Langerhans dendritic cell vaccine bearing mRNA-encoded tumor antigens induces antimyeloma immunity after autotransplant

David J Chung, Sneh Sharma, Madhumitha Rangesa, Susan DeWolf, Yuval Elhanati, Karlo Perica, James W Young, David J Chung, Sneh Sharma, Madhumitha Rangesa, Susan DeWolf, Yuval Elhanati, Karlo Perica, James W Young

Abstract

Posttransplant vaccination targeting residual disease is an immunotherapeutic strategy to improve antigen-specific immune responses and prolong disease-free survival after autologous stem cell transplantation (ASCT) for multiple myeloma (MM). We conducted a phase 1 vaccine trial to determine the safety, toxicity, and immunogenicity of autologous Langerhans-type dendritic cells (LCs) electroporated with CT7, MAGE-A3, and Wilms tumor 1 (WT1) messenger RNA (mRNA), after ASCT for MM. Ten patients received a priming immunization plus 2 boosters at 12, 30, and 90 days, respectively, after ASCT. Vaccines contained 9 × 106 mRNA-electroporated LCs. Ten additional patients did not receive LC vaccines but otherwise underwent identical ASCT and supportive care. At 3 months after ASCT, all patients started lenalidomide maintenance therapy. Vaccinated patients developed mild local delayed-type hypersensitivity reactions after booster vaccines, but no toxicities exceeded grade 1. At 1 and 3 months after vaccines, antigen-specific CD4 and CD8 T cells increased secretion of proinflammatory cytokines (interferon-γ, interleukin-2, and tumor necrosis factor-α) above prevaccine levels, and also upregulated the cytotoxicity marker CD107a. CD4 and CD8 T-cell repertoire analysis showed a trend for increased clonal expansion in the vaccine cohort, which was more pronounced in the CD4 compartment. Although not powered to assess clinical efficacy, treatment responses favored the vaccine arm. Triple antigen-bearing mRNA-electroporated autologous LC vaccination initiated at engraftment after ASCT, in conjunction with standard lenalidomide maintenance therapy for MM, is safe and induces antigen-specific immune reactivity. This trial was registered at www.clinicaltrials.gov as #NCT01995708.

© 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.
Clinical responses after ASCT and mRNA-electroporated LC vaccines. (A) Depth of response pre-ASCT and at days +90 (d90) and +365 (d365) after ASCT. Relapse is also indicated. PFS and OS for patients in the control arm (B) and the vaccine arm (C). D, deceased; R, relapse.
Figure 2.
Figure 2.
Bone marrow immunophenotyping after ASCT and mRNA-electroporated LC vaccines. BMMCs were analyzed by flow cytometry to profile the lymphocyte composition postvaccination in comparison with baseline (prevaccine) and an unvaccinated control cohort. (A) t-Distributed stochastic neighbor embedding plots at 3 months after vaccination showing CD4 and CD8 T cells (far left plot), CD4 and CD8 effector memory cells (TEM; CCR7negCD45RAneg) (middle left plot), CD4 and CD8 naive cells (TN; CCR7+CD45ROneg) (middle right plot), and PD-1–expressing CD4 and CD8 cells (far right plot). CD4 (B) and CD8 (C) effector memory, naive, and PD-1–expressing cells at 1 and 3 months after vaccines. Pooled data (mean ± standard deviation) are shown for patients in the control (red circles) and vaccine (blue squares) arms. ns, not significant; Pre, prevaccine.
Figure 3.
Figure 3.
LC vaccines after ASCT stimulate antigen-specific CD4 and CD8 T-cell cytokine secretion and activation epitope expression. Postvaccination recall responses to CT7, MAGE-A3, and WT1 mRNA-electroporated LCs were measured as the fold change increase above prevaccine baseline using PBMCs at 1 and 3 months after vaccines (A-B) and BMMCs at 3 months after vaccines (C-D). Fold change in CD4 (A,C) and CD8 (B,D) T-cell proinflammatory cytokine (IFN-γ, IL-2, and TNF-α) secretion and activation epitope (CD107a and Mip-1β) expression. Pooled data (mean ± standard deviation) are shown for patients from the control (red circles) and vaccine (blue squares) arms. ns, not significant.
Figure 4.
Figure 4.
