PD-1(hi)TIM-3(+) T cells associate with and predict leukemia relapse in AML patients post allogeneic stem cell transplantation

Y Kong, J Zhang, D F Claxton, W C Ehmann, W B Rybka, L Zhu, H Zeng, T D Schell, H Zheng, Y Kong, J Zhang, D F Claxton, W C Ehmann, W B Rybka, L Zhu, H Zeng, T D Schell, H Zheng

Abstract

Prognosis of leukemia relapse post allogeneic stem cell transplantation (alloSCT) is poor and effective new treatments are urgently needed. T cells are pivotal in eradicating leukemia through a graft versus leukemia (GVL) effect and leukemia relapse is considered a failure of GVL. T-cell exhaustion is a state of T-cell dysfunction mediated by inhibitory molecules including programmed cell death protein 1 (PD-1) and T-cell immunoglobulin domain and mucin domain 3 (TIM-3). To evaluate whether T-cell exhaustion and inhibitory pathways are involved in leukemia relapse post alloSCT, we performed phenotypic and functional studies on T cells from peripheral blood of acute myeloid leukemia patients receiving alloSCT. Here we report that PD-1(hi)TIM-3(+) cells are strongly associated with leukemia relapse post transplantation. Consistent with exhaustion, PD-1(hi)TIM-3(+) T cells are functionally deficient manifested by reduced production of interleukin 2 (IL-2), tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ). In addition, these cells demonstrate a phenotype consistent with exhausted antigen-experienced T cells by losing TN and TEMRA subsets. Importantly, increase of PD-1(hi)TIM-3(+) cells occurs before clinical diagnosis of leukemia relapse, suggesting their predictive value. Results of our study provide an early diagnostic approach and a therapeutic target for leukemia relapse post transplantation.

Figures

Figure 1
Figure 1
Expression of PD-1 and TIM3 is enhanced on T cells from patients with leukemia relapse post transplant. Flow cytometry analysis of surface expression of BTLA, 2B4, LAG3, CD160, TIM-3 and PD-1 was performed on PBMCs collected from AML patients post alloSCT. (a–g) Histogram displays the expression of above receptors on T cells from representative patients of relapse vs remission. CD4+ or CD8+ cells were gated. Percentage of cells expressing each receptor is shown. Panels on right are plots of expression of each receptor on CD4+ or CD8+ T cells from patients with relapse (n=4–5) vs remission (n=6). Each square represents data from one patient. **P<0.01.
Figure 2
Figure 2
PD-1hiTIM3+ cells associate with leukemia relapse post alloSCT. PBMCs from patients with leukemia relapse vs remission were tested for PD-1 and TIM3 expression on CD4+ and CD8+ T cells by flow cytometry. (a) Based on levels of PD-1 and TIM3 expression, cells are divided into six fractions. Shown is the schema of each fraction. (b) Representative flow data from one relapse (patient 09) and one remission (patient 02). Percentage of PD-1hiTIM-3+ among CD4+ or CD8+ cells is shown. (c) Plot of percentage of each fraction among CD4+ or CD8+ cells in patient with relapse (n=5) vs remission (n=6). Each square represents data of an individual patient. *P<0.05, **P<0.01.
Figure 3
Figure 3
PD-1hiTIM3+ cells produce less TNF-a and IL-2 upon PMA/ionomycin stimulation. Production of TNF-α, IFN-γ and IL-2 upon in vitro PMA/ionomycin stimulation. Shown is cytokine release from CD4+ (a) or CD8+ (b) T cells gated on each fraction of cells based on PD-1 and TIM-3 expression. Representative plots from a single relapse patient (09) are shown.
Figure 4
Figure 4
PD-1hiTIM3+ cells produce less cytokines in response to anti-CD3/anti-CD28 stimulation. Production of TNF-α, IFN-γ and IL-2 upon in vitro anti-CD3/anti-CD28 stimulation. Shown is cytokine release from CD4+ (a) or CD8+ (b) T cells gated on each fraction of cells based on PD-1 and TIM-3 expression. Representative plots from a single relapse patient (09) are shown.
Figure 5
Figure 5
PD-1hiTIM3+ cells lose TEMRA in patients with leukemia relapse post transplant. Distribution of TN, TCM, TEM and TEMRA in T cells gated on each fraction of cells based on PD-1 and TIM-3 expression from relapse patients. (a) Representative dot plots from one relapse patient (09). (b) Summary data for five relapse patients (08, 09, 10, 11 and 12).
Figure 6
Figure 6
Increase of PD-1hiTIM3+ cells predict leukemia relapse. PBMCs from a patient (08) who had leukemia relapse 5 month post alloSCT were collected at different time points post transplant. (a) Flow cytometry data of expression of PD-1hiTIM3+ cells among CD4+ or CD8+ T cells. (b) Plot of the percentage of PD-1hiTIM-3+ cells among CD4+ or CD8+ T cells kinetically post transplantation.

