Abstract
Ovarian cancer (OC) is one of the most common cancers in women, with a high mortality rate and very few available and effective treatments. Evidence shows that immunotherapy in OC has not been very successful because immune checkpoint blockers have not achieved satisfactory clinical outcomes. On the other hand, as one of the effective treatment approaches, chimeric antigen receptor T-cell (CAR T-cell) therapy has gained a moral position, especially in blood malignancies. Although in solid tumors, CAR T-cell therapy faces various complications and challenges. One of these challenges is selecting the appropriate tumor antigen targeted by CAR T cells, making the selection difficult due to the expression of antigens by tumor cells and normal cells. In addition, the rate of tumor antigen expression and CAR T-cell access to the desired antigen and proper stimulation of CAR T cells can be other important points in antigen selection. This review summarized common tumor antigens and the challenges of selecting them in CAR T cells therapy of OC.


Similar content being viewed by others
Data availability
Not applicable.
References
Ovarian cancer statistics. [cited 2021 7.1.2021]; Available from: https://www.wcrf.org/dietandcancer/ovarian-cancer-statistics/.
Yan W, Hu H, Tang B. Advances of chimeric antigen receptor T cell therapy in ovarian cancer. Onco Targets Ther. 2019;12:8015.
Webb PM, Jordan SJ. Epidemiology of epithelial ovarian cancer. Best Pract Res Clin Obstet Gynaecol. 2017;41:3–14.
Ovarian Cancer Stages. [cited 2021 1.7.2021]; Available from: https://www.cancer.org/cancer/ovarian-cancer/detection-diagnosis-staging/staging.html.
Bookman M. Standard treatment in advanced ovarian cancer in 2005: the state of the art. Int J Gynecol Cancer. 2005;15:Suppl 3.
Tewari KS, et al. Final overall survival of a randomized trial of bevacizumab for primary treatment of ovarian cancer. J Clin Oncol. 2019;37(26):2317.
Natoli M, et al. Human ovarian cancer intrinsic mechanisms regulate lymphocyte activation in response to immune checkpoint blockade. Cancer Immunol Immunother. 2020;69(8):1391.
Met, Ö., et al. (2019) Principles of adoptive T cell therapy in cancer. in Seminars in immunopathology. Springer.
Mohanty R, et al. CAR T cell therapy: a new era for cancer treatment. Oncol Rep. 2019;42(6):2183–95.
June CH, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361–5.
Marofi F, et al. CAR T cells in solid tumors: challenges and opportunities. Stem Cell Res Ther. 2021;12(1):1–16.
Chekmasova AA, Brentjens RJ. Adoptive T cell immunotherapy strategies for the treatment of patients with ovarian cancer. Discov Med. 2010;9(44):62–70.
Tanyi JL, et al. Possible compartmental cytokine release syndrome in a patient with recurrent ovarian cancer after treatment with mesothelin-targeted CAR-T cells. J Immunother. 2017;40(3):104–7.
Zhu X, et al. CAR-T cell therapy in ovarian cancer: from the bench to the bedside. Oncotarget. 2017;8(38):64607.
Roselli E, Faramand R, Davila ML. Insight into next-generation CAR therapeutics: designing CAR T cells to improve clinical outcomes. J Clin Invest. 2021;131:2.
Zhang C, et al. Engineering car-t cells. Biomark Res. 2017;5(1):1–6.
Styczyński J. A brief history of CAR-T cells: from laboratory to the bedside. Acta Haematol Pol. 2020;51(1):2–5.
Quintás-Cardama A. What CAR will win the CD19 race? Mol Cancer Ther. 2019;18(3):498–506.
Mariuzza RA, Agnihotri P, Orban J. The structural basis of T-cell receptor (TCR) activation: an enduring enigma. J Biol Chem. 2020;295(4):914–25.
June CH, Sadelain M. Chimeric antigen receptor therapy. N Engl J Med. 2018;379(1):64–73.
Silbert S, Yanik GA, Shuman AG. How should we determine the value of CAR T-cell therapy? AMA J Ethics. 2019;21(10):844–51.
Levine BL, et al. Global manufacturing of CAR T cell therapy. Mol Therapy-Methods Clin Develop. 2017;4:92–101.
Turtle CJ, et al. CD19 CAR–T cells of defined CD4+: CD8+ composition in adult B cell ALL patients. J Clin Investig. 2016;126(6):2123–38.