T-cell clonality after ASCT and vaccination with mRNA-electroporated LCs. Next-generation deep sequencing of the TCR-V-β CDR3 was performed on CD4 and CD8 T cells isolated from PBMCs obtained at days 30, 90, and 120 after ASCT and compared with prevaccine levels (day 12). (A) Stacked bar plots of CD4 and CD8 T-cell repertoire composition from 6 representative patients (3 from vaccine arm, 3 from control arm). Each color represents a unique clone, with the higher-percentage clones at the top and lower-percentage clones underneath (low-frequency clones blend together in black). (B) Heat maps of interpatient overlap of CD4 and CD8 T-cell clones calculated using the Morisita similarity index (dark yellow = similar; dark green = dissimilar). V1-V9 along the x- and y-axes represent vaccinated patients. C1-C9 along the x- and y-axes represent control patients. (C) Violin plots representing the mean log fold change in CD4 and CD8 T-cell expansion. Each color represents an individual patient. The red dashed line represents the mean log fold change value in each group. D12, day 12; D30, day 30; D90, day +90; D120, day 120.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7-30.
    1. Mikhael J, Ismaila N, Cheung MC, et al. . Treatment of multiple myeloma: ASCO and CCO Joint Clinical Practice Guideline [published correction appears in J Clin Oncol. 2020;38(21):2469]. J Clin Oncol. 2019;37(14):1228-1263.
    1. Pellat-Deceunynck C, Jego G, Harousseau JL, Vié H, Bataille R. Isolation of human lymphocyte antigens class I-restricted cytotoxic T lymphocytes against autologous myeloma cells. Clin Cancer Res. 1999;5(3):705-709.
    1. Dhodapkar MV, Krasovsky J, Olson K. T cells from the tumor microenvironment of patients with progressive myeloma can generate strong, tumor-specific cytolytic responses to autologous, tumor-loaded dendritic cells. Proc Natl Acad Sci USA. 2002;99(20):13009-13013.
    1. Noonan K, Matsui W, Serafini P, et al. . Activated marrow-infiltrating lymphocytes effectively target plasma cells and their clonogenic precursors. Cancer Res. 2005;65(5):2026-2034.
    1. Goodyear O, Piper K, Khan N, et al. . CD8+ T cells specific for cancer germline gene antigens are found in many patients with multiple myeloma, and their frequency correlates with disease burden. Blood. 2005;106(13):4217-4224.
    1. Dhodapkar MV, Krasovsky J, Osman K, Geller MD. Vigorous premalignancy-specific effector T cell response in the bone marrow of patients with monoclonal gammopathy. J Exp Med. 2003;198(11):1753-1757.
    1. Spisek R, Kukreja A, Chen L-C, et al. . Frequent and specific immunity to the embryonal stem cell-associated antigen SOX2 in patients with monoclonal gammopathy. J Exp Med. 2007;204(4):831-840.
    1. Chung DJ, Pronschinske KB, Shyer JA, et al. . T-cell exhaustion in multiple myeloma relapse after autotransplant: optimal timing of immunotherapy. Cancer Immunol Res. 2016;4(1):61-71.
    1. Rosenblatt J, Avivi I, Vasir B, et al. . Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin Cancer Res. 2013;19(13):3640-3648.
    1. Cohen AD, Lendvai N, Nataraj S, et al. . Autologous lymphocyte infusion supports tumor antigen vaccine-induced immunity in autologous stem cell transplant for multiple myeloma. Cancer Immunol Res. 2019;7(4):658-669.
    1. McCarthy PL, Holstein SA, Petrucci MT, et al. . Lenalidomide maintenance after autologous stem-cell transplantation in newly diagnosed multiple myeloma: a meta-analysis. J Clin Oncol. 2017;35(29):3279-3289.
    1. Görgün G, Calabrese E, Soydan E, et al. . Immunomodulatory effects of lenalidomide and pomalidomide on interaction of tumor and bone marrow accessory cells in multiple myeloma. Blood. 2010;116(17):3227-3237.
    1. Luptakova K, Rosenblatt J, Glotzbecker B, et al. . Lenalidomide enhances anti-myeloma cellular immunity. Cancer Immunol Immunother. 2013;62(1):39-49.
    1. Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M, Reis E Sousa C. Dendritic cells revisited. Annu Rev Immunol. 2021;39(1): 131-166.
    1. Santos PM, Butterfield LH. Dendritic cell-based cancer vaccines. J Immunol. 2018;200(2):443-449.
    1. Ratzinger G, Baggers J, de Cos MA, et al. . Mature human Langerhans cells derived from CD34+ hematopoietic progenitors stimulate greater cytolytic T lymphocyte activity in the absence of bioactive IL-12p70, by either single peptide presentation or cross-priming, than do dermal-interstitial or monocyte-derived dendritic cells. J Immunol. 2004;173(4):2780-2791.
