Systemic and local immunity following adoptive transfer of NY-ESO-1 SPEAR T cells in synovial sarcoma

Indu Ramachandran, Daniel E Lowther, Rebecca Dryer-Minnerly, Ruoxi Wang, Svetlana Fayngerts, Daniel Nunez, Gareth Betts, Natalie Bath, Alex J Tipping, Luca Melchiori, Jean-Marc Navenot, John Glod, Crystal L Mackall, Sandra P D'Angelo, Dejka M Araujo, Warren A Chow, George D Demetri, Mihaela Druta, Brian A Van Tine, Stephan A Grupp, Albiruni R Abdul Razak, Breelyn Wilky, Malini Iyengar, Trupti Trivedi, Erin Van Winkle, Karen Chagin, Rafael Amado, Gwendolyn K Binder, Samik Basu, Indu Ramachandran, Daniel E Lowther, Rebecca Dryer-Minnerly, Ruoxi Wang, Svetlana Fayngerts, Daniel Nunez, Gareth Betts, Natalie Bath, Alex J Tipping, Luca Melchiori, Jean-Marc Navenot, John Glod, Crystal L Mackall, Sandra P D'Angelo, Dejka M Araujo, Warren A Chow, George D Demetri, Mihaela Druta, Brian A Van Tine, Stephan A Grupp, Albiruni R Abdul Razak, Breelyn Wilky, Malini Iyengar, Trupti Trivedi, Erin Van Winkle, Karen Chagin, Rafael Amado, Gwendolyn K Binder, Samik Basu

Abstract

Background: Gene-modified autologous T cells expressing NY-ESO-1c259, an affinity-enhanced T-cell receptor (TCR) reactive against the NY-ESO-1-specific HLA-A*02-restricted peptide SLLMWITQC (NY-ESO-1 SPEAR T-cells; GSK 794), have demonstrated clinical activity in patients with advanced synovial sarcoma (SS). The factors contributing to gene-modified T-cell expansion and the changes within the tumor microenvironment (TME) following T-cell infusion remain unclear. These studies address the immunological mechanisms of response and resistance in patients with SS treated with NY-ESO-1 SPEAR T-cells.

Methods: Four cohorts were included to evaluate antigen expression and preconditioning on efficacy. Clinical responses were assessed by RECIST v1.1. Engineered T-cell persistence was determined by qPCR. Serum cytokines were evaluated by immunoassay. Transcriptomic analyses and immunohistochemistry were performed on tumor biopsies from patients before and after T-cell infusion. Gene-modified T-cells were detected within the TME via an RNAish assay.

Results: Responses across cohorts were affected by preconditioning and intra-tumoral NY-ESO-1 expression. Of the 42 patients reported (data cut-off 4June2018), 1 patient had a complete response, 14 patients had partial responses, 24 patients had stable disease, and 3 patients had progressive disease. The magnitude of gene-modified T-cell expansion shortly after infusion was associated with response in patients with high intra-tumoral NY-ESO-1 expression. Patients receiving a fludarabine-containing conditioning regimen experienced increases in serum IL-7 and IL-15. Prior to infusion, the TME exhibited minimal leukocyte infiltration; CD163+ tumor-associated macrophages (TAMs) were the dominant population. Modest increases in intra-tumoral leukocytes (≤5%) were observed in a subset of subjects at approximately 8 weeks. Beyond 8 weeks post infusion, the TME was minimally infiltrated with a TAM-dominant leukocyte infiltrate. Tumor-associated antigens and antigen presentation did not significantly change within the tumor post-T-cell infusion. Finally, NY-ESO-1 SPEAR T cells trafficked to the TME and maintained cytotoxicity in a subset of patients.

Conclusions: Our studies elucidate some factors that underpin response and resistance to NY-ESO-1 SPEAR T-cell therapy. From these data, we conclude that a lymphodepletion regimen containing high doses of fludarabine and cyclophosphamide is necessary for SPEAR T-cell persistence and efficacy. Furthermore, these data demonstrate that non-T-cell inflamed tumors, which are resistant to PD-1/PD-L1 inhibitors, can be treated with adoptive T-cell based immunotherapy.

Trial registration: ClinicalTrials.gov, NCT01343043 , Registered 27 April 2011.