References

    1. Bishop MR, Alyea EP, 3rd, Cairo MS, Falkenburg JH, June CH, Kroger N, et al. National Cancer Institute's First International Workshop on the Biology, Prevention, and Treatment of Relapse after Allogeneic Hematopoietic Stem Cell Transplantation: summary and recommendations from the organizing committee. Biol Blood Marrow Transplant. 2011;17:443–454.
    1. Frassoni F, Barrett AJ, Granena A, Ernst P, Garthon G, Kolb HJ, et al. Relapse after allogeneic bone marrow transplantation for acute leukaemia: a survey by the E.B.M.T. of 117 cases. Br J Haematol. 1988;70:317–320.
    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;13:1160–1168.
    1. Liu HT, Liu DH, Huang XJ, Artz A, Bishop MR. Treatments of disease relapse after allogeneic stem cell transplantation focusing on donor lymphocyte infusion. Chin Med J. 2013;126:4380–4388.
    1. Elmaagacli AH, Beelen DW, Trenn G, Schmidt O, Nahler M, Schaefer UW. Induction of a graft-versus-leukemia reaction by cyclosporin A withdrawal as immunotherapy for leukemia relapsing after allogeneic bone marrow transplantation. Bone Marrow Transplant. 1999;23:771–777.
    1. Levine JE, Braun T, Penza SL, Beatty P, Cornetta K, Martino R, et al. Prospective trial of chemotherapy and donor leukocyte infusions for relapse of advanced myeloid malignancies after allogeneic stem-cell transplantation. J Clin Oncol. 2002;20:405–412.
    1. Choi SJ, Lee JH, Kim S, Seol M, Lee YS, Lee JS, et al. Treatment of relapsed acute myeloid leukemia after allogeneic bone marrow transplantation with chemotherapy followed by G-CSF-primed donor leukocyte infusion: a high incidence of isolated extramedullary relapse. Leukemia. 2004;18:1789–1797.
    1. Pollyea DA, Artz AS, Stock W, Daugherty C, Godley L, Odenike OM, et al. Outcomes of patients with AML and MDS who relapse or progress after reduced intensity allogeneic hematopoietic cell transplantation. Bone Marrow Transplant. 2007;40:1027–1032.
    1. Bosi A, Laszlo D, Labopin M, Reffeirs J, Michallet M, Gluckman E, et al. Second allogeneic bone marrow transplantation in acute leukemia: results of a survey by the European Cooperative Group for Blood and Marrow Transplantation. J Clin Oncol. 2001;19:3675–3684.
    1. Eapen M, Giralt SA, Horowitz MM, Klein JP, Wagner JE, Zhang MJ, et al. Second transplant for acute and chronic leukemia relapsing after first HLA-identical sibling transplant. Bone Marrow Transplant. 2004;34:721–727.
    1. Schmid C, Labopin M, Nagler A, Niederwieser D, Castagna L, Tabrizi R, et al. Treatment, risk factors, and outcome of adults with relapsed AML after reduced intensity conditioning for allogeneic stem cell transplantation. Blood. 2012;119:1599–1606.
    1. Kolb HJ. Graft-versus-leukemia effects of transplantation and donor lymphocytes. Blood. 2008;112:4371–4383.
    1. Bleakley M, Riddell SR. Molecules and mechanisms of the graft-versus-leukaemia effect. Nat Rev Cancer. 2004;4:371–380.
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454.
    1. Weber JS, Kudchadkar RR, Yu B, Gallenstein D, Horak CE, Inzunza HD, et al. Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J Clin Oncol. 2013;31:4311–4318.
    1. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369:122–133.
    1. Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, Kefford R, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014;384:1109–1117.
