Infusion of donor-derived CD8+ memory T cells for relapse following allogeneic hematopoietic cell transplantation

Lori Muffly, Kevin Sheehan, Randall Armstrong, Kent Jensen, Keri Tate, Andrew R Rezvani, David Miklos, Sally Arai, Judith Shizuru, Laura Johnston, Everett Meyer, Wen-Kai Weng, Ginna G Laport, Robert S Negrin, Sam Strober, Robert Lowsky, Lori Muffly, Kevin Sheehan, Randall Armstrong, Kent Jensen, Keri Tate, Andrew R Rezvani, David Miklos, Sally Arai, Judith Shizuru, Laura Johnston, Everett Meyer, Wen-Kai Weng, Ginna G Laport, Robert S Negrin, Sam Strober, Robert Lowsky

Abstract

Murine models showed that CD8+CD44hi memory T (TM) cells could eradicate malignant cells without inducing graft-versus-host disease (GVHD). We evaluated the feasibility and safety of infusing freshly isolated and purified donor-derived phenotypic CD8+ TM cells into adults with disease relapse after allogeneic hematopoietic cell transplantation (HCT). Phenotypic CD8 TM cells were isolated after unmobilized donor apheresis using a tandem immunomagnetic selection strategy of CD45RA depletion followed by CD8+ enrichment. Fifteen patients received CD8+ TM cells at escalating doses (1 × 106, 5 × 106, or 10 × 106 cells per kg). Thirteen received cytoreduction before CD8+ TM cell infusion, and 9 had active disease at the time of infusion. Mean yield and purity of the CD8+ TM infusion were 38.1% and 92.8%, respectively; >90% had CD8+ T effector memory phenotype, cytokine expression, and secretion profile. No adverse infusional events or dose-limiting toxicities occurred; GVHD developed in 1 patient (grade 2 liver). Ten patients (67%) maintained or achieved response (7 complete response, 1 partial response, 2 stable disease) for at least 3 months after infusion; 4 of the responders had active disease at the time of infusion. With a median follow-up from infusion of 328 days (range, 118-1328 days), median event-free survival and overall survival were 4.9 months (95% confidence interval [CI], 1-19.3 months) and 19.6 months (95% CI, 5.6 months to not reached), respectively. Collection and enrichment of phenotypic CD8+ TM cells is feasible, well tolerated, and associated with a low incidence of GVHD when administered as a manipulated infusion of donor lymphocytes in patients who have relapsed after HCT. This trial was registered at www.clinicaltrials.gov as #NCT01523223.

Conflict of interest statement

Conflict-of-interest disclosure: The authors declare no competing financial interests.

© 2018 by The American Society of Hematology.

Figures

Graphical abstract
Graphical abstract
Figure 1.
Figure 1.
Representative flow analysis of peripheral blood apheresis collections from the preselection, post-CD45RA depletion, and CD8+enrichment steps. Cells were stained for expression of CD4, CD8, CD45RA, and CD45RO. Plots show CD45 gated events.
Figure 2.
Figure 2.
Flow cytometric analysis, cytokine expression, mixed lymphocyte reaction, and cytokine secretion. (A) Representative flow analysis of 1 experiment repeated 5 times that shows cell composition and subsets from start to finish of preselection, post–CD45RA depletion, and post–CD8+ enrichment. Cells were stained for expression of CD45RA, CD45RO, and CD62L, and plots are shown for CD4+ and CD8+ gated cells at each step of the processing procedure. The surface phenotype of the CD45RA–CD8+-enriched cells is consistent with the phenotypic CD8+ TEM subset being predominantly CD45RA–CD45RO+CD62L–. The TEM cells ranged from 81.7% to 98.1% of the final cell composition. (B) Flow cytometric analysis of cytokine expression by enriched cell subsets. Cells were activated by ionomycin and phorbol myristate acetate, treated with monensin, and stained for expression of CD45, CD4, CD8, and CD45RA. Cells were fixed, permeabilized, and stained for INF-γ, and IL-2. Plots show comparison of IL-2 and IFN-γ expression in CD8+CD45RA–, CD8+CD45RA+, and CD4+CD45RA– cells as indicated. (C) Proliferation of CD8+CD45R– and CD4+ cells activated by co-culture with or without irradiated allogeneic stimulators and assessed by 3H-thymidine uptake during the last 24 hours of culture. Mean and standard deviations are shown (n = 4). (D) Cytokine secretion assessment in CD8+CD45RA– and CD4+ cells activated by co-culture with or without irradiated allogeneic stimulators. Supernatants from 7-day cultures were analyzed by flow cytometry using cytokine bead arrays for INF-γ, IL-2, and TNF-α (n = 4). Means with standard deviations are shown. Teff, T effector cells.
Figure 3.
Figure 3.
Kaplan-Meier estimates of event-free survival (EFS) for the 15 patients receiving CD8+TMcell infusion.
Figure 4.
Figure 4.
Kaplan-Meier estimates of overall survival (OS) for the 15 patients receiving CD8+TMcell infusion.

