NY-ESO-1-specific redirected T cells with endogenous TCR knockdown mediate tumor response and cytokine release syndrome

Mikiya Ishihara, Shigehisa Kitano, Shinichi Kageyama, Yoshihiro Miyahara, Noboru Yamamoto, Hidefumi Kato, Hideyuki Mishima, Hiroyoshi Hattori, Takeru Funakoshi, Takashi Kojima, Tetsuro Sasada, Eiichi Sato, Sachiko Okamoto, Daisuke Tomura, Ikuei Nukaya, Hideto Chono, Junichi Mineno, Muhammad Faris Kairi, Phuong Diem Hoang Nguyen, Yannick Simoni, Alessandra Nardin, Evan Newell, Michael Fehlings, Hiroaki Ikeda, Takashi Watanabe, Hiroshi Shiku, Mikiya Ishihara, Shigehisa Kitano, Shinichi Kageyama, Yoshihiro Miyahara, Noboru Yamamoto, Hidefumi Kato, Hideyuki Mishima, Hiroyoshi Hattori, Takeru Funakoshi, Takashi Kojima, Tetsuro Sasada, Eiichi Sato, Sachiko Okamoto, Daisuke Tomura, Ikuei Nukaya, Hideto Chono, Junichi Mineno, Muhammad Faris Kairi, Phuong Diem Hoang Nguyen, Yannick Simoni, Alessandra Nardin, Evan Newell, Michael Fehlings, Hiroaki Ikeda, Takashi Watanabe, Hiroshi Shiku

Abstract

Background: Because of the shortage of ideal cell surface antigens, the development of T-cell receptor (TCR)-engineered T cells (TCR-T) that target intracellular antigens such as NY-ESO-1 is a promising approach for treating patients with solid tumors. However, endogenous TCRs in vector-transduced T cells have been suggested to impair cell-surface expression of transduced TCR while generating mispaired TCRs that can become self-reactive.

Methods: We conducted a first-in-human phase I clinical trial with the TCR-transduced T-cell product (TBI-1301) in patients with NY-ESO-1-expressing solid tumors. In manufacturing TCR-T cells, we used a novel affinity-enhanced NY-ESO-1-specific TCR that was transduced by a retroviral vector that enables siRNA (small interfering RNA)-mediated silencing of endogenous TCR. The patients were divided into two cohorts. Cohort 1 was given a dose of 5×108 cells (whole cells including TCR-T cells) preconditioned with 1500 mg/m2 cyclophosphamide. Cohort 2 was given 5× 109 cells preconditioned with 1500 mg/m2 cyclophosphamide.

Results: In vitro study showed that both the CD8+ and CD4+ T fractions of TCR-T cells exhibited cytotoxic effects against NY-ESO-1-expressing tumor cells. Three patients and six patients were allocated to cohort 1 and cohort 2, respectively. Three of the six patients who received 5×109 cells showed tumor response, while three patients developed early-onset cytokine release syndrome (CRS). One of the patients developed a grade 3 lung injury associated with the infiltration of the TCR-T cells. No siRNA-related adverse events other than CRS were observed. Cytokines including interleukin 6 I and monocyte chemotactic protein-1/chemokine (C-C motif) ligand (CCL2)increased in the sera of patients with CRS. In vitro analysis showed these cytokines were not secreted from the T cells infused. A significant fraction of the manufactured T cells in patients with CRS was found to express either CD244, CD39, or both at high levels.

Conclusions: The trial showed that endogenous TCR-silenced and affinity-enhanced NY-ESO-1 TCR-T cells were safely administered except for grade 3 lung injury. The TCR-T cell infusion exhibited significant tumor response and early-onset CRS in patients with tumors that express NY-ESO-1 at high levels. The differentiation properties of the manufactured T cells may be prognostic for TCR-T-related CRS.

Trial registration number: NCT02366546.

Keywords: cell engineering; clinical trials as topic; cytokines; immunotherapy, adoptive.

