Experimental Study of the In Vitro and In Vivo Functional Activity of NKG2D Chimeric Antigen Receptor

KA Levchuk1, SA Osipova1, AV Onopchenko1, ML Vasyutina1, ER Bulatov2, AKh Valiullina2, ON Demidov1,3,4, AV Petukhov1

1 VA Almazov National Medical Research Center, 2 Akkuratova ul., Saint Petersburg, Russian Federation, 197341

2 Kazan (Privolzhsky) Federal University, 18 Kremlevskaya ul., Kazan, Russian Federation, 420008

3 Institute of Cytology, 4 Tikhoretskii pr-t, Saint Petersburg, Russian Federation, 194064

4 Sirius University of Science and Technology, 1 Olimpiiskii pr-t, Sochi, Russian Federation, 354340

For correspondence: Kseniya Aleksandrovna Levchuk, 2 Akkuratova ul., Saint Petersburg, Russian Federation, 197341; e-mail: levchuk_ka@almazovcentre.ru

For citation: Levchuk KA, Osipova SA, Onopchenko AV, et al. Experimental Study of the In Vitro and In Vivo Functional Activity of NKG2D Chimeric Antigen Receptor. Clinical oncohematology. 2022;15(4):327–39. (In Russ).

DOI: 10.21320/2500-2139-2022-15-4-327-339


ABSTRACT

Aim. To study antitumor cytotoxic effect of CAR-T NKG2D and CAR-T anti-CD19 in vitro and in vivo in order to compare antitumor activity of chimeric antigen receptors (CAR) with different structural and functional properties.

Materials & Methods. CAR constructions were produced by molecular cloning. CAR-T cell populations were obtained by transduction of healthy donor T-lymphocytes with recombinant lentiviral particles coding CAR NKG2D or CD19 target antigen CAR sequences. CAR-T cell proportion was assessed by FusionRed fluorescence and EGFR membrane receptor imaging. Specific in vitro cytotoxic activity of CAR-T effector cells was analyzed by Real-Time Cytotoxicity Assay (RTCA) during co-cultivation with HeLa_CD19 target cell line using xCELLigence. Interferon-γ (IFN-γ) synthesis in vitro and in vivo along with the degree of cytotoxic effect were analyzed by immunoassay of culture medium of co-cultivated effector cells and target cells as well as isolated auto-plasma from the peripheral blood of mice. To assess the in vivo functional activity, CAR-T cell populations were infused into immunodeficient NSG-SGM3 mice (10 000 000 cells/mouse) 12 days after HeLa_CD19 cell injection and confirmation of engraftment and tumor growth. Upon euthanasia, tumors were removed and fixed in paraffin to prepare histological sections. CAR-T cell tumor infiltration was assessed by CD3 antigen immunohistochemical staining.

Results. The highest ligand (molecules MICA, ULBP1/2/3/4/5/6) expression levels were detected in HeLa cell line. The obtained NKG2D CAR-T cells showed a considerable cytotoxic activity against HeLa_CD19 target line (cell index [CI] = 1.27), which was, however, twice as low as that of CAR-T anti-CD19 (CI = 0.60) (= 0.0038). IFN-γ level during co-cultivation of CAR-T anti-CD19 with HeLa_CD19 at the ratio of Е/Т = 1:1 was 64,852 pcg/mL, which was 3.5 times higher than IFN-γ level during co-cultivation of CAR-T NKG2D with HeLa_CD19 (18,635 pcg/mL) (= 0.0360). The degree of tumor infiltration by CAR-T anti-CD19 cells was higher than that by CAR-T NKG2D. The absence of NKG2D proliferating CAR-T cells in mice peripheral blood confirms their low persistence. IFN-γ concentration in mice auto-plasma was 11.89 pcg/mL after CAR-T anti-CD19 infusion and 0.57 pcg/mL after CAR-T NKG2D infusion (= 0.0079). The mean weight of tumor xenografts in experimental groups 10 days after CAR-T anti-CD19 injection was 0.72 g (= 0.0142), after Т-lymphocyte and NKG2D CAR-T cell infusions it was 2.12 g and 1.2 g, respectively.

Conclusion. CAR-T anti-CD19 cells are characterized by more pronounced cytotoxic effect under both in vitro and in vivo experimental conditions compared with CAR-T NKG2D cells. The degree of CAR-T anti-CD19 proliferation and their infiltration in mice xenograft models is considerably higher than the levels reached with NKG2D CAR-T cell injections. A single CAR-T NKG2D injection results only in short-term tumor reduction.

Keywords: CAR-T cell therapy, NKG2D chimeric antigen receptor, co-stimulatory domains, NKG2D ligands, cytotoxic effect, CAR-T infiltration, CAR-T persistence.

