NY-ESO-1-specific TCR-engineered T cells mediate sustained antigen-specific antitumor effects in myeloma

Aaron P Rapoport, Edward A Stadtmauer, Gwendolyn K Binder-Scholl, Olga Goloubeva, Dan T Vogl, Simon F Lacey, Ashraf Z Badros, Alfred Garfall, Brendan Weiss, Jeffrey Finklestein, Irina Kulikovskaya, Sanjoy K Sinha, Shari Kronsberg, Minnal Gupta, Sarah Bond, Luca Melchiori, Joanna E Brewer, Alan D Bennett, Andrew B Gerry, Nicholas J Pumphrey, Daniel Williams, Helen K Tayton-Martin, Lilliam Ribeiro, Tom Holdich, Saul Yanovich, Nancy Hardy, Jean Yared, Naseem Kerr, Sunita Philip, Sandra Westphal, Don L Siegel, Bruce L Levine, Bent K Jakobsen, Michael Kalos, Carl H June, Aaron P Rapoport, Edward A Stadtmauer, Gwendolyn K Binder-Scholl, Olga Goloubeva, Dan T Vogl, Simon F Lacey, Ashraf Z Badros, Alfred Garfall, Brendan Weiss, Jeffrey Finklestein, Irina Kulikovskaya, Sanjoy K Sinha, Shari Kronsberg, Minnal Gupta, Sarah Bond, Luca Melchiori, Joanna E Brewer, Alan D Bennett, Andrew B Gerry, Nicholas J Pumphrey, Daniel Williams, Helen K Tayton-Martin, Lilliam Ribeiro, Tom Holdich, Saul Yanovich, Nancy Hardy, Jean Yared, Naseem Kerr, Sunita Philip, Sandra Westphal, Don L Siegel, Bruce L Levine, Bent K Jakobsen, Michael Kalos, Carl H June

Abstract

Despite recent therapeutic advances, multiple myeloma (MM) remains largely incurable. Here we report results of a phase I/II trial to evaluate the safety and activity of autologous T cells engineered to express an affinity-enhanced T cell receptor (TCR) recognizing a naturally processed peptide shared by the cancer-testis antigens NY-ESO-1 and LAGE-1. Twenty patients with antigen-positive MM received an average 2.4 × 10(9) engineered T cells 2 d after autologous stem cell transplant. Infusions were well tolerated without clinically apparent cytokine-release syndrome, despite high IL-6 levels. Engineered T cells expanded, persisted, trafficked to marrow and exhibited a cytotoxic phenotype. Persistence of engineered T cells in blood was inversely associated with NY-ESO-1 levels in the marrow. Disease progression was associated with loss of T cell persistence or antigen escape, in accordance with the expected mechanism of action of the transferred T cells. Encouraging clinical responses were observed in 16 of 20 patients (80%) with advanced disease, with a median progression-free survival of 19.1 months. NY-ESO-1-LAGE-1 TCR-engineered T cells were safe, trafficked to marrow and showed extended persistence that correlated with clinical activity against antigen-positive myeloma.