    1. Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12:492–499.
    1. Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J Virol. 2003;77:4911–4927.
    1. Wherry EJ, Ahmed R. Memory CD8 T-cell differentiation during viral infection. J Virol. 2004;78:5535–5545.
    1. Fuller MJ, Zajac AJ. Ablation of CD8 and CD4 T cell responses by high viral loads. J Immunol. 2003;170:477–486.
    1. Virgin HW, Wherry EJ, Ahmed R. Redefining chronic viral infection. Cell. 2009;138:30–50.
    1. Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJ, Suresh M, Altman JD, et al. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med. 1998;188:2205–2213.
    1. Trautmann L, Janbazian L, Chomont N, Said EA, Gimmig S, Bessette B, et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med. 2006;12:1198–1202.
    1. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–1544.
    1. Baitsch L, Baumgaertner P, Devevre E, Raghav SK, Legat A, Barba L, et al. Exhaustion of tumor-specific CD8(+) T cells in metastases from melanoma patients. J Clin Invest. 2011;121:2350–2360.
    1. Riches JC, Davies JK, McClanahan F, Fatah R, Iqbal S, Agrawal S, et al. T cells from CLL patients exhibit features of T-cell exhaustion but retain capacity for cytokine production. Blood. 2013;121:1612–1621.
    1. Kozako T, Yoshimitsu M, Fujiwara H, Masamoto I, Horai S, White Y, et al. PD-1/PD-L1 expression in human T-cell leukemia virus type 1 carriers and adult T-cell leukemia/lymphoma patients. Leukemia. 2009;23:375–382.
    1. Mumprecht S, Schurch C, Schwaller J, Solenthaler M, Ochsenbein AF. Programmed death 1 signaling on chronic myeloid leukemia-specific T cells results in T-cell exhaustion and disease progression. Blood. 2009;114:1528–1536.
    1. Zhou Q, Munger ME, Veenstra RG, Weigel BJ, Hirashima M, Munn DH, et al. Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood. 2011;117:4501–4510.
    1. Bachireddy P, Hainz U, Rooney M, Pozdnyakova O, Aldridge J, Zhang W, et al. Reversal of in situ T-cell exhaustion during effective human antileukemia responses to donor lymphocyte infusion. Blood. 2014;123:1412–1421.
    1. Truneh A, Albert F, Golstein P, Schmitt-Verhulst AM. Early steps of lymphocyte activation bypassed by synergy between calcium ionophores and phorbol ester. Nature. 1985;313:318–320.
    1. Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712.
    1. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–763.
    1. Brusa D, Serra S, Coscia M, Rossi D, D'Arena G, Laurenti L, et al. The PD-1/PD-L1 axis contributes to T-cell dysfunction in chronic lymphocytic leukemia. Haematologica. 2013;98:953–963.
    1. Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood. 2009;114:1545–1552.
    1. Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng QR, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2014;28:1280–1288.
    1. Norde WJ, Maas F, Hobo W, Korman A, Quigley M, Kester MG, et al. PD-1/PD-L1 interactions contribute to functional T-cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation. Cancer Res. 2011;71:5111–5122.
    1. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med. 2015;372:311–319.
    1. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015;348:56–61.
