Tumor-associated antigen-specific T cells with nivolumab are safe and persist in vivo in relapsed/refractory Hodgkin lymphoma

Hema Dave, Madeline Terpilowski, Mimi Mai, Keri Toner, Melanie Grant, Maja Stanojevic, Christopher Lazarski, Abeer Shibli, Stephanie A Bien, Philip Maglo, Fahmida Hoq, Reuven Schore, Martha Glenn, Boyu Hu, Patrick J Hanley, Richard Ambinder, Catherine M Bollard, Hema Dave, Madeline Terpilowski, Mimi Mai, Keri Toner, Melanie Grant, Maja Stanojevic, Christopher Lazarski, Abeer Shibli, Stephanie A Bien, Philip Maglo, Fahmida Hoq, Reuven Schore, Martha Glenn, Boyu Hu, Patrick J Hanley, Richard Ambinder, Catherine M Bollard

Abstract

Hodgkin lymphoma (HL) Reed Sternberg cells express tumor-associated antigens (TAA) that are potential targets for cellular therapies. We recently demonstrated that TAA-specific T cells (TAA-Ts) targeting WT1, PRAME, and Survivin were safe and associated with prolonged time to progression in solid tumors. Hence, we evaluated whether TAA-Ts when given alone or with nivolumab were safe and could elicit antitumor effects in vivo in patients with relapsed/refractory (r/r) HL. Ten patients were infused with TAA-Ts (8 autologous and 2 allogeneic) for active HL (n = 8) or as adjuvant therapy after hematopoietic stem cell transplant (n = 2). Six patients received nivolumab priming before TAA-Ts and continued until disease progression or unacceptable toxicity. All 10 products recognized 1 or more TAAs and were polyfunctional. Patients were monitored for safety for 6 weeks after the TAA-Ts and for response until disease progression. The infusions were safe with no clear dose-limiting toxicities. Patients receiving TAA-Ts as adjuvant therapy remain in continued remission at 3+ years. Of the 8 patients with active disease, 1 patient had a complete response and 7 had stable disease at 3 months, 3 of whom remain with stable disease at 1 year. Antigen spreading and long-term persistence of TAA-Ts in vivo were observed in responding patients. Nivolumab priming impacted TAA-T recognition and persistence. In conclusion, treatment of patients with r/r HL with TAA-Ts alone or in combination with nivolumab was safe and produced promising results. This trial was registered at www.clinicaltrials.gov as #NCT022039303 and #NCT03843294.

Trial registration: ClinicalTrials.gov NCT03843294 NCT22039303.

© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Phenotype of TAA-T products. (A) Fold expansion (n = 12). (B) Immunophenotype of the TAA-T products (n = 10). (C) Polyclonality of TAA-T as assessed by TCR-Vβ deep sequencing (n = 6). (D) Lack of cytotoxicity of TAA-T (effectors) against non–antigen-pulsed PHA blasts (targets) at an various effector to target ratios (n = 10). (E) Exhaustion markers on TAA-T products (n = 6). PHA, phytohaemagglutinin
Figure 2.
Figure 2.
Functional characterization of TAA-T products. (A) Tumor antigen specificity as measured by IFNγ ELISPOT assay of the 12 infused products after overnight restimulation with overlapping 15mer pepmixes of actin (irrelevant control antigen), WT1, PRAME, and Survivin. (B) Polyfunctionality as assessed by the release of IFNγ and TNFα by CD3+, CD4+, and CD8+ TAA-Ts in response to irrelevant antigen. Actin and TAA (PRAME) shown in a representative dot plot of TAA-T product and summarized for 6 TAA-T products (C). * and ** denote P < .01 for difference between actin and PRAME.
Figure 3.
Figure 3.
Clinical outcomes. Swimmers plot showing the outcome of patients after receiving TAA-Ts with or without nivolumab. (A) Patients in remission at the time of TAA-T infusion. (B) Patients with measurable disease at the time of TAA-T infusion. The timing of nivolumab is indicated by the blue bar in relation to the TAA-T infusion, which is indicated by an asterisk.
Figure 4.
Figure 4.
Recognition of tumor specific antigens over time. Recovery of antigen-specific T cells and antigen spreading as detected by IFNγ ELISPOT assay in patients receiving TAA-T alone in responders (A-B) and nonresponders (C-D). For each figure, reactivity to the targeted antigens WT1, PRAME, and Survivin is shown in panel i and nontargeted antigens MAGE family, SSX2, and SOX2 is in panel ii.
Figure 5.
Figure 5.
Recognition of tumor specific antigens over time. Recovery of antigen-specific T cells and antigen spreading as detected by IFNγ ELISPOT assay in patients receiving TAA-T and nivolumab in responders (A-D) and nonresponders (E-F). For each figure, reactivity to the targeted antigens WT1, PRAME, and Survivin is shown in panel i and nontargeted antigens MAGE family, SSX2, and SOX2 in panel ii.
Figure 6.
Figure 6.
Impact of nivolumab on persistence of functional TAA-T cells. (A) Comparison of T-cell reactivity to the targeted antigens as detected by IFNγ ELISPOT assay after nivolumab priming and before TAA-T infusion and at 3 months between responding (R) and nonresponding patients (NR). (B) Persistence of polyfunctional TAA-T cells over time in responders. (C) Persistence of polyfunctional TAA-T cells over time in nonresponders. (D) Representative plot of patient demonstrating recovery of in vivo polyfunctional CD4+ and CD8+ TAA-T cells secreting both IFNγ and TNFα was detected after brief ex vivo expansion after restimulation with antigens at several follow-up time points.
Figure 7.
Figure 7.
Persistence of new TAA-T clones expanded in the product that were not present at baseline. (A) Using TCRVβ deep sequencing, new clones detected in the TAA-T product were identified and tracked in PB over time. (B) Longitudinal tracking of new clones expanded in product over time for patients receiving TAA-T alone. Multi-institutional Prospective Research of Expanded Multi-antigen Specifically Oriented Lymphocytes for the Treatment of VEry High Risk Hematopoietic Malignancies (RESOLVE) and TAA-T with nivolumab [TAA-T with nivolumab on Phase I Study Utilizing Tumor Associated Antigen Specific T cells (TAA-T) with PD1 Inhibitor Nivolumab for Relapsed/Refractory Lymphoma (SUSTAIN) trial]. (C) Tracking of clones enriched in TAA-T product and not present at baseline in patient 6.