References

    1. Wingard JR, Majhail NS, Brazauskas R, et al. . Long-term survival and late deaths after allogeneic hematopoietic cell transplantation. J Clin Oncol. 2011;29(16):2230-2239.
    1. D’Souza A, Zhu X Current Uses and Outcomes of Hematopoietic Cell Transplantation (HCT): CIBMTR Summary Slides, 2016. . Accessed 1 June 2017.
    1. Mielcarek M, Storer BE, Flowers ME, Storb R, Sandmaier BM, Martin PJ. Outcomes among patients with recurrent high-risk hematologic malignancies after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2007;13(10):1160-1168.
    1. Oran B, Giralt S, Couriel D, et al. . Treatment of AML and MDS relapsing after reduced-intensity conditioning and allogeneic hematopoietic stem cell transplantation. Leukemia. 2007;21(12):2540-2544.
    1. Schmid C, Labopin M, Nagler A, et al. . Treatment, risk factors, and outcome of adults with relapsed AML after reduced intensity conditioning for allogeneic stem cell transplantation. Blood. 2012;119(6):1599-1606.
    1. Poon LM, Hamdi A, Saliba R, et al. . Outcomes of adults with acute lymphoblastic leukemia relapsing after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2013;19(7):1059-1064.
    1. Bejanyan N, Weisdorf DJ, Logan BR, et al. . Survival of patients with acute myeloid leukemia relapsing after allogeneic hematopoietic cell transplantation: a Center for International Blood and Marrow Transplant research study. Biol Blood Marrow Transplant. 2015;21(3):454-459.
    1. Schmid C, Labopin M, Nagler A, et al. . Donor lymphocyte infusion in the treatment of first hematological relapse after allogeneic stem-cell transplantation in adults with acute myeloid leukemia: a retrospective risk factors analysis and comparison with other strategies by the EBMT Acute Leukemia Working Party. J Clin Oncol. 2007;25(31):4938-4945.
    1. Kolb HJ, Mittermüller J, Clemm C, et al. . Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood. 1990;76(12):2462-2465.
    1. Bär BM, Schattenberg A, Mensink EJ, et al. . Donor leukocyte infusions for chronic myeloid leukemia relapsed after allogeneic bone marrow transplantation. J Clin Oncol. 1993;11(3):513-519.
    1. Kolb HJ, Schattenberg A, Goldman JM, et al. ; European Group for Blood and Marrow Transplantation Working Party Chronic Leukemia. Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood. 1995;86(5):2041-2050.
    1. Collins RH Jr, Shpilberg O, Drobyski WR, et al. . Donor leukocyte infusions in 140 patients with relapsed malignancy after allogeneic bone marrow transplantation. J Clin Oncol. 1997;15(2):433-444.
    1. Mackinnon S, Papadopoulos EB, Carabasi MH, et al. . Adoptive immunotherapy evaluating escalating doses of donor leukocytes for relapse of chronic myeloid leukemia after bone marrow transplantation: separation of graft-versus-leukemia responses from graft-versus-host disease. Blood. 1995;86(4):1261-1268.
    1. Alyea EP, Soiffer RJ, Canning C, et al. . Toxicity and efficacy of defined doses of CD4(+) donor lymphocytes for treatment of relapse after allogeneic bone marrow transplant. Blood. 1998;91(10):3671-3680.
    1. Giralt S, Hester J, Huh Y, et al. . CD8-depleted donor lymphocyte infusion as treatment for relapsed chronic myelogenous leukemia after allogeneic bone marrow transplantation. Blood. 1995;86(11):4337-4343.
    1. Anderson BE, McNiff J, Yan J, et al. . Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest. 2003;112(1):101-108.
    1. Zheng H, Matte-Martone C, Li H, et al. . Effector memory CD4+ T cells mediate graft-versus-leukemia without inducing graft-versus-host disease. Blood. 2008;111(4):2476-2484.
    1. Zhang Y, Joe G, Zhu J, et al. . Dendritic cell-activated CD44hiCD8+ T cells are defective in mediating acute graft-versus-host disease but retain graft-versus-leukemia activity. Blood. 2004;103(10):3970-3978.
    1. Chen BJ, Cui X, Sempowski GD, Liu C, Chao NJ. Transfer of allogeneic CD62L- memory T cells without graft-versus-host disease. Blood. 2004;103(4):1534-1541.
    1. Zheng H, Matte-Martone C, Jain D, McNiff J, Shlomchik WD. Central memory CD8+ T cells induce graft-versus-host disease and mediate graft-versus-leukemia. J Immunol. 2009;182(10):5938-5948.
    1. Dutt S, Tseng D, Ermann J, et al. . Naive and memory T cells induce different types of graft-versus-host disease. J Immunol. 2007;179(10):6547-6554.
