr‐09‐0737

[7]

Wang SS, Luong K, Gracey FM, Jabar S, McColl B, Cross RS, et al. A novel peptide‐MHC targeted chimeric antigen receptor T cell forms a T cell‐like immune synapse. Biomedicines. 2021;9(12): 1875. https://doi.org/10.3390/biomedicines9121875

[8]

Ordóñez NG. Application of mesothelin immunostaining in tumor diagnosis. Am J Surg Pathol. 2003;27(11): 1418–28. https://doi.org/10.1097/00000478‐200311000‐00003

[9]

Haas AR, Tanyi JL, O’Hara MH, Gladney WL, Lacey SF, Torigian DA, et al. Phase Ⅰ study of lentiviral‐transduced chimeric antigen receptor‐modified T cells recognizing mesothelin in advanced solid cancers. Mol Ther. 2019;27(11): 1919–29. https://www.sciencedirect.com/science/article/pii/S1525001619303284

[10]

Adusumilli PS, Zauderer MG, Rivière I, Solomon SB, Rusch VW, O’Cearbhaill RE, et al. A phase Ⅰ trial of regional mesothelin‐targeted CAR T‐cell therapy in patients with malignant pleural disease, in combination with the anti‐PD‐1 agent pembrolizumab. Cancer Discov. 2021;11(11): 2748–63. https://doi.org/10.1158/2159‐8290.cd‐21‐0407

[11]

Tanyi J, Haas A, Aggarwal C, O’Hara M, Lacey S, Golden R, et al. Phase Ⅰ study of autologous T cells bearing fully‐humanized chimeric antigen receptors targeting mesothelin in mesothelin‐expressing cancers (314). Gynecol Oncol [Internet]. 2022;166: S164–5. https://www.sciencedirect.com/science/article/pii/S0090825822015372

[12]

Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. Antitumor activity and long‐term fate of chimeric antigen receptor‐positive T cells in patients with neuroblastoma. Blood. 2011;118(23): 6050–6. https://doi.org/10.1182/blood‐2011‐05‐354449

[13]

Heczey A, Louis CU, Savoldo B, Dakhova O, Durett A, Grilley B, et al. CAR T cells administered in combination with lymphodepletion and PD‐1 inhibition to patients with neuroblastoma. Mol Ther. 2017;25(9): 2214–24. https://doi.org/10.1016/j.ymthe.2017.05.012

[14]

Straathof K, Flutter B, Wallace R, Jain N, Loka T, Depani S, et al. Antitumor activity without on‐target off‐tumor toxicity of GD2‐chimeric antigen receptor T cells in patients with neuroblastoma. Sci Transl Med. 2020;12(571): eabd6169. https://doi.org/10.1126/scitranslmed.abd6169

[15]

Yu L, Huang L, Lin D, Lai X, Wu L, Liao X, et al. GD2‐specific chimeric antigen receptor‐modified T cells for the treatment of refractory and/or recurrent neuroblastoma in pediatric patients. J Cancer Res Clin Oncol. 2022;148(10): 2643–52. https://doi.org/10.1007/s00432‐021‐03839‐5

[16]

Majzner RG, Ramakrishna S, Yeom KW, Patel S, Chinnasamy H, Schultz LM, et al. GD2‐CAR T cell therapy for H3K27M‐mutated diffuse midline gliomas. Nature. 2022;603(7903): 934–41. https://doi.org/10.1038/s41586‐022‐04489‐4

[17]

Shi D, Shi Y, Kaseb AO, Qi X, Zhang Y, Chi J, et al. Chimeric antigen receptor‐glypican‐3 T‐cell therapy for advanced hepatocellular carcinoma: results of phase Ⅰ trials. Clin Cancer Res. 2020;26(15): 3979–89. https://doi.org/10.1158/1078‐0432.ccr‐19‐3259

[18]

Fang W, Fu Q, Zhao Q, Zheng Y, Liu L, Li Z, et al. Phase Ⅰ trial of fourth‐generation chimeric antigen receptor T‐cells targeting glypican‐3 for advanced hepatocellular carcinoma. JCO [Internet]. 2021;39(15_suppl): 4088. https://ascopubs.org/doi/abs/10.1200/JCO.2021.39.15_suppl.4088