Keywords: Adoptive immunotherapy; Antigen loss; Checkpoint therapy; Cyclophosphamide; Cytokine; Engineered cell therapy; Fludarabine; IL-15; NY-ESO-1; Synovial sarcoma; T cell; TCR.

Conflict of interest statement

IR, DEL, RDM, RW, SF, DN, GB, NB, AJT, LM, JMN, MI, TT, EVW, KC, RA, GKB, and SB: employees of Adaptimmune and have stock or other ownership interests in Adaptimmune.

JG: None

CLM: Consulting for Adaptimmune, GlaxoSmithKline, Allogene, Nektar, Vor, PACT, Unum, Bryologyx, Apricity, Roche, Lyell. Equity in Allogene, Vor, PACT, Unum, Lyell. Research Funding from: Obsidian.

SPD: Consulting for Amgen, EMD Serono, Nektar, travel support from Adaptimmune

DMA: None

WAC: Speaker’s bureau and research funding from Novartis, data safety monitoring board for Advenchen

GDD: None

BAVT: Basic science grant funding from Pfizer, Tracon, and Merck; consulting fees from Epizyme, Lilly, CytRX, Janssen, Immune Design, Daiichi Sankyo, Plexxicon and Adaptimmune; speaking fees from Caris, Janseen, and Lilly

MD: Consulting and speaker’s bureau for Eisai and Lilly

SG: Research and/or clinical trial support from Novartis, Servier, Adaptimmune, and Kite. Consulting, study steering committees, or scientific advisory boards: Novartis, Cellectis, Adaptimmune, Eureka, TCR2, Juno, GlaxoSmithKline, Vertex, Cure Genetics, Humanigen, and Roche.

ARAR: Consulting for Lilly, Merck, Boehringer Ingelheim, research funding from CASI Pharmaceuticals, Boehringer Ingelheim

BW: Research support from Merck, Pfizer, Agenus, and consulting for Lilly and Immune Design

Figures

Fig. 1
Fig. 1
Clinical outcome in SS patients following NY-ESO-1 SPEAR T-cell infusion. Comparison of maximal tumor regression curves (waterfall plot) in 42 patients treated with NY-ESO-1 SPEAR T cells across four cohorts: a cohort 1, b cohort 2, c cohort 3, d cohort 4. Spider plots of tumor burden changes following NY-ESO-1 SPEAR T-cell infusion in 42 patients across four cohorts: e cohort 1, f cohort 2, g cohort 3, h cohort 4
Fig. 2
Fig. 2
Pre-conditioning lymphodepletion regimen influences NY-ESO-1 SPEAR T-cell engraftment. a Peak expansion of transduced T cells in non-responders versus responders across all four cohorts was determined by measuring peak vector copies/μg of DNA in 42 patients treated with NY-ESO-1 SPEAR T cells. b IL-7 and c IL-15 levels in serum samples from 40 patients across all four cohorts were evaluated prior to (Pre-) and following (Post-) administration of pre-conditioning therapy, but prior to T-cell infusion. Box plots depict median, first and third quartiles. Dotted lines connect Pre- and Post- samples from the same patient. p-values between pre- and post-lymphodepletion in paired specimen in each cohort were calculated by the Wilcoxon matched-pairs signed-rank test
Fig. 3
Fig. 3
SPEAR T-cell therapy alters cellular infiltrate in tumor microenvironment. a Markers associated with immune cells and their function were evaluated at pre-infusion (red), and post- infusion at week 8 (blue) or beyond week 8 (grey) by IHC and plotted as a percentage of marker area within tumor area. Statistical significance in marker positivity between time points was determined by a two-way ANOVA test. b Immune marker expression in a representative region of pre- and post- infusion biopsies in one patient with increased leukocyte infiltration at week 8, and in another patient with minimal changes at >week 8 time point. Scale bar = 50 μm
Fig. 4
Fig. 4
SPEAR T-cell therapy does not affect antigen expression or presentation. a Representative IHC images of NY-ESO-1 expression at each of the time points evaluated. Scale bar = 100 μm. b NY-ESO-1 protein expression H-scores as determined by IHC in pre- and post-infusion biopsies from all patients where a minimum of one post-infusion biopsy was evaluable (N = 15). Where > 1 biopsy per timepoint was evaluated, average H-score is shown. Mann-Whitney statistical test was used to evaluate changes between pre- and post-infusion time points. Tumor-associated antigen (c) and antigen-processing machinery (d). RNA expression shown as normalized counts as determined by the NanoString assay performed on pre- and post-infusion biopsies. Where more than one biopsy was collected and tested separately, points show the mean. Box-plots depict the median, along with first and third quartiles
Fig. 5
Fig. 5
Adoptively transferred NY-ESO-1 SPEAR T cells maintain functionality long after infusion. a Representative fields for detection of negative control RNA (DapB), positive control RNA (PPIB, POLR2A), and CD3 or NY-ESO-1c259TCR RNA by RNAish in one patient’s tumor collected over 2 years post-infusion. b A375 target killing shown as green object count as determined by the Incucyte killing assay performed on flow sorted CD3+CD8+dextramer+ T cells (red line) and CD3+CD8+dextramer− T cells (grey line) from a patient’s PBMCs collected 12 months post-infusion and on A375 alone (blue line)