References

    1. Bollard CM, Gottschalk S, Torrano V, et al. . Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J Clin Oncol. 2014;32(8):798-808.
    1. Bollard CM, Tripic T, Cruz CR, et al. . Tumor-specific T-cells engineered to overcome tumor immune evasion induce clinical responses in patients with relapsed Hodgkin lymphoma. J Clin Oncol. 2018;36(11):1128-1139.
    1. McLaughlin LP, Rouce R, Gottschalk S, et al. . EBV/LMP-specific T cells maintain remissions of T- and B-cell EBV lymphomas after allogeneic bone marrow transplantation. Blood. 2018;132(22):2351-2361.
    1. Kochenderfer JN, Somerville RPT, Lu T, et al. . Long-duration complete remissions of diffuse large B cell lymphoma after Anti-CD19 chimeric antigen receptor T cell therapy. Mol Ther. 2017;25(10):2245-2253.
    1. Maude SL. Tisagenlecleucel in pediatric patients with acute lymphoblastic leukemia. Clin Adv Hematol Oncol. 2018;16(10):664-666.
    1. Ramos CA, Ballard B, Zhang H, et al. . Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes. J Clin Invest. 2017;127(9):3462-3471.
    1. Ramos CA, Grover NS, Beaven AW, et al. . Anti-CD30 CAR-T cell therapy in relapsed and refractory Hodgkin lymphoma. J Clin Oncol. 2020;38(32):3794-3804.
    1. Wang CM, Wu ZQ, Wang Y, et al. . Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin lymphoma: an open-label phase I trial. Clin Cancer Res. 2017;23(5):1156-1166.
    1. Grover NS, Savoldo B. Challenges of driving CD30-directed CAR-T cells to the clinic. BMC Cancer. 2019;19(1):203.
    1. Ansell SM, Lesokhin AM, Borrello I, et al. . PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372(4):311-319.
    1. Armand P, Engert A, Younes A, et al. . Nivolumab for relapsed/refractory classic Hodgkin lymphoma after failure of autologous hematopoietic cell transplantation: extended follow-up of the multicohort single-arm phase II CheckMate 205 trial [correction published in J Clin Oncol. 2018;36(26):2748]. J Clin Oncol. 2018;36(14):1428-1439.
    1. Armand P, Janssens AM, Gritti G, et al. . Efficacy and safety results from CheckMate 140, a phase 2 study of nivolumab for relapsed/refractory follicular lymphoma. Blood. 2021;137(5):637-645.
    1. Chen R, Zinzani PL, Lee HJ, et al. . Pembrolizumab in relapsed or refractory Hodgkin lymphoma: 2-year follow-up of KEYNOTE-087. Blood. 2019;134(14):1144-1153.
    1. Younes A, Santoro A, Shipp M, et al. . Nivolumab for classical Hodgkin’s lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol. 2016;17(9):1283-1294.
    1. Hont AB, Cruz CR, Ulrey R, et al. . Immunotherapy of relapsed and refractory solid tumors with ex vivo expanded multi-tumor associated antigen specific cytotoxic T lymphocytes: a phase I study. J Clin Oncol. 2019;37(26):2349-2359.
    1. Cheson BD. Staging and response assessment in lymphomas: the new Lugano classification. Linchuang Zhongliuxue Zazhi. 2015;4(1):5.
    1. Gerdemann U, Katari U, Christin AS, et al. . Cytotoxic T lymphocytes simultaneously targeting multiple tumor-associated antigens to treat EBV negative lymphoma. Mol Ther. 2011;19(12):2258-2268.
    1. Tanna JG, Ulrey R, Williams KM, Hanley PJ. Critical testing and parameters for consideration when manufacturing and evaluating tumor-associated antigen-specific T cells. Cytotherapy. 2019;21(3):278-288.
    1. Weber G, Caruana I, Rouce RH, et al. . Generation of tumor antigen-specific T cell lines from pediatric patients with acute lymphoblastic leukemia: implications for immunotherapy. Clin Cancer Res. 2013;19(18):5079-5091.