Figures

Figure 1. Overview of clinical study
Figure 1. Overview of clinical study
Patient screening, including HLA testing and tumor antigen testing, and apheresis scheduling requires 2-4 weeks. Manufacture of gene-modified cells takes 3-4 weeks. Patients received high dose melphalan two days prior to stem cell infusion, and four days prior to T-cell infusion. Response assessments were performed at day 42, 100, 180 and quarterly thereafter. Optional bone marrow biopsies are indicated by asterisk. For eligible patients, maintenance lenalidomide was given starting at day 100. Once off study, patients are monitored for up to 15 years for delayed adverse events in accordance with FDA Guidance.
Figure 2. Persistence and function of gene-modified…
Figure 2. Persistence and function of gene-modified cells in blood and marrow
(a). The total number of gene-modified cells from infusion to 1 year on study is shown, as measured by Q-RT-PCR. The red line represents the median across patients. The limit of detection is approximately at 2.5 cells/microliter. (b). The percent of gene marking in blood over time is shown, using the left vertical axis. The red bold line represents the average WBC count using the right vertical axis. Standard deviation is represented by the error bars. (c). The total number of vector copies per microgram of DNA in bone marrow is represented for patients with two or more marrow collections post infusion. The percent of cells marked was calculated assuming a copy number of 1 per cell. The limit of detection for the assay is 10 copies and is represented by the red dotted line. (d). NY-ESO TCR-positive CD8 T-cells were evaluated for functionality in the cell product (MP) and at multiple timepoints post infusion by measuring production of IFN-γ, granzyme B and CD107a in response to antigen loaded T2 target cells. Non-antigen loaded T2 target cells were used as a negative control for all samples and background was subtracted from the values shown. Subsets of IFN-γ positive cells with +/− expression of granzyme B and CD107a are represented by the various colors in each histogram. Data from four responding patients with durable persistence are shown.
Figure 3. Tumor and T-cell infiltration in…
Figure 3. Tumor and T-cell infiltration in marrow
Core marrow samples were collected from patient 258 at day 7. (a). Marrow was stained for the plasma cell marker CD138 which is expressed on normal plasma and myeloma cells, and for CD8 to evaluate T-cell infiltration. (b). Mononuclear cells were isolated from fresh marrow aspirates, and stained with anti-CD3 antibody to detect T-cells, and NY-ESO TCR specific peptide/HLA-A2 dextramer reagent to detect gene-modified cells. The dextramer FMO is shown as a negative control. Gene-modified cells in the controls and test panel are denoted by the box, and the number to the left of the box represents the overall percent positive cells from the CD3 positive fraction. (c). Patient 253 received a second infusion of NY-ESO TCR T-cells in the absence of ASCT, after progressing following ASCT and first infusion. Core marrow biopsies were collected just prior to 1 infusion, and at day 24 and 38 after infusion, and were stained for the CD138 plasma cell/myeloma tumor marker, and for CD8 T-cell infiltration. Bars shown in (a) and (b) represent 20 microns.
Figure 4. Clinical response in patient 250…
Figure 4. Clinical response in patient 250 correlates with engineered T cell expansion
(a). The number of NY-ESO specific T-cells per microliter of blood is shown for patient 250, and is overlaid with the corresponding tumor burden assessed at days 42 - 180 post infusion. The kappa/lambda light chain ratio is shown on the right axis; n.b. that when ratio becomes normalized, plot is superimposed on X-axis. (b). The levels of TCR expression on CD8 T-cells over time were measured by peptide/HLA-A2 dextramer in marrow and blood; samples were gated on CD3+ lymphocytes. The percent double positive cells is represented by the number in the upper right hand corner. (c). H&E stain of a marrow biopsy pre and post treatment, demonstrating normalization of cellularity at two months. Note the magnification is slightly higher in the left panel. (d). Abdominal CT scan showing a pancreatic plasmacytoma (confirmed by biopsy) at baseline, and loss of the tumor two months after treatment. Bars in panel (c) are 10 microns and bars in panel (d) are 4 centimeters.
Figure 5. CD138, LAGE-1 and NY-ESO-1 expression…
Figure 5. CD138, LAGE-1 and NY-ESO-1 expression in marrow
Transcripts for the plasma cell and myeloma marker CD138 (a), and the tumor antigens LAGE-1 (b) and NY-ESO-1 (c) were evaluated by QRT-PCR in all marrow collections performed post infusion, and the percent change from baseline values was calculated at day 100 (left panels) and 180 (right panels). The numbers along the x-axis are patient numbers and correspond to numbers in other figures. Bars are colored to reflect the clinical response of each patient at day 100, which is the pre-specified timepoint for response assessment in the study. Note Loss of engineered T cells (red asterisk) and relapse (black asterisk) is indicated.
Figure 6. Clinical responses and durability
Figure 6. Clinical responses and durability
(a). OS and PFS functions estimated by the Kaplan-Meier approach for the 20 patient cohort. Times-to-event are truncated as 24 months. Surviving (censored) patients are represented by tick marks; the number of patients available for assessment at each 6 month interval is indicated below the graph. (b). Swimmer plot showing the duration of clinical response (black bars) and survival following progression (hatched bars). The depth of each response as measured at day 100 on study is represented by the triangles overlying each bar at that time point. The number of gene-marked cells per microliter of blood, as determined by Q-RT-PCR, is noted above each patient bar for day 100, 180 and 360 post-infusion.