References

    1. Thway K, Fisher C. Synovial sarcoma: defining features and diagnostic evolution. Ann Diagn Pathol. 2014;18(6):369–380. doi: 10.1016/j.anndiagpath.2014.09.002.
    1. Paoluzzi L, Cacavio A, Ghesani M, Karambelkar A, Rapkiewicz A, Weber J, et al. Response to anti-PD1 therapy with nivolumab in metastatic sarcomas. Clin Sarcoma Res. 2016;6:24. doi: 10.1186/s13569-016-0064-0.
    1. Tawbi HA, Burgess M, Bolejack V, Van Tine BA, Schuetze SM, Hu J, et al. Pembrolizumab in advanced soft-tissue sarcoma and bone sarcoma (SARC028): a multicentre, two-cohort, single-arm, open-label, phase 2 trial. Lancet Oncol. 2017;18(11):1493–1501. doi: 10.1016/S1470-2045(17)30624-1.
    1. Toulmonde M, Penel N, Adam J, Chevreau C, Blay JY, Le Cesne A, et al. Use of PD-1 targeting, macrophage infiltration, and IDO pathway activation in sarcomas: a phase 2 clinical trial. JAMA Oncol. 2018;4(1):93–97. doi: 10.1001/jamaoncol.2017.1617.
    1. Rizvi H, Sanchez-Vega F, La K, Chatila W, Jonsson P, Halpenny D, et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung cancer profiled with targeted next-generation sequencing. J Clin Oncol. 2018;36(7):633–641. doi: 10.1200/JCO.2017.75.3384.
    1. Oike N, Kawashima H, Ogose A, Hotta T, Hatano H, Ariizumi T, et al. Prognostic impact of the tumor immune microenvironment in synovial sarcoma. Cancer Sci. 2018;109(10):3043–3054. doi: 10.1111/cas.13769.
    1. Pollack SM, He Q, Yearley JH, Emerson R, Vignali M, Zhang Y, et al. T-cell infiltration and clonality correlate with programmed cell death protein 1 and programmed death-ligand 1 expression in patients with soft tissue sarcomas. Cancer. 2017;123(17):3291–3304. doi: 10.1002/cncr.30726.
    1. Services USDoHaH. The Cancer Genome Atlas 2017 [Available from: .
    1. Jungbluth AA, Chen YT, Stockert E, Busam KJ, Kolb D, Iversen K, et al. Immunohistochemical analysis of NY-ESO-1 antigen expression in normal and malignant human tissues. Int J Cancer. 2001;92(6):856–860. doi: 10.1002/ijc.1282.
    1. Lai JP, Robbins PF, Raffeld M, Aung PP, Tsokos M, Rosenberg SA, et al. NY-ESO-1 expression in synovial sarcoma and other mesenchymal tumors: significance for NY-ESO-1-based targeted therapy and differential diagnosis. Mod Pathol. 2012;25(6):854–858. doi: 10.1038/modpathol.2012.31.
    1. Satie AP, Rajpert-De Meyts E, Spagnoli GC, Henno S, Olivo L, Jacobsen GK, et al. The cancer-testis gene, NY-ESO-1, is expressed in normal fetal and adult testes and in spermatocytic seminomas and testicular carcinoma in situ. Lab Investig. 2002;82(6):775–780. doi: 10.1097/01.LAB.0000017169.26718.5F.
    1. Pollack SM, Ingham M, Spraker MB, Schwartz GK. Emerging targeted and immune-based therapies in sarcoma. J Clin Oncol. 2018;36(2):125–135. doi: 10.1200/JCO.2017.75.1610.
    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(7):917–924. doi: 10.1200/JCO.2010.32.2537.
    1. D'Angelo SP, Melchiori L, Merchant MS, Bernstein D, Glod J, Kaplan R, et al. Antitumor activity associated with prolonged persistence of adoptively transferred NY-ESO-1 (c259) T cells in synovial sarcoma. Cancer Discov. 2018;8(8):944–957. doi: 10.1158/-17-1417.