[19]

Sun H, Xing C, Jiang S, Yu K, Dai S, Kong H, et al. Long term complete response of advanced hepatocellular carcinoma to glypican‐3 specific chimeric antigen receptor T‐Cells plus sorafenib, a case report. Front Immunol. 2022;13: 963031. https://doi.org/10.3389/fimmu.2022.963031

[20]

Zhao Z, Guo W, Fang S, Song S, Song J, Teng F, et al. An armored GPC3‐directed CAR‐T for refractory or relapsed hepatocellular carcinoma in China: A phase Ⅰ trial. JCO [Internet]. 2021;39(15_suppl): 4095. https://ascopubs.org/doi/10.1200/JCO.2021.39.15_suppl.4095

[21]

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): 1248–58. https://doi.org/10.1016/j.ymthe.2017.03.010

[22]

Katz SC, Moody AE, Guha P, Hardaway JC, Prince E, LaPorte J, et al. HITM‐SURE: hepatic immunotherapy for metastases phase Ib anti‐CEA CAR‐T study utilizing pressure enabled drug delivery. J Immunother Cancer. 2020;8(2): e001097. https://doi.org/10.1136/jitc‐2020‐001097

[23]

Hegde M, DeRenzo CC, Zhang H, Mata M, Gerken C, Shree A, et al. Expansion of HER2‐CAR T cells after lymphodepletion and clinical responses in patients with advanced sarcoma. JCO [Internet]. 2017;35(15_suppl): 10508. https://ascopubs.org/doi/10.1200/JCO.2017.35.15_suppl.10508

[24]

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): 1094–101. https://doi.org/10.1001/jamaoncol.2017.0184

[25]
Feng K, Liu Y, Guo Y, Qiu J, Wu Z, Dai H, et al. Phase Ⅰ study of chimeric antigen receptor modified T cells in treating HER2‐positive advanced biliary tract cancers and pancreatic cancers. Protein Cell. 2018;9(10): 838–47. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6160389/
DOI
[26]

Zhan X, Wang B, Li Z, Li J, Wang H, Chen L, et al. Phase Ⅰ trial of Claudin 18.2‐specific chimeric antigen receptor T cells for advanced gastric and pancreatic adenocarcinoma. JCO [Internet]. 2019;37(15_suppl):2509. https://ascopubs.org/doi/abs/10.1200/JCO.2019.37.15_suppl.2509

[27]

Qi C, Gong J, Li J, Liu D, Qin Y, Ge S, et al. Claudin18.2‐specific CAR T cells in gastrointestinal cancers: phase 1 trial interim results. Nat Med. 2022;28(6):1189–98. https://www.nature.com/articles/s41591‐022‐01800‐8

[28]

Haanen JB, Mackensen A, Koenecke C, Alsdorf W, Wagner‐Drouet E, Heudobler D, et al. Abstract CT002: BNT211: a Phase Ⅰ trial to evaluate safety and efficacy of CLDN6 CAR‐T cells and CARVac‐mediated in vivo expansion in patients with CLDN6‐positive advanced solid tumors. Cancer Res. 2022;82(12_Supplement):CT002. https://doi.org/10.1158/1538‐7445.AM2022‐CT002

[29]

Lin Y, Chen S, Zhong S, An H, Yin H, McGowan E. Phase Ⅰ clinical trial of PD‐1 knockout anti‐MUC1 CAR‐T cells in the treatment of patients with non‐small cell lung cancer. Ann Oncol. 2019;30:xi12. https://www.annalsofoncology.org/article/S0923‐7534(20)34414‐8/fulltext

[30]

Li Q, Wang Y, Lin M, Xia L, Bao Y, Sun X, et al. Abstract A014: phase Ⅰ clinical trial with PD‐1/MUC1 CAR‐pNK92 immunotherapy. Cancer Immunol Res. 2019;7(2_Supplement):A014. https://doi.org/10.1158/2326‐6074.CRICIMTEATIAACR18‐A014

[31]