    1. Hanley PJ, Melenhorst JJ, Nikiforow S, et al. . CMV-specific T cells generated from naïve T cells recognize atypical epitopes and may be protective in vivo. Sci Transl Med. 2015;7(285):285ra63.
    1. Robins HS, Campregher PV, Srivastava SK, et al. . Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood. 2009;114(19):4099-4107.
    1. Carlson CS, Emerson RO, Sherwood AM, et al. . Using synthetic templates to design an unbiased multiplex PCR assay. Nat Commun. 2013;4(1):2680.
    1. Robins H, Desmarais C, Matthis J, et al. . Ultra-sensitive detection of rare T cell clones. J Immunol Methods. 2012;375(1-2):14-19.
    1. Vasileiou S, Lulla PD, Tzannou I, et al. . T-cell therapy for lymphoma using nonengineered multiantigen-targeted T cells is safe and produces durable clinical effects [correction published in J Clin Oncol. 2021;39(18):2053]. J Clin Oncol. 2021;39(13):1415-1425.
    1. Lulla P, Naik S, Vasileiou S, et al. . Clinical effects of administering leukemia-specific donor T cells to patients with AML/MDS post-allogeneic transplant. Blood. 2021;137(19):2585-2597.
    1. Lulla PD, Tzannou I, Vasileiou S, et al. . The safety and clinical effects of administering a multiantigen-targeted T cell therapy to patients with multiple myeloma. Sci Transl Med. 2020;12(554):eaaz3339.
    1. Kvistborg P, Philips D, Kelderman S, et al. . Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med. 2014; 6(254):254ra128.
    1. Robert L, Tsoi J, Wang X, et al. . CTLA4 blockade broadens the peripheral T-cell receptor repertoire. Clin Cancer Res. 2014;20(9):2424-2432.
    1. Yuan J, Adamow M, Ginsberg BA, et al. . Integrated NY-ESO-1 antibody and CD8+ T-cell responses correlate with clinical benefit in advanced melanoma patients treated with ipilimumab. Proc Natl Acad Sci USA. 2011;108(40):16723-16728.
    1. Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162-174.
    1. De Sanctis F, Sandri S, Ferrarini G, et al. . The emerging immunological role of post-translational modifications by reactive nitrogen species in cancer microenvironment. Front Immunol. 2014;5:69.
    1. Jain MD, Zhao H, Wang X, et al. . Tumor interferon signaling and suppressive myeloid cells are associated with CAR T-cell failure in large B-cell lymphoma. Blood. 2021;137(19):2621-2633.
    1. Ramchandren R, Domingo-Domènech E, Rueda A, et al. . Nivolumab for newly diagnosed advanced-stage classic Hodgkin lymphoma: safety and efficacy in the phase II CheckMate 205 study. J Clin Oncol. 2019;37(23):1997-2007.
    1. Kanate AS, Kumar A, Dreger P, et al. . Maintenance therapies for Hodgkin and non-Hodgkin lymphomas after autologous transplantation: a consensus project of ASBMT, CIBMTR, and the Lymphoma Working Party of EBMT. JAMA Oncol. 2019;5(5):715-722.
    1. Moskowitz CH, Walewski J, Nademanee A, et al. . Five-year PFS from the AETHERA trial of brentuximab vedotin for Hodgkin lymphoma at high risk of progression or relapse. Blood. 2018;132(25):2639-2642.
    1. Steidl C, Connors JM, Gascoyne RD. Molecular pathogenesis of Hodgkin’s lymphoma: increasing evidence of the importance of the microenvironment. J Clin Oncol. 2011;29(14):1812-1826.
    1. Koyama S, Akbay EA, Li YY, et al. . Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun. 2016;7(1):10501.
    1. Lichtenegger FS, Rothe M, Schnorfeil FM, et al. . Targeting LAG-3 and PD-1 to enhance T cell activation by antigen-presenting cells. Front Immunol. 2018;9:385.
    1. Foster AE, Dotti G, Lu A, et al. . Antitumor activity of EBV-specific T lymphocytes transduced with a dominant negative TGF-beta receptor. J Immunother. 2008;31(5):500-505.

Source: PubMed

3
Abonneren