    1. Rapoport AP, Stadtmauer EA, Binder-Scholl GK, Goloubeva O, Vogl DT, Lacey SF, et al. NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma. Nat Med. 2015;21(8):914–921. doi: 10.1038/nm.3910.
    1. Cesano A. nCounter((R)) PanCancer immune profiling panel (NanoString Technologies, Inc., Seattle, WA) J Immunother Cancer. 2015;3:42. doi: 10.1186/s40425-015-0088-7.
    1. The R project for statistical computing. Vienna: The R Foundation; 2018 [Available from: .
    1. Iura K, Maekawa A, Kohashi K, Ishii T, Bekki H, Otsuka H, et al. Cancer-testis antigen expression in synovial sarcoma: NY-ESO-1, PRAME, MAGEA4, and MAGEA1. Hum Pathol. 2017;61:130–139. doi: 10.1016/j.humpath.2016.12.006.
    1. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra25. doi: 10.1126/scitranslmed.3008226.
    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(6):540–549. doi: 10.1200/JCO.2014.56.2025.
    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(9967):517–528. doi: 10.1016/S0140-6736(14)61403-3.
    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(16):1507–1517. doi: 10.1056/NEJMoa1407222.
    1. Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123–2138. doi: 10.1172/JCI85309.
    1. Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8(355):355ra116. doi: 10.1126/scitranslmed.aaf8621.
    1. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439–448. doi: 10.1056/NEJMoa1709866.
    1. Berdeja JG, Lin Y, Raje N, Munshi N, Siegel D, Liedtke M, et al. Durable clinical responses in heavily pretreated patients with relapsed/refractory multiple myeloma: updated results from a multicenter study of bb2121 Anti-Bcma CAR T cell therapy. NEJM. 2017;130(Suppl 1):740.
    1. Blank CU, Haanen JB, Ribas A, Schumacher TN. CANCER IMMUNOLOGY. The “cancer immunogram”. Science. 2016;352(6286):658–660. doi: 10.1126/science.aaf2834.
    1. McBride MJ, Pulice JL, Beird HC, Ingram DR, D’Avino AR, Shern JF, et al. The SS18-SSX fusion oncoprotein hijacks BAF complex targeting and function to drive synovial sarcoma. Cancer Cell. 2018;33(6):1128–41.e7. doi: 10.1016/j.ccell.2018.05.002.
    1. Lowe KL, Mackall CL, Norry E, Amado R, Jakobsen BK, Binder G. Fludarabine and neurotoxicity in engineered T-cell therapy. Gene Ther. 2018;25(3):176–191. doi: 10.1038/s41434-018-0019-6.
    1. Fraietta JA, Nobles CL, Sammons MA, Lundh S, Carty SA, Reich TJ, et al. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018;558(7709):307–312. doi: 10.1038/s41586-018-0178-z.
    1. Heczey A, Louis CU, Savoldo B, Dakhova O, Durett A, Grilley B, et al. CAR T cells administered in combination with Lymphodepletion and PD-1 inhibition to patients with neuroblastoma. Mol Ther. 2017;25(9):2214–2224. doi: 10.1016/j.ymthe.2017.05.012.
    1. Kochenderfer JN, Somerville RPT, Lu T, Shi V, Bot A, Rossi J, et al. Lymphoma remissions caused by anti-CD19 chimeric antigen receptor T cells are associated with high serum Interleukin-15 levels. J Clin Oncol. 2017;35(16):1803–1813. doi: 10.1200/JCO.2016.71.3024.
    1. Bao L, Dunham K, Lucas K. MAGE-A1, MAGE-A3, and NY-ESO-1 can be upregulated on neuroblastoma cells to facilitate cytotoxic T lymphocyte-mediated tumor cell killing. Cancer Immunol Immunother. 2011;60(9):1299–1307. doi: 10.1007/s00262-011-1037-z.