Conflict of interest statement

Competing interests: SO, DT, IN, HC, and JM are employees of Takara Bio. The Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, to which SKa, YM, TW, and HS belonged, was funded by Takara Bio. MFK, PDHN, YS, AN, EN, and MF are employees or consultants of ImmunoScape. MI received honoraria from Chugai, Eisai, MSD, Ono Pharmaceutical, Daiichi Sankyo, and Eli Lilly. As a potential conflict of interest, SKi and NY received research grants from Takara Bio.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Preclinical study of G50A+A51E T cell receptor (TCR)-transduced T cells. (A) Construct of retroviral vector, pMS3-NY-ESO-1-siTCR. siRNAs, siRNA sequences for endogenous TCR-α and TCR-β; EU, untranslated region 5 of human EF1a; coTCRβ, codon-optimized TCR-β gene sequences for NY-ESO-1; 2A, 2A peptide; coTCRα, codon-optimized TCR-α gene sequences for NY-ESO-1; pA, polyA signal addition; Ψ+, packaging signal; 5'-LTR and R region of 3'-LTR are Moloney murine sarcoma virus-derived, U3 region of 3’-LTR is PCC4-cell-passaged myeloproliferative sarcoma virus-derived. The coTCRα and coTCRβ are formed by codon optimization and are not affected by small interfering RNAs (siRNAs). (B) Transduction of TCR gene to CD8+ T cells and CD4+ T cells. Using the vector of pMS3-NY-ESO-1-siTCR, the TCR gene was transduced to peripheral blood of a healthy volunteer and cultured for 10 days. CD8+ or CD4+ T cells were stained with the NY-ESO-1 peptidemajor histocompatibity complex (MHC) tetramer. 87.6% of CD8+ T cells and 89.0% of CD4+ T cells were positive for the tetramer. (C) Suppression of endogenous TCR-α and TCR-β chain expression by siTCR. RNAs were extracted from 5×106 cells of TCR-gene modified T cells (GMCs) and the T cells that were not gene-transduced (NGMCs). Reverse transcription PCR for endogenous TCR-α and TCR-β chains was performed. 35.3% (±7.9%) and 33.8% (±6.2%) reductions in TCR-α and TCR-β chain expression, respectively. Three experiments using T cells from healthy volunteers were performed in each TCR chain. (D) Interferon gamma (IFN-γ) ELISPOT assay of the NY-ESO-1-TCR-gene transduced CD8+ and CD4+ T cells from healthy volunteers. After TCR-gene transduction and 10-day culture, CD8+ T cells and CD4+ T cells were separated using microbeads. SK-MEL37 is a NY-ESO-1(+) and HLA-A*02:01(+) melanoma cell line. 397mel is a NY-ESO-1(+) and HLA-A*02:01(−) melanoma cell line. HLA-A*02:01-gene or HLA-A*02:06-gene transduced 397mel cells were used as target cells. (E) IFN-γ ELISPOT assay of NY-ESO-1-TCR (G50A+A51E)-gene transduced T cells targeting tumor cells. 1G4-clone is a wild-type (WT) TCR whose affinity is not modified, and G50A+A51E is the affinity-enhanced TCR that was used in the clinical trial. SK-MEL37 cells, 397mel cells, and HLA-A*02:01-transfected 397mel cells were used as target cells. (F) Cytotoxicity assay of G50A+A51E TCR-gene transduced CD8+ and CD4+ T cells. Cytotoxicity was determined by 51Cr-release assay. T cells were transduced with the NY-ESO-1-TCR(G50A+A51E) transgene, and CD8+ and CD4+ T cells were enriched after sorting with NY-ESO-1/HLA-A*02:01-tetramer. As target cells, T2 cells pulsed with NY-ESO-1 peptide (ESO1-T2), T2 cells pulsed with MAGE-A4 peptide (MAGE-T2), melanoma cell line (NW-MEL-38, NY-ESO-1+/HLA-A*02:01+) and colon tumor cell line (HCT116, NY-ESO-1−/HLA-A*02:01+) were used. 51Cr-labeled target cells were cocultured with CD8+ and CD4+ T cells for 12 hours. (G) Cross-reactivity of the NY-ESO-1-TCR with other peptides with amino acid sequences similar to that of the NY-ESO-1 peptide. WT and nine similar amino acid sequences. The WT NY-ESO-1/HLA*02:01-restricted peptide, SLLMWITQC, is listed at the top. Nine peptides are prepared by replacing each amino acid with alanine. (H) Nine peptides were synthesized, S1A, L2A, L3A, M4A, W5A, I6A, T7A, Q8A, C9A, by replacing each amino acid with alanine. TCR-T cells that responded to WT peptide and the synthesized nine peptides were analyzed by intracellular IFN-γ staining. The response to WT peptide was a positive control. Four amino acids (M, W, I, Q) were evaluated to be essential to TCR recognition. (I) Intracellular IFN-γ staining