Brudno JN, Kochenderfer JN. Chimeric antigen receptor T-cell therapies for lymphoma. Nat Rev Clin Oncol. 2018;15(1):31.
Siddiqi HF, Staser KW, Nambudiri VE. Research techniques made simple: CAR T-cell therapy. J Invest Dermatol. 2018;138(12):2501–4.
Kim DW, Cho J-Y. Recent advances in allogeneic CAR-T cells. Biomolecules. 2020;10(2):263.
Luangwattananun P, et al. Fourth-generation chimeric antigen receptor T cells targeting folate receptor alpha antigen expressed on breast cancer cells for adoptive T cell therapy. Breast Cancer Res Treat. 2021;186(1):25–36.
Yeku OO, Brentjens RJ. Armored CAR T-cells: utilizing cytokines and pro-inflammatory ligands to enhance CAR T-cell anti-tumour efficacy. Biochem Soc Trans. 2016;44(2):412–8.
Chen H, et al. CD27 enhances the killing effect of CAR T cells targeting trophoblast cell surface antigen 2 in the treatment of solid tumors. Cancer Immunol Immunother. 2021;70(7):2059–71.
Chacon JA, et al. Co-stimulation through 4–1BB/CD137 improves the expansion and function of CD8+ melanoma tumor-infiltrating lymphocytes for adoptive T-cell therapy. PLoS ONE. 2013;8(4): e60031.
Zuccolotto G, et al. PSMA-specific car-engineered T cells for prostate cancer: CD28 outperforms combined CD28–41BB “Super-stimulation”. Front. Oncol. 2021.
Song D-G, Powell DJ. Pro-survival signaling via CD27 costimulation drives effective CAR T-cell therapy. Oncoimmunology. 2012;1(4):547–9.
Schubert M-L, et al. Third-generation CAR T cells targeting CD19 are associated with an excellent safety profile and might improve persistence of CAR T cells in treated patients. DC: American Society of Hematology Washington; 2019.
Chmielewski M, Abken H. TRUCKs: the fourth generation of CARs. Expert Opin Biol Ther. 2015;15(8):1145–54.
Tokarew N, et al. Teaching an old dog new tricks: next-generation CAR T cells. Br J Cancer. 2019;120(1):26–37.
Siroy A, et al. MUC1 is expressed at high frequency in early-stage basal-like triple-negative breast cancer. Hum Pathol. 2013;44(10):2159–66.
Maeda T, et al. MUC1-C induces PD-L1 and immune evasion in triple-negative breast cancer. Can Res. 2018;78(1):205–15.
Zhou R, et al. CAR T cells targeting the tumor MUC1 glycoprotein reduce triple-negative breast cancer growth. Front Immunol. 2019;10:1149.
Aghili M, et al. Triple-negative breast cancer survival in Iranian patients. Acta Medica Iranica. 2013;560–566.
Zuo B-L, et al. Targeting and suppression of HER3-positive breast cancer by T lymphocytes expressing a heregulin chimeric antigen receptor. Cancer Immunol Immunother. 2018;67(3):393–401.
Lamers CH, 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.
Lamers CH, et al. Treatment of metastatic renal cell carcinoma (mRCC) with CAIX CAR-engineered T-cells–a completed study overview. Biochem Soc Trans. 2016;44(3):951–9.
Jiang H, et al. Claudin18. 2-specific chimeric antigen receptor engineered T cells for the treatment of gastric cancer. JNCI J Nat Cancer Instit. 2019;111(4):409–18.
Tao K, et al. Development of NKG2D-based chimeric antigen receptor-T cells for gastric cancer treatment. Cancer Chemother Pharmacol. 2018;82(5):815–27.
Song Y, et al. Effective and persistent antitumor activity of HER2-directed CAR-T cells against gastric cancer cells in vitro and xenotransplanted tumors in vivo. Protein Cell. 2018;9(10):867–78.
Jung M, et al. Chimeric antigen receptor T cell therapy targeting ICAM-1 in gastric cancer. Molecular Therapy-Oncolytics. 2020;18:587–601.
Chen X, et al. Combined DLL3-targeted bispecific antibody with PD-1 inhibition is efficient to suppress small cell lung cancer growth. J Immunotherapy Cancer. 2020;8:1.