Junghans RP, Ma Q, Rathore R, Gomes EM, Bais AJ, Lo ASY, 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):1257–70. https://doi.org/10.1002/pros.23214

[32]

Narayan V, Barber‐Rotenberg JS, Jung IY, Lacey SF, Rech AJ, Davis MM, et al. PSMA‐targeting TGFβ‐insensitive armored CAR T cells in metastatic castration‐resistant prostate cancer: a phase 1 trial. Nat Med. 2022;28(4):724–34. https://doi.org/10.1038/s41591‐022‐01726‐1

[33]

Slovin SF, Dorff TB, Falchook GS, Wei XX, Gao X, McKay RR, et al. Phase 1 study of P‐PSMA‐101 CAR‐T cells in patients with metastatic castration‐resistant prostate cancer (mCRPC). JCO. 2022;40(6_suppl):98. https://ascopubs.org/doi/abs/10.1200/JCO.2022.40.6_suppl.098

[34]

Bharadwaj U, Li M, Chen C, Yao Q. Mesothelin‐induced pancreatic cancer cell proliferation involves alteration of cyclin E via activation of signal transducer and activator of transcription protein 3. Mol Cancer Res. 2008;6(11):1755–65. https://doi.org/10.1158/1541‐7786.mcr‐08‐0095

[35]

Greenbaum U, Yalniz FF, Srour SA, Rezvani K, Singh H, Olson A, et al. Chimeric antigen receptor therapy: how are we driving in solid tumors? Biol Blood Marrow Transplant. 2020;26(10):1759–69. https://doi.org/10.1016/j.bbmt.2020.06.020

[36]

Castelletti L, Yeo D, van Zandwijk N, Rasko JEJ. Anti‐Mesothelin CAR T cell therapy for malignant mesothelioma. Biomarker Res. 2021;9(1):11. https://doi.org/10.1186/s40364‐021‐00264‐1

[37]

Beatty GL, O’Hara MH, Lacey SF, Torigian DA, Nazimuddin F, Chen F, et al. Activity of mesothelin‐specific chimeric antigen receptor T cells against pancreatic carcinoma metastases in a phase 1 trial. Gastroenterology. 2018;155(1):29–32. https://doi.org/10.1053/j.gastro.2018.03.029

[38]

Nazha B, Inal C, Owonikoko TK. Disialoganglioside GD2 expression in solid tumors and role as a target for cancer therapy. Front Oncol. 2020;10:1000. https://doi.org/10.3389/fonc.2020.01000

[39]

Richman SA, Nunez‐Cruz S, Moghimi B, Li LZ, Gershenson ZT, Mourelatos Z, et al. High‐affinity GD2‐specific CAR T cells induce fatal encephalitis in a preclinical neuroblastoma model. Cancer Immunol Res. 2018;6(1):36–46. https://doi.org/10.1158/2326‐6066.CIR‐17‐0211

[40]

Lammie G, Cheung N, Gerald W, Rosenblum M, Cordoncardo C. Ganglioside GD(2) expression in the human nervous‐system and in neuroblastomas – an immunohistochemical study. Int J Oncol. 1993;3(5):909–15. https://doi.org/10.3892/ijo.3.5.909

[41]

Zheng X, Liu X, Lei Y, Wang G, Liu M. Glypican‐3: a novel and promising target for the treatment of hepatocellular carcinoma. Front Oncol. 2022;12:824208. https://doi.org/10.3389/fonc.2022.824208

[42]

Baumhoer D, Tornillo L, Stadlmann S, Roncalli M, Diamantis EK, Terracciano LM. Glypican 3 expression in human nonneoplastic, preneoplastic, and neoplastic tissues: a tissue microarray analysis of 4,387 tissue samples. Am J Clin Pathol. 2008;129(6):899–906. https://doi.org/10.1309/hcqwpwd50xhd2dw6

[43]

Liu X, Onda M, Watson N, Hassan R, Ho M, Bera TK, et al. Highly active CAR T cells that bind to a juxtamembrane region of mesothelin and are not blocked by shed mesothelin. Proc Natl Acad Sci U S A. 2022;119(19):e2202439119. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9171807/

[44]