    1. Klechevsky E, Morita R, Liu M, et al. . Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity. 2008;29(3):497-510.
    1. Romano E, Cotari JW, Barreira da Silva R, et al. . Human Langerhans cells use an IL-15R-α/IL-15/pSTAT5-dependent mechanism to break T-cell tolerance against the self-differentiation tumor antigen WT1. Blood. 2012;119(22):5182-5190.
    1. Banchereau J, Thompson-Snipes L, Zurawski S, et al. . The differential production of cytokines by human Langerhans cells and dermal CD14(+) DCs controls CTL priming. Blood. 2012;119(24):5742-5749.
    1. Banchereau J, Palucka AK, Dhodapkar M, et al. . Immune and clinical responses in patients with metastatic melanoma to CD34(+) progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61(17):6451-6458.
    1. Chung DJ, Carvajal RD, Postow MA, et al. . Langerhans-type dendritic cells electroporated with TRP-2 mRNA stimulate cellular immunity against melanoma: results of a phase I vaccine trial. OncoImmunology. 2017;7(1):e1372081.
    1. Nair SK, Boczkowski D, Morse M, Cumming RI, Lyerly HK, Gilboa E. Induction of primary carcinoembryonic antigen (CEA)-specific cytotoxic T lymphocytes in vitro using human dendritic cells transfected with RNA. Nat Biotechnol. 1998;16(4):364-369.
    1. Boczkowski D, Nair SK, Nam JH, Lyerly HK, Gilboa E. Induction of tumor immunity and cytotoxic T lymphocyte responses using dendritic cells transfected with messenger RNA amplified from tumor cells. Cancer Res. 2000;60(4):1028-1034.
    1. Muul LM, Tuschong LM, Soenen SL, et al. . Persistence and expression of the adenosine deaminase gene for 12 years and immune reaction to gene transfer components: long-term results of the first clinical gene therapy trial. Blood. 2003;101(7):2563-2569.
    1. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, et al. . LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003;302(5644):415-419.
    1. Jungbluth AA, Ely S, DiLiberto M, et al. . The cancer-testis antigens CT7 (MAGE-C1) and MAGE-A3/6 are commonly expressed in multiple myeloma and correlate with plasma-cell proliferation. Blood. 2005;106(1):167-174.
    1. Tinguely M, Jenni B, Knights A, et al. . MAGE-C1/CT-7 expression in plasma cell myeloma: sub-cellular localization impacts on clinical outcome. Cancer Sci. 2008;99(4):720-725.
    1. Tyler EM, Jungbluth AA, O’Reilly RJ, Koehne G. WT1-specific T-cell responses in high-risk multiple myeloma patients undergoing allogeneic T cell-depleted hematopoietic stem cell transplantation and donor lymphocyte infusions. Blood. 2013;121(2):308-317.
    1. Nardiello T, Jungbluth AA, Mei A, et al. . MAGE-A inhibits apoptosis in proliferating myeloma cells through repression of Bax and maintenance of survivin. Clin Cancer Res. 2011;17(13):4309-4319.
    1. Hatta Y, Takeuchi J, Saitoh T, et al. . WT1 expression level and clinical factors in multiple myeloma. J Exp Clin Cancer Res. 2005;24(4):595-599.
    1. Tyler EM, Jungbluth AA, O’Reilly RJ, Koehne G. Wilms’ tumor 1 protein-specific T-cell responses in high-risk multiple myeloma patients undergoing T-cell depleted allogeneic hematopoietic stem cell transplantation and donor lymphocyte infusion. Blood. 2013;121(2):308-317.
    1. Tyler EM, Jungbluth AA, Gnjatic S, O’Reilly RJ, Koehne G. Cancer-testis antigen 7 expression and immune responses following allogeneic stem cell transplantation for multiple myeloma. Cancer Immunol Res. 2014;2(6):547-558.
    1. Azuma T, Otsuki T, Kuzushima K, Froelich CJ, Fujita S, Yasukawa M. Myeloma cells are highly sensitive to the granule exocytosis pathway mediated by WT1-specific cytotoxic T lymphocytes. Clin Cancer Res. 2004;10(21):7402-7412.
    1. Tsuboi A, Oka Y, Nakajima H, et al. . Wilms tumor gene WT1 peptide-based immunotherapy induced a minimal response in a patient with advanced therapy-resistant multiple myeloma. Int J Hematol. 2007;86(5):414-417.
    1. Yao TJ, Begg CB, Livingston PO. Optimal sample size for a series of pilot trials of new agents. Biometrics. 1996;52(3):992-1001.