    1. Blazar BR, Carreno BM, Panoskaltsis-Mortari A, Carter L, Iwai Y, Yagita H, et al. Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism. J Immunol. 2003;171:1272–1277.
    1. Luhder F, Hoglund P, Allison JP, Benoist C, Mathis D. Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) regulates the unfolding of autoimmune diabetes. J Exp Med. 1998;187:427–432.
    1. Wang HB, Shi FD, Li H, Chambers BJ, Link H, Ljunggren HG. Anti-CTLA-4 antibody treatment triggers determinant spreading and enhances murine myasthenia gravis. J Immunol. 2001;166:6430–6436.
    1. Kyi C, Postow MA. Checkpoint blocking antibodies in cancer immunotherapy. FEBS Lett. 2014;588:368–376.
    1. Bashey A, Medina B, Corringham S, Pasek M, Carrier E, Vrooman L, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood. 2009;113:1581–1588.
    1. Anderson BE, McNiff J, Yan J, Doyle H, Mamula M, Shlomchik MJ, et al. Memory CD4+ T cells do not induce graft-versus-host disease. J Clin Invest. 2003;112:101–108.
    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;103:1534–1541.
    1. Zhang Y, Joe G, Zhu J, Carroll R, Levine B, Hexner E, 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;103:3970–3978.
    1. Zheng H, Matte-Martone C, Li H, Anderson BE, Venketesan S, Sheng Tan H, et al. Effector memory CD4+ T cells mediate graft-versus-leukemia without inducing graft-versus-host disease. Blood. 2008;111:2476–2484.
    1. Koestner W, Hapke M, Herbst J, Klein C, Welte K, Fruehauf J, et al. PD-L1 blockade effectively restores strong graft-versus-leukemia effects without graft-versus-host disease after delayed adoptive transfer of T-cell receptor gene-engineered allogeneic CD8+ T cells. Blood. 2011;117:1030–1041.
    1. Eapen M, O'Donnell P, Brunstein CG, Wu J, Barowski K, Mendizabal A, et al. Mismatched related and unrelated donors for allogeneic hematopoietic cell transplantation for adults with hematologic malignancies. Biol Blood Marrow Transplant. 2014;20:1485–1492.
    1. Laughlin MJ, Barker J, Bambach B, Koc ON, Rizzieri DA, Wagner JE, et al. Hematopoietic engraftment and survival in adult recipients of umbilical-cord blood from unrelated donors. N Engl J Med. 2001;344:1815–1822.
    1. Laughlin MJ, Eapen M, Rubinstein P, Wagner JE, Zhang MJ, Champlin RE, et al. Outcomes after transplantation of cord blood or bone marrow from unrelated donors in adults with leukemia. N Engl J Med. 2004;351:2265–2275.
    1. Chapuis AG, Ragnarsson GB, Nguyen HN, Chaney CN, Pufnock JS, Schmitt TM, et al. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci Transl Med. 2013;5:174ra27.
    1. Dao T, Liu C, Scheinberg DA. Approaching untargetable tumor-associated antigens with antibodies. Oncoimmunology. 2013;2:e24678.
    1. Van Driessche A, Gao L, Stauss HJ, Ponsaerts P, Van Bockstaele DR, Berneman ZN, et al. Antigen-specific cellular immunotherapy of leukemia. Leukemia. 2005;19:1863–1871.
    1. Gros A, Robbins PF, Yao X, Li YF, Turcotte S, Tran E, et al. PD-1 identifies the patient-specific CD8(+) tumor-reactive repertoire infiltrating human tumors. J Clin Invest. 2014;124:2246–2259.
    1. Dziubianau M, Hecht J, Kuchenbecker L, Sattler A, Stervbo U, Rodelsperger C, et al. TCR repertoire analysis by next generation sequencing allows complex differential diagnosis of T cell-related pathology. Am J Transplant. 2013;13:2842–2854.

Source: PubMed

3
Abonnieren