Optimized T-cell receptor-mimic chimeric antigen receptor T cells directed toward the intracellular Wilms Tumor 1 antigen

S Rafiq, T J Purdon, A F Daniyan, M Koneru, T Dao, C Liu, D A Scheinberg, R J Brentjens, S Rafiq, T J Purdon, A F Daniyan, M Koneru, T Dao, C Liu, D A Scheinberg, R J Brentjens

Abstract

CD19-directed chimeric antigen receptor (CAR) T cells are clinically effective in a limited set of leukemia patients. However, CAR T-cell therapy thus far has been largely restricted to targeting extracellular tumor-associated antigens (TAA). Herein, we report a T-cell receptor-mimic (TCRm) CAR, termed WT1-28z, that is reactive to a peptide portion of the intracellular onco-protein Wilms Tumor 1(WT1), as it is expressed on the surface of the tumor cell in the context of HLA-A*02:01. T cells modified to express WT1-28z specifically targeted and lysed HLA-A*02:01+ WT1+ tumors and enhanced survival of mice engrafted with HLA-A*02:01+, WT1+ leukemia or ovarian tumors. This in vivo functional validation of TCRm CAR T cells provides the proof-of-concept necessary to expand the range of TAA that can be effectively targeted for immunotherapy to include attractive intracellular targets, and may hold great potential to expand on the success of CAR T-cell therapy.

Conflict of interest statement

CONFLICT OF INTEREST

RJB is a co-founder, stockholder and consultant for Juno Therapeutics Inc. DAS and TD are inventors of TCRm technology owned by MSKCC, licensed to Novartis.