Hillerdal V, Essand M. Chimeric antigen receptor-engineered T cells for the treatment of metastatic prostate cancer. BioDrugs. 2015;29(2):75–89.
Weimin S, et al. Chimeric cytokine receptor enhancing PSMA-CAR-T cell-mediated prostate cancer regression. Cancer Biol Ther. 2020;21(6):570–80.
Junghans RP, 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.
Slovin SF, et al. Chimeric antigen receptor (CAR+) modified T cells targeting prostate specific membrane antigen (PSMA) in patients (pts) with castrate metastatic prostate cancer (CMPC). Am Soc Clin Oncol. 2013.
Batra SA, et al. Glypican-3–specific CAR T cells coexpressing IL15 and IL21 have superior expansion and antitumor activity against hepatocellular carcinoma. Cancer Immunol Res. 2020;8(3):309–20.
Jin L, et al. CXCR1-or CXCR2-modified CAR T cells co-opt IL-8 for maximal antitumor efficacy in solid tumors. Nat Commun. 2019;10(1):1–13.
Zhang Y, et al. Novel cellular immunotherapy using NKG2D CAR-T for the treatment of cervical cancer. Biomed Pharmacother. 2020;131: 110562.
Murad JP, et al. Effective targeting of TAG72+ peritoneal ovarian tumors via regional delivery of CAR-engineered T cells. Front Immunol. 2018;9:2268.
Chekmasova AA, et al. Successful eradication of established peritoneal ovarian tumors in SCID-Beige mice following adoptive transfer of T cells genetically targeted to the MUC16 antigen. Clin Cancer Res. 2010;16(14):3594–606.
Yeku OO, et al. Interleukin-12 armored chimeric antigen receptor (CAR) T cells for heterogeneous antigen-expressing ovarian cancer. Am Soc Clin Oncol. 2018.
Hung C-F, et al. Development of anti-human mesothelin-targeted chimeric antigen receptor messenger RNA–transfected peripheral blood lymphocytes for ovarian cancer therapy. Hum Gene Ther. 2018;29(5):614–25.
Lu Y, Low PS. Immunotherapy of folate receptor-expressing tumors: review of recent advances and future prospects. J Control Release. 2003;91(1–2):17–29.
Zuo S, et al. Modification of cytokine-induced killer cells with folate receptor alpha (FRα)-specific chimeric antigen receptors enhances their antitumor immunity toward FRα-positive ovarian cancers. Mol Immunol. 2017;85:293–304.
Song D-G, et al. Chimeric NKG2D CAR-expressing T cell-mediated attack of human ovarian cancer is enhanced by histone deacetylase inhibition. Hum Gene Ther. 2013;24(3):295–305.
Rodriguez-Garcia A, et al. CAR T cells targeting MISIIR for the treatment of ovarian cancer and other gynecologic malignancies. Mol Ther. 2020;28(2):548–60.
Fu J, et al. Chimeric Antigen receptor-T (CAR-T) cells targeting Epithelial cell adhesion molecule (EpCAM) can inhibit tumor growth in ovarian cancer mouse model. J Veter Med Sci. 2020;20–0455.
Wrigley E, et al. 5T4 oncofetal antigen expression in ovarian carcinoma. Int J Gynecol Cancer. 1995;5:4.
Owens GL, et al. Preclinical assessment of CAR T-cell therapy targeting the tumor antigen 5T4 in ovarian cancer. J Immun (Hagerstown, Md: 1997). 2018;41(3):130.
Li T, Wang J. Therapeutic effect of dual CAR-T targeting PDL1 and MUC16 antigens on ovarian cancer cells in mice. BMC Cancer. 2020;20(1):1–13.
Shu R, et al. Engineered CAR-T cells targeting TAG-72 and CD47 in ovarian cancer. Mol Therapy-Oncol. 2021;20:325–41.
Jiang G, et al. Dual CAR-T cells to treat cancers co-expressing NKG2D and PD1 ligands in xenograft models of peritoneal metastasis. Cancer Immunol Immunotherapy. 2022;1–12.
Wang W, et al. LGR5 CAR-T cells: a novel potential treatment against high grade serous ovarian cancer. Can Res. 2022;82(12_Supplement):5183–5183.