Hall C, Clarke L, Pal A, Buchwald P, Eglinton T, Wakeman C, et al. A review of the role of carcinoembryonic antigen in clinical practice. Ann Coloproctol. 2019;35(6):294–305. https://doi.org/10.3393/ac.2019.11.13

[45]

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. 2022;3(Suppl 1):24–31. https://doi.org/10.1002/jha2.356

[46]

Wu S, Gu W. Association of T Stage and serum CEA levels in determining survival of rectal cancer. Front Med. 2019;6:270. https://doi.org/10.3389/fmed.2019.00270

[47]

Lee JH, Lee SW. The roles of carcinoembryonic antigen in liver metastasis and therapeutic approaches. Gastroenterol Res Pract. 2017;2017:7521987. https://doi.org/10.1155/2017/7521987

[48]

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):1425–36. https://doi.org/10.1007/s00262‐017‐2034‐7

[49]

Adamczyk A, Grela‐Wojewoda A, Domagała‐Haduch M, Ambicka A, Harazin‐Lechowska A, Janecka A, et al. Proteins involved in HER2 signalling pathway, their relations and influence on metastasis‐free survival in HER2‐positive breast cancer patients treated with Trastuzumab in adjuvant setting. J Cancer. 2017;8(1):131–9. https://doi.org/10.7150/jca.16239

[50]

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):843–51. https://doi.org/10.1038/mt.2010.24

[51]

Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, et al. Human epidermal growth factor receptor 2 (HER2) – specific chimeric antigen receptor–modified T cells for the immunotherapy of HER2‐positive sarcoma. JCO. 2015;33(15):1688–96. https://ascopubs.org/doi/full/10.1200/JCO.2014.58.0225

[52]

Hegde M, Joseph SK, Pashankar F, DeRenzo C, Sanber K, Navai S, et al. Tumor response and endogenous immune reactivity after administration of HER2 CAR T cells in a child with metastatic rhabdomyosarcoma. Nat Commun. 2020;11(1):3549. https://doi.org/10.1038/s41467‐020‐17175‐8

[53]

Arnold A, Daum S, von Winterfeld M, Berg E, Hummel M, Rau B, et al. Prognostic impact of Claudin 18.2 in gastric and esophageal adenocarcinomas. Clin Transl Oncol. 2020;22(12):2357–63. https://doi.org/10.1007/s12094‐020‐02380‐0

[54]

Kyuno D, Takasawa A, Takasawa K, Ono Y, Aoyama T, Magara K, et al. Claudin‐18.2 as a therapeutic target in cancers: cumulative findings from basic research and clinical trials. Tissue Barriers. 2022;10(1):1967080. https://doi.org/10.1080/21688370.2021.1967080

[55]

Xu X, Jin H, Liu Y, Liu L, Wu Q, Guo Y, et al. The expression patterns and correlations of claudin‐6, methy‐CpG binding protein 2, DNA methyltransferase 1, histone deacetylase 1, acetyl‐histone H3 and acetyl‐histone H4 and their clinicopathological significance in breast invasive ductal carcinomas. Diagn Pathol. 2012;7(1):33. https://doi.org/10.1186/1746‐1596‐7‐33

[56]

Stadler CR, Bähr‐Mahmud H, Plum LM, Schmoldt K, Kölsch AC, Türeci Ö, et al. Characterization of the first‐in‐class T‐cell‐engaging bispecific single‐chain antibody for targeted immunotherapy of solid tumors expressing the oncofetal protein claudin 6. OncoImmunology. 2016;5(3):e1091555. https://doi.org/10.1080/2162402x.2015.1091555

[57]

Reinhard K, Rengstl B, Oehm P, Michel K, Billmeier A, Hayduk N, et al. An RNA vaccine drives expansion and efficacy of claudin‐CAR‐T cells against solid tumors. Science. 2020;367(6476):446–53. https://doi.org/10.1126/science.aay5967

[58]

Taylor‐Papadimitriou J, Burchell JM, Graham R, Beatson R. Latest developments in MUC1 immunotherapy. Biochem Soc Trans. 2018;46(3):659–68. https://doi.org/10.1042/BST20170400

[59]