Figures

Figure 1
Figure 1
Generation and validation of WT1-28z CAR T cells. (a) Schematic representation of the WT1-28z constructs. WT1/HLA-A*02:01-specific scFv derived from the heavy (VH) and light (VL) chain variable regions of the ESK1 antibody; CD28: human CD28 transmembrane and cytoplasmic signaling domains; z-chain: human TCR zeta chain cytoplasmic signaling domain; black box: κ leader sequence; gray box: (Gly4Ser)3 linker. (b) Flow cytometry histograms depicting the expression of WT1-28z in retrovirally transduced into primary human T cells, as detected by binding to a fluorescently-labeled WT1/HLA-A*02:01 Tetramer (representative figure of n > 10). (c) WT1-28z CAR T cells are more toxic than control irrelevant antigen-specific control CAR T cells 4H11-28z or 19-28z CAR T cells against AML-14, BV173 and OVCAR3 in standard 51Cr release assays (representative figures, n =3 for each cell line). (d) WT1-28z CAR T cells when co-cultured with AML-14, BV173, or OVCAR3 cell lines for 24 h have significantly enhanced release of IFN-γ (*P = <0.0001, 0.03, or 0.0013 respectively) and IL-2 (*P ≤ 0.0001, 0.013, or <0.0001 respectively), as compared with control 4H11-28z or 19-28z CAR T cells (n =3 for each cell line). (e) Standard 51Cr release assays with antigen-specific WT1-28z CAR T cells showed enhanced cytotoxicity against primary AML samples as compared with a control irrelevant antigen-specific 4H11-28z CAR T cells (n =5) (*P =0.029). (f) WT1-28z CAR T cells have significantly enhanced release of IFN-γ and IL-2 (*P = 0.04) when co-cultured with AML patient samples for 24 h, as compared with 4H11-28z CAR T cells (n =3).
Figure 2
Figure 2
Generation and validation of WT1-28z/IL-12 CAR T cells. (a) Schematic representation of the WT1-28z/IL-12 CAR constructs. WT1/HLA-A*02:01-specific scFv derived from heavy (VH) and light (VL) chain variable regions of the ESK1 antibody; CD28: human CD28 transmembrane and cytoplasmic signaling domains; z-chain: human TCR zeta chain cytoplasmic signaling domain; flexi human IL-12 (hIL-12 f); LTR: 50 and 30 long terminal repeat; black box: κ leader sequence; gray box: (Gly4Ser)3 linker. (b) IL-12 expression by viral producer cell lines stably transduced with CAR constructs show that only 293Galv9 cells transduced with WT1-28z/IL-12 express IL-12 (n =3 independent experiments) (P =0.01). (c) The IL-12 produced by the WT1-28z/IL-12 CAR in 293Galv9 is functional, as determined by increases in levels of IFN-γ produced by peripheral blood mononuclear cell cultured in supernatant (n =3 independent experiments) (P =0.03). (d) Flow cytometry histograms depicting the expression of WT1-28z/IL-12 CARs in retrovirally transduced primary human T cells, as detected by binding to a fluorescently-labeled WT1/HLA-A*02:01 tetramer (representative figure of n > 10). (e) WT1-28z/IL-12 CAR T cells have significantly enhanced release of IFN-γ (*P =0.008, 0.01, respectively) and IL-2 (*P = 0.002, 0.002, respectively), when co-cultured with Set2 cells for 24 hours, as compared to 19-28z or 19-28z/IL-12 CAR T cells (n =4). (f) WT1-28z/IL-12 CAR T cells proliferate after co-culture with Set2 cells (n =4).
Figure 3
Figure 3
WT1-28z and WT1-28z/IL-12 CAR T cells are specific to WT1/HLA-A*02:01. (a and b) WT1-28z or WT1-28z/IL-12 CAR T cells are significantly more toxic than control irrelevant antigen-specific CAR T cells 4H11-28z and 4H11-28z/IL-12 CAR T cells against Set2 and KARPAS-620 in standard 51Cr release assays (representative figures, n =3 for each cell line). WT1-28z significantly more lysed Set2 cells than 4H11-28z CAR T cells at all the effector to target ratios tested (P ≤ 0.005). WT1-28z/IL-12 significantly more lysed Set2 cells than 4H11-28z/IL-12 CAR T cells at 40:1, 20:1 and 10:1 effector to target ratios tested (P<0.0006). WT1-28z significantly more lysed KARPAS-620 than 4H11-28z CAR T cells at all the effector to target ratios tested (P<0.007). WT1-28z/IL-12 significantly more lysed KARPAS-620 than 4H11-28z/IL-12 CAR T cells at 40:1, 20:1, and 10:1 effector to target ratios tested (P<0.0003). (c) WT1-28z and WT1-28z/IL-12 CAR T cells show no toxicity toward the HLA-A*02:01-positive, WT1-negative cell lines SKLY-16 compared with control 4H11-28z or 4H11-28z/IL-12 CAR T cells in standard 51Cr release assays (n =3). (d) WT1-28z and WT1-28z/IL-12 CAR T cells show no toxicity toward the WT1-positive, HLA-A*02:01-negative cell line HL-60 as compared with control 4H11-28z or 4H11-28z/IL-12 CAR T cells in standard 51Cr release assays (n =3).
Figure 4
Figure 4
WT1-28z/IL-12 CAR T cells eradicates established tumors in a human xenograft mouse models. (a) SCID/Beige mice were injected i.v. with 5E6 Set2 cells and treated with i.v. injection of 2E7 CAR-positive primary human T cells 7 days post-tumor injection. WT1-28z CAR T cells significantly improved survival of mice compared to untreated mice (*P =0.006) or mice treated with control 4H11-28z CAR T cells (*P =0.01). WT1-28z/IL-12 CAR T cells had significant survival over mice treated with 4H11-28z/IL-12 CAR T cells (*P =0.004). (n =3 separate experiments). (b) SCID/Beige mice were i.v. injected with 5E6 Set2 cells and treated with i.v. injection of 2E7 CAR-positive primary human T cells 14 days post-tumor injection. WT1-28z/IL-12 CAR T cells significantly enhanced survival of mice in a more established disease model compared with WT1-28z CAR T cells (*P = 0.006). (c) SCID/Beige mice were i.p. injected with 1E7 OVCAR3 cells and treated 41 days post-tumor injection with i.p. injection of 5E6 CAR-positive primary human T cells. WT1-28z CAR T cells mediated significantly enhanced survival compared with treatment with control 19-28z CAR T cells (*P =0.009) or untreated mice (*P =0.0034). A single dose of WT1-28z/IL-12 CAR T cells mediated significantly enhanced survival compared with treatment with control 19-28z/IL-12 CAR T cells (*P ≤ 0.0001). Mice treated with WT1-28z/IL-12 CAR T cells exhibited enhanced survival as compared with WT1-28z CAR T cells, but this difference was not statistically significant (P =0.23) (n =2 independent experiments). (d) Bioluminescent imaging of mice on day 58 after OVCAR3 tumor injection show eradication of tumor in mice treated with WT1-28z or WT1-28z/IL-12 CAR T cells.
Figure 5
Figure 5
Differential gene expression of WT1-28z/IL-12 CAR T cells compared with WT1-28z CAR T cells when stimulated with OVCAR3 tumor cells for 48 hs. (a) Heat map of statistically significant (P ≤ 0.05) differential gene expression of cytokines between four normal donor T cells expressing WT1-28z/IL-12 CAR or WT1-28z CAR. (b) Heat map of statistically significant (P ≤ 0.05) differential expression of genes involved in chemokine or chemotaxis functions between four normal donor T cells expressing WT1-28z/IL-12 CAR or WT1-28z CAR. (c) Heat map of statistically significant (P ≤ 0.05) genes showing a distinct transcriptional profile between four normal donor T cells expressing WT1-28z/IL-12 CAR or WT1-28z CAR.