Schepisi G, et al. Immunotherapy and its development for gynecological (ovarian, endometrial and cervical) tumors: from immune checkpoint inhibitors to chimeric antigen receptor (CAR)-T cell therapy. Cancers. 2021;13(4):840.
CAR T cell receptor immunotherapy targeting mesothelin for patients with metastatic cancer. 2021 [cited 2021 7.7.2021]; Available from: https://clinicaltrials.gov/ct2/show/NCT01583686.
Herbel C, et al. Identification of a novel tumor marker combination THY1-EPCAM for adaptor CAR T cell therapy in ovarian cancer. Cancer Res. 2022;82(12_Supplement):2813–2813.
Rao TD, et al. Novel monoclonal antibodies against the proximal (carboxy-terminal) portions of MUC16. Appl Immunohistochem Mol Morphol: AIMM/Off Publ Soc Appl Immunohistochem. 2010;18(5):462.
Kollmorgen G, et al. A re-engineered immunotoxin shows promising preclinical activity in ovarian cancer. Sci Rep. 2017;7(1):1–13.
Hassan R, et al. Mesothelin is overexpressed in pancreaticobiliary adenocarcinomas but not in normal pancreas and chronic pancreatitis. Am J Clin Pathol. 2005;124(6):838–45.
Pastan I, Hassan R. Discovery of mesothelin and exploiting it as a target for immunotherapy. Can Res. 2014;74(11):2907–12.
Li X, et al. PLAP-CAR T cells mediate high specific cytotoxicity against colon cancer cells. Front Biosci (Landmark Ed). 2020;25:1765–86.
Golubovskaya VM, et al. PLAP (placental alkaline phosphatase)-CAR-T cells specifically target colorectal cancer. Cancer Res. 2020;80:16.
Zhan X, et al. Phase I trial of Claudin 18.2-specific chimeric antigen receptor T cells for advanced gastric and pancreatic adenocarcinoma. Am Soc Clin Oncol. 2019.
Reinhard K, et al. An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors. Science. 2020;367(6476):446–53.
Levitsky K, et al. Allogeneic anti-PTK7 CAR-T cells for the treatment of solid tumors. Cancer Res. 2020;80:16.
Sachdev J, et al. A phase 1 study of PF-06647020, an antibody-drug conjugate (ADC) targeting protein tyrosine kinase 7 (PTK7), in patients with advanced solid tumors including platinum resistant ovarian cancer (OVCA). Ann Oncol. 2017;27:vi570.
Kershaw MH, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. 2006;12(20):6106–15.
Song D-G, et al. In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatory signaling through CD137 (4–1BB). Can Res. 2011;71(13):4617–27.
Delaine-Smith RM, et al. Modelling TGFβR and Hh pathway regulation of prognostic matrisome molecules in ovarian cancer. Iscience. 2021;24(6): 102674.
Joy JD, et al. TGFβ-mediated targeting of the extracellular matrix enhances the migration and cytotoxicity of CAR-T cells in 3D models of ovarian cancer. Cancer Res. 2022;82(12_Supplement):693–693.
Acknowledgements
None.
Funding
This work was supported by Public Technology Applied Research Projects of Zhejiang Province (LGF22H060023 to WQL), Medical and Health Research Project of Zhejiang Province (2022KY433 to WQL), Traditional Chinese Medicine Science and Technology Projects of Zhejiang Province (2022ZB382 to WQL), Research Fund Projects of The Affiliated Hospital of Zhejiang Chinese Medicine University (2021FSYYZY45 to WQL).
Author information
Authors and Affiliations
Contributions
W L and Q L: conception, design and inviting co-authors to participate. H D, J Z and F Z: writing original manuscript draft. W L, Q L, Y X and Y Y: review and editing of manuscript critically for important intellectual content and provided comments and feedback for the scientific contents of the manuscript. All authors read, revised and approved the final manuscript.
Corresponding authors
Ethics declarations
Conflict of interests
All authors of this article declare that they have no conflict of interest.
Ethical approval
This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.
Consent to participate
Not applicable.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Ding, H., Zhang, J., Zhang, F. et al. The challenge of selecting tumor antigens for chimeric antigen receptor T-cell therapy in ovarian cancer. Med Oncol 39, 232 (2022). https://doi.org/10.1007/s12032-022-01824-7
Received:
Accepted:
Published:
DOI: https://doi.org/10.1007/s12032-022-01824-7