    1. Jungbluth AA, Chen YT, Busam KJ, et al. . CT7 (MAGE-C1) antigen expression in normal and neoplastic tissues. Int J Cancer. 2002;99(6):839-845.
    1. Chung DJ, Romano E, Pronschinske KB, et al. . Langerhans-type and monocyte-derived human dendritic cells have different susceptibilities to mRNA electroporation with distinct effects on maturation and activation: implications for immunogenicity in dendritic cell-based immunotherapy. J Transl Med. 2013;11(1):166.
    1. Roshal M, Flores-Montero JA, Gao Q, et al. . MRD detection in multiple myeloma: comparison between MSKCC 10-color single-tube and EuroFlow 8-color 2-tube methods. Blood Adv. 2017;1(12):728-732.
    1. Muraro PA, Robins H, Malhotra S, et al. . T cell repertoire following autologous stem cell transplantation for multiple sclerosis. J Clin Invest. 2014;124(3):1168-1172.
    1. Sheikh N, Cham J, Zhang L, et al. . Clonotypic diversification of intratumoral T cells following sipuleucel-T treatment in prostate cancer subjects. Cancer Res. 2016;76(13):3711-3718.
    1. Postow MA, Manuel M, Wong P, et al. . Peripheral T cell receptor diversity is associated with clinical outcomes following ipilimumab treatment in metastatic melanoma. J Immunother Cancer. 2015;3(1):23.
    1. Ratta M, Fagnoni F, Curti A, et al. . Dendritic cells are functionally defective in multiple myeloma: the role of interleukin-6. Blood. 2002;100(1): 230-237.
    1. Kukreja A, Hutchinson A, Dhodapkar K, et al. . Enhancement of clonogenicity of human multiple myeloma by dendritic cells. J Exp Med. 2006; 203(8):1859-1865.
    1. Leone P, Berardi S, Frassanito MA, et al. . Dendritic cells accumulate in the bone marrow of myeloma patients where they protect tumor plasma cells from CD8+ T-cell killing. Blood. 2015;126(12):1443-1451.
    1. Racanelli V, Leone P, Frassanito MA, et al. . Alterations in the antigen processing-presenting machinery of transformed plasma cells are associated with reduced recognition by CD8+ T cells and characterize the progression of MGUS to multiple myeloma. Blood. 2010;115(6):1185-1193.
    1. Görgün GT, Whitehill G, Anderson JL, et al. . Tumor-promoting immune-suppressive myeloid-derived suppressor cells in the multiple myeloma microenvironment in humans. Blood. 2013;121(15):2975-2987.
    1. Braga WM, da Silva BR, de Carvalho AC, et al. . FOXP3 and CTLA4 overexpression in multiple myeloma bone marrow as a sign of accumulation of CD4(+) T regulatory cells. Cancer Immunol Immunother. 2014;63(11):1189-1197.
    1. Asimakopoulos F, Kim J, Denu RA, et al. . Macrophages in multiple myeloma: emerging concepts and therapeutic implications. Leuk Lymphoma. 2013;54(10):2112-2121.
    1. Minnie SA, Kuns RD, Gartlan KH, et al. . Myeloma escape after stem cell transplantation is a consequence of T-cell exhaustion and is prevented by TIGIT blockade [published correction appears in Blood. 2019;134(21):1878]. Blood. 2018;132(16):1675-1688.
    1. Parmar H, Gertz M, Anderson EI, Kumar S, Kourelis TV. Microenvironment immune reconstitution patterns correlate with outcomes after autologous transplant in multiple myeloma. Blood Adv. 2021;5(7):1797-1804.
    1. Visram A, Kourelis T, Dasari S, Anderson E, Kumar SK. Describing the cellular and humoral immune tumor microenvironment and malignant transcriptome across the multiple myeloma disease spectrum. Blood. 2020;136(suppl 1):39-40.
    1. Shah N, Chari A, Scott E, Mezzi K, Usmani SZ. B-cell maturation antigen (BCMA) in multiple myeloma: rationale for targeting and current therapeutic approaches. Leukemia. 2020;34(4):985-1005.
    1. Akahori Y, Wang L, Yoneyama M, et al. . Antitumor activity of CAR-T cells targeting the intracellular oncoprotein WT1 can be enhanced by vaccination. Blood. 2018;132(11):1134-1145.
    1. Reinhard K, Rengstl B, Oehm P, et al. . An RNA vaccine drives expansion and efficacy of claudin-CAR-T cells against solid tumors. Science. 2020;367(6476):446-453.

Source: PubMed

3
Předplatit