References

    1. Tricot G, et al. Graft-versus-myeloma effect: proof of principle. Blood. 1996;87:1196–1198.
    1. Alyea E, et al. T-cell--depleted allogeneic bone marrow transplantation followed by donor lymphocyte infusion in patients with multiple myeloma: induction of graft-versus-myeloma effect. Blood. 2001;98:934–939.
    1. Lokhorst HM, et al. The occurrence of graft-versus-host disease is the major predictive factor for response to donor lymphocyte infusions in multiple myeloma. Blood. 2004;103:4362–4364.
    1. Barlogie B, et al. Superiority of tandem autologous transplantation over standard therapy for previously untreated multiple myeloma. Blood. 1997;89:789–793.
    1. Attal M, et al. A prospective, randomized trial of autologous bone marrow transplantation and chemotherapy in multiple myeloma. Intergroupe Francais du Myelome. The New England journal of medicine. 1996;335:91–97.
    1. Child JA, et al. High-dose chemotherapy with hematopoietic stem-cell rescue for multiple myeloma. The New England journal of medicine. 2003;348:1875–1883.
    1. Porrata LF, et al. Early lymphocyte recovery predicts superior survival after autologous hematopoietic stem cell transplantation in multiple myeloma or non-Hodgkin lymphoma. Blood. 2001;98:579–585.
    1. Porrata LF, Markovic SN. Timely reconstitution of immune competence affects clinical outcome following autologous stem cell transplantation. Clinical and experimental medicine. 2004;4:78–85.
    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 U S A. 2002;99:13009–13013.
    1. Noonan K, et al. Activated marrow-infiltrating lymphocytes effectively target plasma cells and their clonogenic precursors. Cancer research. 2005;65:2026–2034.
    1. Rapoport AP, et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nature medicine. 2005;11:1230–1237.
    1. Rapoport AP, et al. Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma. Blood. 2011;117:788–797.
    1. Rapoport AP, et al. Combination Immunotherapy after ASCT for Multiple Myeloma Using MAGE-A3/Poly-ICLC Immunizations Followed by Adoptive Transfer of Vaccine-Primed and Costimulated Autologous T Cells. Clin Cancer Res. 2014;20:1355–1365.
    1. Rapoport AP, et al. Rapid immune recovery and graft-versus-host disease-like engraftment syndrome following adoptive transfer of Costimulated autologous T cells. Clinical cancer research : an official journal of the American Association for Cancer Research. 2009;15:4499–4507.
    1. Stadtmauer EA, et al. Transfer of influenza vaccine-primed costimulated autologous T cells after stem cell transplantation for multiple myeloma leads to reconstitution of influenza immunity: results of a randomized clinical trial. Blood. 2011;117:63–71.
    1. Rosenblatt J, 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;19:3640–3648.
    1. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nature medicine. 2004;10:909–915.
    1. Aleksic M, et al. Different affinity windows for virus and cancer-specific T-cell receptors: implications for therapeutic strategies. Eur J Immunol. 2012;42:3174–3179.
    1. Purbhoo MA, et al. Quantifying and imaging NY-ESO-1/LAGE-1-derived epitopes on tumor cells using high affinity T cell receptors. Journal of immunology. 2006;176:7308–7316.
    1. Mittal D, Gubin MM, Schreiber RD, Smyth MJ. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Current opinion in immunology. 2014;27:16–25.
    1. Maude SL, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England journal of medicine. 2014;371:1507–1517.
    1. Davila ML, et al. Chimeric antigen receptors for the adoptive T cell therapy of hematologic malignancies. International journal of hematology. 2014;99:361–371.
    1. Lee DW, 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. 2014
    1. Morgan RA, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science. 2006;314:126–129.
    1. Johnson LA, et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood. 2009;114:535–546.
    1. Burns WR, Zheng Z, Rosenberg SA, Morgan RA. Lack of specific gamma-retroviral vector long terminal repeat promoter silencing in patients receiving genetically engineered lymphocytes and activation upon lymphocyte restimulation. Blood. 2009;114:2888–2899.
    1. Li Y, et al. Directed evolution of human T-cell receptors with picomolar affinities by phage display. Nature biotechnology. 2005;23:349–354.
    1. Robbins PF, et al. Single and dual amino acid substitutions in TCR CDRs can enhance antigen-specific T cell functions. Journal of immunology. 2008;180:6116–6131.
    1. Robbins PF, et al. Tumor regression in patients with metastatic synovial cell sarcoma and melanoma using genetically engineered lymphocytes reactive with NY-ESO-1. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2011;29:917–924.
    1. Robbins PF, 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. Clinical cancer research : an official journal of the American Association for Cancer Research. 2014