Bose M, Mukherjee P. Potential of anti‐MUC1 antibodies as a targeted therapy for gastrointestinal cancers. Vaccines (Basel). 2020;8(4):659. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7712407/

[60]

Saeland E, Belo AI, Mongera S, van Die I, Meijer GA, van Kooyk Y. Differential glycosylation of MUC1 and CEACAM5 between normal mucosa and tumour tissue of colon cancer patients. Int J Cancer. 2012;131(1):117–28. https://doi.org/10.1002/ijc.26354

[61]

Horm TM, Schroeder JA. MUC1 and metastatic cancer: expression, function and therapeutic targeting. Cell Adh Migr. 2013;7(2):187–98. https://doi.org/10.4161/cam.23131

[62]

Ghosh SK, Pantazopoulos P, Medarova Z, Moore A. Expression of underglycosylated MUC1 antigen in cancerous and adjacent normal breast tissues. Clin Breast Cancer. 2013;13(2):109–18. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3578066/

[63]

Caromile LA, Shapiro LH. PSMA redirects MAPK to PI3K‐AKT signaling to promote prostate cancer progression. Mol Cell Oncol. 2017;4(4):e1321168. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5540209/

[64]

Gladney W, Vultur A, Schweizer M, Fraietta J, Rech A, June C, et al. 335 Analyses of severe immune‐mediated toxicity in patients with advanced mCRPC treated with a PSMA‐targeted armored CAR T‐cells. J Immunother Cancer. 2022;10(Suppl 2). https://jitc.bmj.com/content/10/Suppl_2/A353

[65]

Barros LRC, Couto SCF, da Silva Santurio D, Paixão EA, Cardoso F, da Silva VJ, et al. Systematic review of available CAR‐T cell trials around the world. Cancers. 2022;14(11):2667. https://doi.org/10.3390/cancers14112667

[66]

Barrett D, Chagin K, Fountaine TJ, Moore A, Ka M, Gladney W, et al. TmPSMA‐02: a CD2 endodomain containing double armored PSMA CAR T with enhanced efficacy and lower immune toxicity. JCO. 2022;40(6_suppl):158. https://ascopubs.org/doi/abs/10.1200/JCO.2022.40.6_suppl.158

[67]

Morris EC, Neelapu SS, Giavridis T, Sadelain M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat Rev Immunol. 2022;22(2):85–96. https://www.nature.com/articles/s41577‐021‐00547‐6

[68]

Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, et al. Chimeric antigen receptor T‐cell therapy — assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15(1):47–62. https://www.nature.com/articles/nrclinonc.2017.148

[69]

Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, Groot C, et al. Treatment of metastatic renal cell carcinoma with CAIX CAR‐engineered T cells: clinical evaluation and management of on‐target toxicity. Mol Ther. 2013;21(4):904–12. https://doi.org/10.1038/mt.2013.17

[70]

Silver DA, Pellicer I, Fair WR, Heston WD, Cordon‐Cardo C. Prostate‐specific membrane antigen expression in normal and malignant human tissues. Clin Cancer Res. 1997;3(1):81–5.

[71]

Bonini C, Ferrari G, Verzeletti S, Servida P, Zappone E, Ruggieri L, et al. HSV‐TK gene transfer into donor lymphocytes for control of allogeneic graft‐versus‐leukemia. Science. 1997;276(5319):1719–24. https://doi.org/10.1126/science.276.5319.1719

[72]

Kalinin RS, Petukhov AV, Knorre VD, Maschan MA, Stepanov AV, Gabibov AG. Molecular approaches to safe and controlled engineered T‐cell therapy. Acta Naturae. 2018;10(2):16–23. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6087824/

[73]

Caruso HG, Hurton LV, Najjar A, Rushworth D, Ang S, Olivares S, et al. Tuning sensitivity of CAR to EGFR density limits recognition of normal tissue while maintaining potent antitumor activity. Cancer Res. 2015;75(17):3505–18. https://doi.org/10.1158/0008‐5472.CAN‐15‐0139

[74]

Liu X, Jiang S, Fang C, Yang S, Olalere D, Pequignot EC, 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):3596–607. https://doi.org/10.1158/0008‐5472.CAN‐15‐0159