Received: June 27, 2022

Accepted: September 12, 2022

Read in PDF

Статистика Plumx английский

REFERENCES

  1. Saez-Borderias A, Guma M, Angulo A, et al. Expression and function of NKG2D in CD4+ T cells specific for human cytomegalovirus. Eur J Immunol. 2006;36(12):3198–206. doi: 10.1002/eji.200636682.
  2. Allez M, Tieng V, Nakazawa A, et al. CD4+NKG2D+ T cells in Crohn’s disease mediate inflammatory and cyto-toxic responses through MICA interactions. Gastroenterology. 2007;132(7):2346–58. doi: 10.1053/j.gastro.2007.03.025.
  3. Dai Z, Turtle CJ, Booth GC, et al. Normally occurring NKG2D+CD4+ T cells are immunosuppressive and inversely correlated with disease activity in juvenile-onset lupus. J Exp Med. 2009;206(4):793–805. doi: 10.1084/jem.20081648.
  4. Raulet DH. Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol. 2003;3(10):781–90. doi: 10.1038/nri1199.
  5. Le Bert N, Gasser S. Advances in NKG2D ligand recognition and responses by NK cells. Immunol Cell Biol. 2014;92(3):230–6. doi: 10.1038/icb.2013.111.
  6. Rabinovich B, Li J, Wolfson M, et al. NKG2D splice variants: a reexamination of adaptor molecule associations. Immunogenetics. 2006;58(2–3):81–8. doi: 10.1007/s00251-005-0078-x.
  7. Lanier LL. DAP10- and DAP12-associated receptors in innate immunity. Immunol Rev. 2009;227(1):150–60. doi: 10.1111/j.1600-065X.2008.00720.x.
  8. Raulet DH, Gasser S, Gowen BG, et al. Regulation of ligands for the NKG2D activating receptor. Annu Rev Immunol. 2013;31(1):413–41. doi: 10.1146/annurev-immunol-032712-095951.
  9. Luo QZ, Lin L, Gong Z, et al. Positive association of major histocompatibility complex class I chain-related gene A polymorphism with leukemia susceptibility in the people of Han nationality of Southern China. Tissue Antigens. 2011;78(3):178–84. doi: 10.1111/j.1399-0039.2011.01748.x.
  10. Kim H, Byun JE, Yoon SR, et al. SARS-CoV-2 peptides bind to NKG2D and increase NK cell activity. Cell Immunol. 2022;371:104454. doi: 10.1016/j.cellimm.2021.104454.
  11. Farzad F, Yaghoubi N, Jabbari-Azad F, et al. Prognostic Value of Serum MICA Levels as a Marker of Severity in COVID-19 Patients. Immunol Invest. 2022:1–11. doi: 10.1080/08820139.2022.2069035.
  12. Groh V, Rhinehart R, Randolph-Habecker J, et al. Costimulation of CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat Immunol. 2001;2(3):255–60. doi: 10.1038/85321.
  13. Ehrlich LI, Ogasawara K, Hamerman JA, et al. Engagement of NKG2D by cognate ligand or antibody alone is insufficient to mediate costimulation of human and mouse CD8+ T cells. J Immunol. 2005;174(4):1922–31. doi: 10.4049/jimmunol.174.4.1922.
  14. O’Hayre M, Salanga CL, Handel TM, Allen SJ. Chemokines and cancer: migration, intracellular signalling and intercellular communication in the microenvironment. Biochem J. 2008;409(3):635–49. doi: 10.1042/BJ20071493.
  15. Zhang T, Lemoi BA, Sentman CL. Chimeric NK-receptor-bearing T cells mediate antitumor immunotherapy. Blood. 2005;106(5):1544–51. doi: 10.1182/blood-2004-11-4365.
  16. Zhang T, Barber A, Sentman CL. Generation of antitumor responses by genetic modification of primary human T cells with a chimeric NKG2D receptor. Cancer Res. 2006;66(11):5927–33. doi: 10.1158/0008-5472.CAN-06-0130.
  17. Chang YH, Connolly J, Shimasaki N, et al. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013;73(6):1777–86. doi: 10.1158/0008-5472.CAN-12-3558.
  18. Song DG, Ye Q, Santoro S, 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. doi: 10.1089/hum.2012.143.
  19. Lehner M, Gotz G, Proff J, et al. Redirecting T cells to Ewing’s sarcoma family of tumors by a chimeric NKG2D receptor expressed by lentiviral transduction or mRNA transfection. PLoS One. 2012;7(2):e31210. doi: 10.1371/journal.pone.0031210.
  20. Sentman CL, Meehan KR. NKG2D CARs as cell therapy for cancer. Cancer J. 2014;20(2):156–9. doi: 10.1097/PPO.0000000000000029.
  21. Barber A, Zhang T, DeMars LR, et al. Chimeric NKG2D receptor-bearing T cells as immunotherapy for ovarian cancer. Cancer Res. 2007;67(10):5003–8. doi: 10.1158/0008-5472.CAN-06-4047.
  22. Chang YH, Connolly J, Shimasaki N, et al. A chimeric receptor with NKG2D specificity enhances natural killer cell activation and killing of tumor cells. Cancer Res. 2013;73(6):1777–86. doi: 10.1158/0008-5472.CAN-12-3558.
  23. Barber A, Meehan KR, Sentman CL. Treatment of multiple myeloma with adoptively transferred chimeric NKG2D receptor-expressing T cells. Gene Ther. 2011;18(5):509–16. doi: 10.1038/gt.2010.174.
  24. Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: Summary and perspective. J Cell Immunother. 2016;2(2):59–68. doi: 10.1016/j.jocit.2016.08.001.
  25. Ng YY, Tay JCK, Li Z, et al. T Cells Expressing NKG2D CAR with a DAP12 Signaling Domain Stimulate Lower Cytokine Production While Effective in Tumor Eradication. Mol Ther. 2021;29(1):75–85. doi: 10.1016/j.ymthe.2020.08.016.
  26. Fontaine M, Demoulin B, Bornschein S, et al. Next generation NKG2D-based CAR T-cells (CYAD-02): co-expression of a single shRNA targeting MICA and MICB improves cell persistence and anti-tumor efficacy in vivo. Blood. 2019;134(Suppl_1):3931. doi: 10.1182/blood-2019-129998.
  27. Breman E, Demoulin B, Agaugue S, et al. Overcoming Target Driven Fratricide for T Cell Therapy. Front Immunol. 2018;9:2940. doi: 10.3389/fimmu.2018.02940.