    1. Dutt S, Baker J, Kohrt HE, et al. . CD8+CD44(hi) but not CD4+CD44(hi) memory T cells mediate potent graft antilymphoma activity without GVHD. Blood. 2011;117(11):3230-3239.
    1. Edinger M, Hoffmann P, Ermann J, et al. . CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med. 2003;9(9):1144-1150.
    1. Strober S, Benike C, Krishnaswamy S, Engleman EG, Grumet FC. Clinical transplantation tolerance twelve years after prospective withdrawal of immunosuppressive drugs: studies of chimerism and anti-donor reactivity. Transplantation. 2000;69(8):1549-1554.
    1. Hikono H, Kohlmeier JE, Takamura S, Wittmer ST, Roberts AD, Woodland DL. Activation phenotype, rather than central- or effector-memory phenotype, predicts the recall efficacy of memory CD8+ T cells. J Exp Med. 2007;204(7):1625-1636.
    1. Lefrançois L, Obar JJ. Once a killer, always a killer: from cytotoxic T cell to memory cell. Immunol Rev. 2010;235(1):206-218.
    1. Chen BJ, Deoliveira D, Cui X, et al. . Inability of memory T cells to induce graft-versus-host disease is a result of an abortive alloresponse. Blood. 2007;109(7):3115-3123.
    1. Xystrakis E, Bernard I, Dejean AS, Alsaati T, Druet P, Saoudi A. Alloreactive CD4 T lymphocytes responsible for acute and chronic graft-versus-host disease are contained within the CD45RChigh but not the CD45RClow subset. Eur J Immunol. 2004;34(2):408-417.
    1. Bar M, Sandmaier BM, Inamoto Y, et al. . Donor lymphocyte infusion for relapsed hematological malignancies after allogeneic hematopoietic cell transplantation: prognostic relevance of the initial CD3+ T cell dose. Biol Blood Marrow Transplant. 2013;19(6):949-957.
    1. Boumédine RS, Roy DC. Elimination of alloreactive T cells using photodynamic therapy. Cytotherapy. 2005;7(2):134-143.
    1. Mielke S, Solomon SR, Barrett AJ. Selective depletion strategies in allogeneic stem cell transplantation. Cytotherapy. 2005;7(2):109-115.
    1. Willinger T, Freeman T, Hasegawa H, McMichael AJ, Callan MF. Molecular signatures distinguish human central memory from effector memory CD8 T cell subsets. J Immunol. 2005;175(9):5895-5903.
    1. Kolb HJ, Schattenberg A, Goldman JM, et al. . Graft-versus-leukemia effect of donor lymphocyte transfusions in marrow grafted patients. Blood. 1995;86(5):2041-2050.
    1. Bleakley M, Heimfeld S, Loeb KR, et al. . Outcomes of acute leukemia patients transplanted with naive T cell-depleted stem cell grafts. J Clin Invest. 2015;125(7):2677-2689.
    1. Touzot F, Neven B, Dal-Cortivo L, et al. . CD45RA depletion in HLA-mismatched allogeneic hematopoietic stem cell transplantation for primary combined immunodeficiency: A preliminary study. J Allergy Clin Immunol. 2015;135(5):1303-1309.e1-e3.
    1. Pillai A, Teo P, George T, Mukhopadhyay A, Dejbakhsh-Jones S, Strober S. Alloantigen recognition is critical for CD8 T cell-mediated graft anti-tumor activity against murine BCL1 lymphoma after myeloablative bone marrow transplantation. Bone Marrow Transplant. 2007;40(5):487-497.
    1. Ford ML, Kirk AD, Larsen CP. Donor-reactive T-cell stimulation history and precursor frequency: barriers to tolerance induction. Transplantation. 2009;87(9 Suppl):S69-S74.
    1. Adams AB, Pearson TC, Larsen CP. Heterologous immunity: an overlooked barrier to tolerance. Immunol Rev. 2003;196:147-160.
    1. Brehm MA, Markees TG, Daniels KA, Greiner DL, Rossini AA, Welsh RM. Direct visualization of cross-reactive effector and memory allo-specific CD8 T cells generated in response to viral infections. J Immunol. 2003;170(8):4077-4086.
    1. Murali-Krishna K, Ahmed R.. Cutting edge: naive T cells masquerading as memory cells. J Immunol. 2000;165(4):1733-1737.
    1. Bekisz J, Sato Y, Johnson C, Husain SR, Puri RK, Zoon KC. Immunomodulatory effects of interferons in malignancies. J Interferon Cytokine Res. 2013;33(4):154-161.
    1. Holtan SG, DeFor TE, Lazaryan A, et al. . Composite end point of graft-versus-host disease-free, relapse-free survival after allogeneic hematopoietic cell transplantation. Blood. 2015;125(8):1333-1338.
    1. Kohrt HE, Müller A, Baker J, et al. . Donor immunization with WT1 peptide augments antileukemic activity after MHC-matched bone marrow transplantation. Blood. 2011;118(19):5319-5329.

Source: PubMed

3
Subscribe