of TCR-T cells responding to the 11 analogous peptide-pulsed target cells. The 11 peptides are derived from known human proteins. YLNMWITTC, taste receptor type 2 member 8; GMDMWSTQD, claudin-18 isoform 1 precursor; QYLMWLTQK, phosphatidylinositol 3-kinase regulatory subunit alpha; AMVAWVTQE, serine/threonine-protein phosphatase 6 regulatory subunit 1; FQDMWITAA, actin-related protein T3; FQQMWITKQ, actin, alpha skeletal muscle; GQKMWITNG, medium-chain specific acyl-CoA dehydrogenase, mitochondrial; GQPLWITAT, ectonucleotide pyrophosphatase/phosphodiesterase family; YVIMWITYM, Niemann-Pick C1 protein precursor; LVGLWITQW, phosphatidylcholine:ceramide cholinephosphotransferase 2; KIALWITYG, gasdermin-C. The response to WT peptide was a positive control (figure 1H).
Figure 2
Figure 2
Clinical course of patient no 8. (A) Clinical course of patient no 8, who developed grade 3 lung injury after grade 2 cytokine release syndrome. (B) Chest X-ray (days 2, 9, and 14) and chest CT scan (days 5 and 9) of patient no 8. (C) Flow cytometry analysis of NY-ESO-1 tetramer–positive T cells in peripheral blood (PB) and bronchoalveolar lavage fluid (BALF) on day 10. Mononuclear cells were separated and were ex vivo stained with anti-CD8 antibody and NY-ESO-1 peptide/MHC tetramer. 25.0% of CD8+ T cells in PB were tetramer positive, and 53.6% of CD8+ T cells in BALF were also positive. TCR-T, T-cell receptor-engineered T cells.
Figure 3
Figure 3
Tumor responses after TBI-1301 infusion. (A) Waterfall plot of seven patients. Seven of the nine patients had measurable lesions that could be evaluated for tumor response. Patient nos 2 and 14 had no measurable tumor lesions. Patient nos 7, 8, and 16 had synovial sarcoma. Patient nos 8, 9, and 16 developed CRS. (B) Swimmer plot for the nine patients treated with TBI-1301. Patient no 2 had no measurable lesion. CRS, cytokine release syndrome; TBI-1301, TCR-transduced T cell product; TCR-T, T-cell receptor-engineered T cells.
Figure 4
Figure 4
Chronological serum cytokine level in six patients who received 5×109 TBI-1301 cells. (A) Serum IL-6, IFN-γ, TNF-α, and IL-2 levels in three patients with CRS. (B) Cytokine levels in three patients without CRS. (C) Multiple cytokines levels measured by Bio-Plex Pro human cytokine screening 48-Plex panel in three patients with CRS. (D) Cytokine levels in two patients without CRS. The serum sample from patient no 7 was unavailable. (E) Cytokine levels in the supernatants of TBI-1301 products from six patients after culturing with NY-ESO-1 peptide–pulsed target cells. Data from patients with CRS are shown on the left and those from patients without CRS are on the right. (F) Cytokine profiles responding to NY-ESO-1-peptide stimulation of TBI-1301 manufactured products in patients with and without CRS. CRS, cytokine release syndrome; IFN-γ, interferon gamma; IL, interleukin; TBI-1301, TCR-transduced T cell product; TNF-α, tumor necrosis factor alpha.
Figure 5
Figure 5
T cell phenotypes and high-dimensional immune profiles of TBI-1301 manufactured products. (A) T cell phenotypes of TBI-1301 manufactured products. Three patients, nos 8, 9, and 16, with CRS had significantly more frequent CD8+ T cells with effector memory phenotypes (p=0.008). Stem cell-like memory T cells, CD45RA+/CCR7+; central memory T cells, CD45RA−/CCR7+; effector memory T cells, CD45RA−/CCR7−; terminally differentiated T cells, CD45RA+/CCR7−. (B and C) High-dimensional immune profiles of NY-ESO-1 tetramer+ CD8+ T cells in the manufactured T cell product. CD244 and CD39 levels in NY-ESO-1-specific CD8+ T cells among the manufactured T cell products. Nos 8, 9, and 16 developed CRS, whereas nos 7, 14, and 15 did not. (D) Cells located in either cluster A (area enclosed with red line), cluster B (area enclosed with orange line), or both in infusion products of patients with CRS are not seen in infusion products of patients without CRS. No statistical difference was found between the two groups. (E) Clusters A and B are characterized by high levels of CD244 and CD39 and a lack of CCR7. CRS, cytokine release syndrome; TBI-1301, TCR-transduced T cell product; UMAP, uniform manifold approximation and projection.