    1. Griffiths EA, Srivastava P, Matsuzaki J, Brumberger Z, Wang ES, Kocent J, et al. NY-ESO-1 vaccination in combination with decitabine induces antigen-specific T-lymphocyte responses in patients with myelodysplastic syndrome. Clin Cancer Res. 2018;24(5):1019–1029. doi: 10.1158/1078-0432.CCR-17-1792.
    1. Klar AS, Gopinadh J, Kleber S, Wadle A, Renner C. Treatment with 5-Aza-2′-Deoxycytidine induces expression of NY-ESO-1 and facilitates cytotoxic T lymphocyte-mediated tumor cell killing. PLoS One. 2015;10(10):e0139221. doi: 10.1371/journal.pone.0139221.
    1. Thomas R, Al-Khadairi G, Roelands J, Hendrickx W, Dermime S, Bedognetti D, et al. NY-ESO-1 based immunotherapy of cancer: current perspectives. Front Immunol. 2018;9:947. doi: 10.3389/fimmu.2018.00947.
    1. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368(16):1509–1518. doi: 10.1056/NEJMoa1215134.
    1. Gardner R, Wu D, Cherian S, Fang M, Hanafi LA, Finney O, et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood. 2016;127(20):2406–2410. doi: 10.1182/blood-2015-08-665547.
    1. O’Rourke Donald M., Nasrallah MacLean P., Desai Arati, Melenhorst Jan J., Mansfield Keith, Morrissette Jennifer J. D., Martinez-Lage Maria, Brem Steven, Maloney Eileen, Shen Angela, Isaacs Randi, Mohan Suyash, Plesa Gabriela, Lacey Simon F., Navenot Jean-Marc, Zheng Zhaohui, Levine Bruce L., Okada Hideho, June Carl H., Brogdon Jennifer L., Maus Marcela V. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Science Translational Medicine. 2017;9(399):eaaa0984. doi: 10.1126/scitranslmed.aaa0984.
    1. Rodig Scott J., Gusenleitner Daniel, Jackson Donald G., Gjini Evisa, Giobbie-Hurder Anita, Jin Chelsea, Chang Han, Lovitch Scott B., Horak Christine, Weber Jeffrey S., Weirather Jason L., Wolchok Jedd D., Postow Michael A., Pavlick Anna C., Chesney Jason, Hodi F. Stephen. MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Science Translational Medicine. 2018;10(450):eaar3342. doi: 10.1126/scitranslmed.aar3342.
    1. Sade-Feldman M, Jiao YJ, Chen JH, Rooney MS, Barzily-Rokni M, Eliane JP, et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat Commun. 2017;8(1):1136. doi: 10.1038/s41467-017-01062-w.
    1. McGranahan N, Rosenthal R, Hiley CT, Rowan AJ, Watkins TBK, Wilson GA, et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell. 2017;171(6):1259–71.e11. doi: 10.1016/j.cell.2017.10.001.
    1. Paulson KG, Voillet V, McAfee MS, Hunter DS, Wagener FD, Perdicchio M, et al. Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat Commun. 2018;9(1):3868. doi: 10.1038/s41467-018-06300-3.
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6382):1350–1355. doi: 10.1126/science.aar4060.
    1. Halse H, Colebatch AJ, Petrone P, Henderson MA, Mills JK, Snow H, et al. Multiplex immunohistochemistry accurately defines the immune context of metastatic melanoma. Sci Rep. 2018;8(1):11158. doi: 10.1038/s41598-018-28944-3.
    1. Elliott LA, Doherty GA, Sheahan K, Ryan EJ. Human tumor-infiltrating myeloid cells: phenotypic and functional diversity. Front Immunol. 2017;8:86. doi: 10.3389/fimmu.2017.00086.
    1. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41(1):49–61. doi: 10.1016/j.immuni.2014.06.010.

Source: PubMed

3
Sottoscrivi