[75]

Park S, Shevlin E, Vedvyas Y, Zaman M, Park S, Hsu YMS, et al. Micromolar affinity CAR T cells to ICAM‐1 achieves rapid tumor elimination while avoiding systemic toxicity. Sci Rep. 2017;7(1):14366. https://doi.org/10.1038/s41598‐017‐14749‐3

[76]

Vander Mause ER, Atanackovic D, Lim CS, Luetkens T. Roadmap to affinity‐tuned antibodies for enhanced chimeric antigen receptor T cell function and selectivity. Trends Biotechnol. 2022;40(7):875–90. https://www.sciencedirect.com/science/article/pii/S0167779921003152

[77]

Mansilla‐Soto J, Eyquem J, Haubner S, Hamieh M, Feucht J, Paillon N, et al. HLA‐independent T cell receptors for targeting tumors with low antigen density. Nat Med. 2022;28(2):345–52. https://doi.org/10.1038/s41591‐021‐01621‐1

[78]

You Y, Lai X, Pan Y, Zheng H, Vera J, Liu S, et al. Artificial intelligence in cancer target identification and drug discovery. Signal Transduct Target Ther. 2022;7(1):156. https://doi.org/10.1038/s41392‐022‐00994‐0

[79]

Rose M, Cardon T, Aboulouard S, Hajjaji N, Kobeissy F, Duhamel M, et al. Surfaceome proteomic of glioblastoma revealed potential targets for immunotherapy. Front Immunol. 2021;12:746168. https://doi.org/10.3389/fimmu.2021.746168

[80]

Lee JK, Bangayan NJ, Chai T, Smith BA, Pariva TE, Yun S, et al. Systemic surfaceome profiling identifies target antigens for immune‐based therapy in subtypes of advanced prostate cancer. Proc Natl Acad Sci U S A. 2018;115(19):E4473–82. https://doi.org/10.1073/pnas.1802354115

[81]

Bogetofte Barnkob M, Vitting‐Seerup K, Rønn Olsen L. Target isoforms are an overlooked challenge and opportunity in chimeric antigen receptor cell therapy. Immunother Adv. 2022;2(1):ltac009. https://doi.org/10.1093/immadv/ltac009

[82]

Raglow Z, McKenna MK, Bonifant CL, Wang W, Pasca di Magliano M, Stadlmann J, et al. Targeting glycans for CAR therapy: the advent of sweet CARs. Mol Ther. 2022;30(9):2881–90. https://www.sciencedirect.com/science/article/pii/S1525001622004270

[83]

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. https://www.science.org/doi/full/10.1126/scitranslmed.aaa0984

[84]

Mao R, Kong W, He Y. The affinity of antigen‐binding domain on the antitumor efficacy of CAR T cells: moderate is better. Front Immunol. 2022;13:1032403. https://doi.org/10.3389/fimmu.2022.1032403

[85]

Karam A, Mjaess G, Martinez Chanza N, Aoun F, Bou Kheir G, Younes H, et al. CAR‐T cell therapy for solid tumors: are we still that far? A systematic review of literature. Cancer Invest. 2022;40(10):923–37. https://doi.org/10.1080/07357907.2022.2125004

[86]

Mehrabadi AZ, Ranjbar R, Farzanehpour M, Shahriary A, Dorostkar R, Hamidinejad MA, et al. Therapeutic potential of CAR T cell in malignancies: a scoping review. Biomed Pharmacother. 2022;146:112512. https://doi.org/10.1016/j.biopha.2021.112512

[87]

Abbott RC, Hughes‐Parry HE, Jenkins MR. To go or not to go? Biological logic gating engineered T cells. J Immunother Cancer. 2022;10(4):e004185. https://jitc.bmj.com/lookup/doi/10.1136/jitc‐2021‐004185

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 10 February 2023
Accepted: 03 April 2023
Published: 19 May 2023
Issue date: June 2023

Copyright

© 2023 The Authors. Tsinghua University Press.

Acknowledgements

ACKNOWLEDGMENTS

None.

Rights and permissions

This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

Return