References

    1. Maude SL, Teachey DT, Porter DL, et al. . CD19-targeted chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Blood 2015;125:4017–23. 10.1182/blood-2014-12-580068
    1. Neelapu SS, Locke FL, Bartlett NL, et al. . Axicabtagene Ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med 2017;377:2531–44. 10.1056/NEJMoa1707447
    1. Chen YT, Scanlan MJ, Sahin U, et al. . A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening. Proc Natl Acad Sci U S A 1997;94:1914–8. 10.1073/pnas.94.5.1914
    1. Jungbluth AA, Chen YT, Stockert E, et al. . Immunohistochemical analysis of NY-ESO-1 antigen expression in normal and malignant human tissues. Int J Cancer 2001;92:856–60. 10.1002/ijc.1282
    1. Schmid DA, Irving MB, Posevitz V, et al. . Evidence for a TCR affinity threshold delimiting maximal CD8 T cell function. J Immunol 2010;184:4936–46. 10.4049/jimmunol.1000173
    1. Robbins PF, Li YF, El-Gamil M, et al. . Single and dual amino acid substitutions in TCR CDRs can enhance antigen-specific T cell functions. J Immunol 2008;180:6116–31. 10.4049/jimmunol.180.9.6116
    1. Robbins PF, Morgan RA, Feldman SA, 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;29:917–24. 10.1200/JCO.2010.32.2537
    1. Robbins PF, Kassim SH, Tran TLN, et al. . A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin Cancer Res 2015;21:1019–27. 10.1158/1078-0432.CCR-14-2708
    1. Sommermeyer D, Neudorfer J, Weinhold M, et al. . Designer T cells by T cell receptor replacement. Eur J Immunol 2006;36:3052–9. 10.1002/eji.200636539
    1. Heemskerk MHM, Hagedoorn RS, van der Hoorn MAWG, et al. . Efficiency of T-cell receptor expression in dual-specific T cells is controlled by the intrinsic qualities of the TCR chains within the TCR-CD3 complex. Blood 2007;109:235–43. 10.1182/blood-2006-03-013318
    1. Bendle GM, Linnemann C, Hooijkaas AI, et al. . Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nat Med 2010;16:565–70. 10.1038/nm.2128
    1. Schober K, Müller TR, Gökmen F, et al. . Orthotopic replacement of T-cell receptor α- and β-chains with preservation of near-physiological T-cell function. Nat Biomed Eng 2019;3:974–84. 10.1038/s41551-019-0409-0
    1. Stadtmauer EA, Fraietta JA, Davis MM, et al. . CRISPR-engineered T cells in patients with refractory cancer. Science 2020;367:eaba7365. 10.1126/science.aba7365
    1. Okamoto S, Mineno J, Ikeda H, et al. . Improved expression and reactivity of transduced tumor-specific TCRs in human lymphocytes by specific silencing of endogenous TCR. Cancer Res 2009;69:9003–11. 10.1158/0008-5472.CAN-09-1450
    1. Okamoto S, Amaishi Y, Goto Y, et al. . A promising vector for TCR gene therapy: differential effect of siRNA, 2A peptide, and disulfide bond on the introduced TCR expression. Mol Ther Nucleic Acids 2012;1:e63. 10.1038/mtna.2012.52