    1. Hunder NN, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NYESO-1. The New England journal of medicine. 2008;358:2698–2703.
    1. Yuan J, et al. CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A. 2008;105:20410–20415.
    1. van Baren N, et al. Genes encoding tumor-specific antigens are expressed in human myeloma cells. Blood. 1999;94:1156–1164.
    1. Jungbluth AA, 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;106:167–174.
    1. Condomines M, et al. Cancer/testis genes in multiple myeloma: expression patterns and prognosis value determined by microarray analysis. Journal of immunology. 2007;178:3307–3315.
    1. Atanackovic D, et al. Cancer-testis antigens are commonly expressed in multiple myeloma and induce systemic immunity following allogeneic stem cell transplantation. Blood. 2007;109:1103–1112.
    1. van Rhee F, et al. NY-ESO-1 is highly expressed in poor-prognosis multiple myeloma and induces spontaneous humoral and cellular immune responses. Blood. 2005;105:3939–3944.
    1. Hoos A, et al. Improved endpoints for cancer immunotherapy trials. Journal of the National Cancer Institute. 2010;102:1388–1397.
    1. Lee DW, et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood. 2014;124:188–195.
    1. Barrett DM, Teachey DT, Grupp SA. Toxicity management for patients receiving novel T-cell engaging therapies. Current opinion in pediatrics. 2014;26:43–49.
    1. Brentjens RJ, 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. Corrigan-Curay J, et al. T-cell immunotherapy: looking forward. Mol Ther. 2014;22:1564–1574.
    1. Merchant Melinda S., M.C.C., Stadtmauer Edward A., Tap William D., D'Angelo Sandra P., Grupp Stephan A., Holdich Tom, Binder-Scholl Gwendolyn, Jakobsen Bent K, Odunsi Kunle, Rapoport Aaron, Mackall Crystal. American Society of Clinical Oncology. Illinois; Chicago: 2015. Genetically engineered NY-ESO-1 specific T cells in HLA-A201+ patients wtih advanced cancers.
    1. Cameron BJ, et al. Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Science translational medicine. 2013;5:197ra103.
    1. Linette GP, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013;122:863–871.
    1. Kalos M, June CH. Adoptive T cell transfer for cancer immunotherapy in the era of synthetic biology. Immunity. 2013;39:49–60.
    1. Stein S, et al. Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nature medicine. 2010;16:198–204.
    1. Levine BL, et al. Effects of CD28 costimulation on long-term proliferation of CD4+ T cells in the absence of exogenous feeder cells. Journal of immunology. 1997;159:5921–5930.
    1. Scholler J, et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Science translational medicine. 2012;4:132ra153.
    1. Krishnan A, et al. Autologous haemopoietic stem-cell transplantation followed by allogeneic or autologous haemopoietic stem-cell transplantation in patients with multiple myeloma (BMT CTN 0102): a phase 3 biological assignment trial. The lancet oncology. 2011;12:1195–1203.
    1. Sonneveld P, et al. Bortezomib-based versus nonbortezomib-based induction treatment before autologous stem-cell transplantation in patients with previously untreated multiple myeloma: a meta-analysis of phase III randomized, controlled trials. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2013;31:3279–3287.
    1. Richardson P. Novel strategies in the treatment of relapsed/refractory multiple myeloma. From the Multiple Myeloma Research Foundation. Oncology. 2003;17:1063–1065.
    1. Armand P, et al. Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial. J Clin Oncol. 2013;31:4199–4206.
    1. Galustian C, et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer immunology, immunotherapy : CII. 2009;58:1033–1045.
    1. Ramsay AG, et al. Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. The Journal of clinical investigation. 2008;118:2427–2437.
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74.
    1. Rajkumar SV, et al. Consensus recommendations for the uniform reporting of clinical trials: report of the International Myeloma Workshop Consensus Panel 1. Blood. 2011;117:4691–4695.
    1. Mark T, et al. Atypical serum immunofixation patterns frequently emerge in immunomodulatory therapy and are associated with a high degree of response in multiple myeloma. British journal of haematology. 2008;143:654–660.
    1. Dull T, et al. A third-generation lentivirus vector with a conditional packaging system. Journal of virology. 1998;72:8463–8471.
    1. Janetzki S, et al. “MIATA”-minimal information about T cell assays. Immunity. 2009;31:527–528.
    1. Kalos M, et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Science translational medicine. 2011;3:95ra73.
    1. Robins HS, et al. Comprehensive assessment of T-cell receptor beta-chain diversity in alphabeta T cells. Blood. 2009;114:4099–4107.
    1. Rizopoulos D. Joint Models for Longitudinal and Time-to-Event Data. Chapman & Hall/CRC; 2012.

Source: PubMed

3
Abonnieren