References

    1. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5:177ra38.
    1. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517.
    1. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385:517–528.
    1. Brentjens RJ, Riviere I, Park JH, Davila ML, Wang X, Stefanski J, et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. 2011;118:4817–4828.
    1. Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7:303ra139.
    1. Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011;121:1822–1826.
    1. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M, et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. 2015;33:540–549.
    1. Wang X, Popplewell LL, Wagner JR, Naranjo A, Blanchard MS, Mott MR, et al. Phase I studies of central-memory-derived CD19 CAR T cell therapy following autologous HSCT in patients with B-cell NHL. Blood. 2016;127:2980–2990.
    1. Jackson HJ, Rafiq S, Brentjens RJ. Driving CAR T cells forward. Nat Rev Clin Oncol. 2016;13:370–383.
    1. Sugiyama H. WT1 (Wilms’ tumor gene 1): biology and cancer immunotherapy. Jpn J Clin Oncol. 2010;40:377–387.
    1. Hohenstein P, Hastie ND. The many facets of the Wilms’ tumour gene, WT1. Hum Mol Genet. 2006;15:R196–R201.
    1. Morrison AA, Viney RL, Ladomery MR. The post-transcriptional roles of WT1, a multifunctional zinc-finger protein. Biochim Biophys Acta. 2008;1785:55–62.
    1. Davies R, Moore A, Schedl A, Bratt E, Miyahawa K, Ladomery M, et al. Multiple roles for the Wilms’ tumor suppressor, WT1. Cancer Res. 1999;59:1747s–1750s. discussion 51s.
    1. Di Stasi A, Jimenez AM, Minagawa K, Al-Obaidi M, Rezvani K. Review of the results of WT1 peptide vaccination strategies for myelodysplastic syndromes and acute myeloid leukemia from nine different studies. Front Immunol. 2015;6:36.
    1. Inoue K, Sugiyama H, Ogawa H, Nakagawa M, Yamagami T, Miwa H, et al. WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood. 1994;84:3071–3079.
    1. Ogawa H, Tamaki H, Ikegame K, Soma T, Kawakami M, Tsuboi A, et al. The usefulness of monitoring WT1 gene transcripts for the prediction and management of relapse following allogeneic stem cell transplantation in acute type leukemia. Blood. 2003;101:1698–1704.
    1. Dao T, Scheinberg DA. Peptide vaccines for myeloid leukaemias. Best Pract Res Clin Haematol. 2008;21:391–404.
    1. Maslak PG, Dao T, Krug LM, Chanel S, Korontsvit T, Zakhaleva V, et al. Vaccination with synthetic analog peptides derived from WT1 oncoprotein induces T-cell responses in patients with complete remission from acute myeloid leukemia. Blood. 2010;116:171–179.
    1. Krug LM, Dao T, Brown AB, Maslak P, Travis W, Bekele S, et al. WT1 peptide vaccinations induce CD4 and CD8 T cell immune responses in patients with mesothelioma and non-small cell lung cancer. Cancer Immunol Immunother. 2010;59:1467–1479.
    1. Dao T, Yan S, Veomett N, Pankov D, Zhou L, Korontsvit T, et al. Targeting the intracellular WT1 oncogene product with a therapeutic human antibody. Sci Transl Med. 2013;5:176ra33.
    1. Veomett N, Dao T, Liu H, Xiang J, Pankov D, Dubrovsky L, et al. Therapeutic efficacy of an Fc-enhanced TCR-like antibody to the intracellular WT1 oncoprotein. Clin Cancer Res. 2014;20:4036–4046.
    1. Dubrovsky L, Pankov D, Brea EJ, Dao T, Scott A, Yan S, et al. A TCR-mimic antibody to WT1 bypasses tyrosine kinase inhibitor resistance in human BCR-ABL+ leukemias. Blood. 2014;123:3296–3304.
    1. Dao T, Pankov D, Scott A, Korontsvit T, Zakhaleva V, Xu Y, et al. Therapeutic bispecific T-cell engager antibody targeting the intracellular oncoprotein WT1. Nat Biotechnol. 2015;33:1079–1086.