    1. Ochi T, Fujiwara H, Okamoto S, et al. . Novel adoptive T-cell immunotherapy using a WT1-specific TCR vector encoding silencers for endogenous TCRs shows marked antileukemia reactivity and safety. Blood 2011;118:1495–503. 10.1182/blood-2011-02-337089
    1. Miyahara Y, Naota H, Wang L, et al. . Determination of cellularly processed HLA-A2402-restricted novel CTL epitopes derived from two cancer germ line genes, MAGE-A4 and SAGE. Clin Cancer Res 2005;11:5581–9. 10.1158/1078-0432.CCR-04-2585
    1. Altman JD, Moss PA, Goulder PJ, et al. . Phenotypic analysis of antigen-specific T lymphocytes. Science 1996;274:94–6. 10.1126/science.274.5284.94
    1. Lee DW, Gardner R, Porter DL, et al. . Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014;124:188–95. 10.1182/blood-2014-05-552729
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47. 10.1016/j.ejca.2008.10.026
    1. Fujii K, Miyahara Y, Harada N, et al. . Identification of an immunogenic neo-epitope encoded by mouse sarcoma using CXCR3 ligand mRNAs as sensors. Oncoimmunology 2017;6:e1306617. 10.1080/2162402X.2017.1306617
    1. Gattinoni L, Lugli E, Ji Y, et al. . A human memory T cell subset with stem cell-like properties. Nat Med 2011;17:1290–7. 10.1038/nm.2446
    1. Newell EW, Sigal N, Nair N, et al. . Combinatorial tetramer staining and mass cytometry analysis facilitate T-cell epitope mapping and characterization. Nat Biotechnol 2013;31:623–9. 10.1038/nbt.2593
    1. Fehlings M, Jhunjhunwala S, Kowanetz M, et al. . Late-differentiated effector neoantigen-specific CD8+ T cells are enriched in peripheral blood of non-small cell lung carcinoma patients responding to atezolizumab treatment. J Immunother Cancer 2019;7:1–15. 10.1186/s40425-019-0695-9
    1. Becht E, McInnes L, Healy J, et al. . Dimensionality reduction for visualizing single-cell data using UMAP. Nat Biotechnol 2018:4314. 10.1038/nbt.4314
    1. D'Angelo SP, Melchiori L, Merchant MS, et al. . Antitumor activity associated with prolonged persistence of adoptively transferred NY-ESO-1c259T cells in synovial sarcoma. Cancer Discov 2018;8:944–57. 10.1158/-17-1417
    1. Morgan RA, Chinnasamy N, Abate-Daga D, et al. . Cancer regression and neurological toxicity following anti-MAGE-A3 TCR gene therapy. J Immunother 2013;36:133–51. 10.1097/CJI.0b013e3182829903
    1. Linette GP, Stadtmauer EA, Maus MV, et al. . Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood 2013;122:863–71. 10.1182/blood-2013-03-490565
    1. Cameron BJ, Gerry AB, Dukes J, et al. . Identification of a titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med 2013;5:197ra103. 10.1126/scitranslmed.3006034
    1. Shah NN, Highfill SL, Shalabi H, et al. . CD4/CD8 T-cell selection affects chimeric antigen receptor (CAR) T-cell potency and toxicity: updated results from a phase I anti-CD22 CAR T-cell trial. J Clin Oncol 2020;38:1938–50. 10.1200/JCO.19.03279
    1. Turtle CJ, Hanafi L, Berger C. CD19 CAR – T cells of defined CD4 +: CD8 + composition in adult B cell all patients. J Clin Invest 2016;1:1–16.
    1. Ferris ST, Durai V, Wu R, et al. . cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity. Nature 2020;584:624–9. 10.1038/s41586-020-2611-3
    1. Cachot A, Bilous M, Liu Y-C, et al. . Tumor-specific cytolytic CD4 T cells mediate immunity against human cancer. Sci Adv 2021;7:eabe3348. 10.1126/sciadv.abe3348
    1. Imai N, Tawara I, Yamane M, et al. . CD4+ T cells support polyfunctionality of cytotoxic CD8+ T cells with memory potential in immunological control of tumor. Cancer Sci 2020;111:1958–68. 10.1111/cas.14420
    1. Giavridis T, van der Stegen SJC, Eyquem J, et al. . CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med 2018;24:731–8. 10.1038/s41591-018-0041-7
    1. Norelli M, Camisa B, Barbiera G, et al. . Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med 2018;24:410.1038/s41591-018-0036-4
    1. Butler MO, Sotov V, Saibil S, et al. . Adoptive T cell therapy with TBI-1301 results in gene-engineered T cell persistence and anti-tumour responses in patients with NY-ESO-1 expressing solid tumours. Ann Oncol 2019;30:v481.1010.1093/annonc/mdz253.009
    1. Harper J, Adams KJ, Bossi G, et al. . An approved in vitro approach to preclinical safety and efficacy evaluation of engineered T cell receptor anti-CD3 bispecific (ImmTAC) molecules. PLoS One 2018;13:e0205491. 10.1371/journal.pone.0205491
    1. Coles CH, Mulvaney RM, Malla S, et al. . TCRs with distinct specificity profiles use different binding modes to engage an identical Peptide-HLA complex. J Immunol 2020;204:1943–53. 10.4049/jimmunol.1900915
    1. Speiser DE, Colonna M, Ayyoub M, et al. . The activatory receptor 2B4 is expressed in vivo by human CD8+ effector alpha beta T cells. J Immunol 2001;167:6165–70. 10.4049/jimmunol.167.11.6165
    1. Kis-Toth K, Comte D, Karampetsou MP, et al. . Selective loss of signaling lymphocytic activation molecule family member 4-Positive CD8+ T cells contributes to the decreased cytotoxic cell activity in systemic lupus erythematosus. Arthritis Rheumatol 2016;68:164–73. 10.1002/art.39410
    1. Gupta PK, Godec J, Wolski D, et al. . Cd39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog 2015;11:e1005177. 10.1371/journal.ppat.1005177
    1. Canale FP, Ramello MC, Núñez N, et al. . CD39 expression defines cell exhaustion in tumor-infiltrating CD8+ T cells. Cancer Res 2018;78:115–28. 10.1158/0008-5472.CAN-16-2684
    1. Simoni Y, Becht E, Fehlings M, et al. . Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 2018;557:575–9. 10.1038/s41586-018-0130-2
    1. Duhen T, Duhen R, Montler R, et al. . Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun 2018;9:2724. 10.1038/s41467-018-05072-0
    1. Kortekaas KE, Santegoets SJ, Sturm G, et al. . CD39 identifies the CD4+tumor-specific T-cell population in human cancer. Cancer Immunol Res 2020;8:1311–21. 10.1158/2326-6066.CIR-20-0270

Source: PubMed

3
Subscribe