    1. Fraietta JA, Schwab RD, Maus MV. Improving therapy of chronic lymphocytic leukemia with chimeric antigen receptor T cells. Semin Oncol. 2016;43:291–299.
    1. Pegram HJ, Lee JC, Hayman EG, Imperato GH, Tedder TF, Sadelain M, et al. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood. 2012;119:4133–4141.
    1. Pegram HJ, Purdon TJ, van Leeuwen DG, Curran KJ, Giralt SA, Barker JN, et al. IL-12-secreting CD19-targeted cord blood-derived T cells for the immunotherapy of B-cell acute lymphoblastic leukemia. Leukemia. 2015;29:415–422.
    1. Lasek W, Zagozdzon R, Jakobisiak M. Interleukin 12: still a promising candidate for tumor immunotherapy? Cancer Immunol Immunother. 2014;63:419–435.
    1. Koneru M, Purdon TJ, Spriggs D, Koneru S, Brentjens RJ. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors. Oncoimmunology. 2015;4:e994446.
    1. Ghani K, Wang X, de Campos-Lima PO, Olszewska M, Kamen A, Riviere I, et al. Efficient human hematopoietic cell transduction using RD114- and GALV-pseudotyped retroviral vectors produced in suspension and serum-free media. Hum Gene Ther. 2009;20:966–974.
    1. Chekmasova AA, Rao TD, Nikhamin Y, Park KJ, Levine DA, Spriggs DR, 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;16:3594–3606.
    1. 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;13:5426–5435.
    1. Ferrone CR, Perales MA, Goldberg SM, Somberg CJ, Hirschhorn-Cymerman D, Gregor PD, et al. Adjuvanticity of plasmid DNA encoding cytokines fused to immunoglobulin Fc domains. Clin Cancer Res. 2006;12:5511–5519.
    1. Lund RJ, Loytomaki M, Naumanen T, Dixon C, Chen Z, Ahlfors H, et al. Genome-wide identification of novel genes involved in early Th1 and Th2 cell differentiation. J Immunol. 2007;178:3648–3660.
    1. Szabo SJ, Sullivan BM, Peng SL, Glimcher LH. Molecular mechanisms regulating Th1 immune responses. Annu Rev Immunol. 2003;21:713–758.
    1. Zhao Q, Ahmed M, Tassev DV, Hasan A, Kuo TY, Guo HF, et al. Affinity maturation of T-cell receptor-like antibodies for Wilms tumor 1 peptide greatly enhances therapeutic potential. Leukemia. 2015;29:2238–2247.
    1. Oren R, Hod-Marco M, Haus-Cohen M, Thomas S, Blat D, Duvshani N, et al. Functional comparison of engineered T cells carrying a native TCR versus TCR-like antibody-based chimeric antigen receptors indicates affinity/avidity thresholds. J Immunol. 2014;193:5733–5743.
    1. Long AH, Haso WM, Shern JF, Wanhainen KM, Murgai M, Ingaramo M, et al. 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med. 2015;21:581–590.
    1. Chmielewski M, Kopecky C, Hombach AA, Abken H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 2011;71:5697–5706.
    1. Kerkar SP, Muranski P, Kaiser A, Boni A, Sanchez-Perez L, Yu Z, et al. Tumor-specific CD8+ T cells expressing interleukin-12 eradicate established cancers in lymphodepleted hosts. Cancer Res. 2010;70:6725–6734.
    1. Chinnasamy D, Yu Z, Kerkar SP, Zhang L, Morgan RA, Restifo NP, et al. Local delivery of interleukin-12 using T cells targeting VEGF receptor-2 eradicates multiple vascularized tumors in mice. Clin Cancer Res. 2012;18:1672–1683.
    1. Morgan RA, Chinnasamy N, Abate-Daga D, Gros A, Robbins PF, Zheng Z, et al. Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother. 2013;36:133–151.
    1. 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;17:4550–4557.
    1. Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. J Clin Oncol. 2011;29:917–924.
    1. Chapuis AG, Ragnarsson GB, Nguyen HN, Chaney CN, Pufnock JS, Schmitt TM, et al. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci Transl Med. 2013;5:174ra27.

Source: PubMed

3
購読する