A multicenter, phase I study of nivolumab for relapsed hematologic malignancies after allogeneic transplantation.

CTLA-4 blockade augments the graft-vs-tumor effect in relapsed hematologic malignancies (HMs) after allogeneic hematopoietic cell transplantation (alloHCT). PD-1/PDL-1 interactions also contribute to functional T cell impairment, but retrospective studies of anti-PD-1 therapy following alloHCT reported substantial toxicity from GVHD. Here, we report the results of a prospective clinical trial of PD-1 blockade for relapsed HMs after alloHCT (ClinicalTrials.gov, NCT01822509). The primary objectives in this phase I multicenter, investigator-initiated study were to determine maximum tolerated dose and safety. Secondary objectives were to assess efficacy and immunologic activity. Patients with relapsed HMs following alloHCT were eligible. Nivolumab was administered every 2 weeks until progression or unacceptable toxicity, starting with a 1 mg/kg cohort, with planned de-escalation based on toxicity to a 0.5 mg/kg cohort. Twenty-eight patients were treated (n=19 myeloid, n=9 lymphoid). Median age was 57 (range 27-76), and median time from alloHCT to enrollment was 21 months (range 5.6-108.5). Two of six patients treated at 1 mg/kg experienced dose-limiting toxicity (DLT) from immune-related adverse events (irAEs). Twenty-two patients were treated at 0.5 mg/kg, and 4 DLTs occurred, including 2 irAEs and 2 with fatal GVHD. The overall response rate in efficacy evaluable patients was 32% (8/25). With a median follow-up of 11 months, the 1-year PFS and OS were 23% and 56%, respectively. In this first prospective clinical trial of an anti-PD-1 antibody for post-alloHCT relapse, GVHD and irAEs occurred, requiring dose de-escalation, with only modest anti-tumor activity. Further studies of anti-PD-1 therapy post alloHCT may require specific toxicity mitigation strategies.

[1]  J. McCarty,et al.  Managing post allograft relapse of myeloid neoplasms: azacitidine and donor lymphocyte infusions as salvage therapy , 2019, Leukemia & lymphoma.

[2]  M. Brock,et al.  Addition of Low-Dose Decitabine to Anti-PD-1 Antibody Camrelizumab in Relapsed/Refractory Classical Hodgkin Lymphoma. , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[3]  G. Tonon,et al.  Immune signature drives leukemia escape and relapse after hematopoietic cell transplantation , 2019, Nature Medicine.

[4]  C. Craddock,et al.  Combination Lenalidomide and Azacitidine: A Novel Salvage Therapy in Patients Who Relapse After Allogeneic Stem-Cell Transplantation for Acute Myeloid Leukemia , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[5]  R. Claus,et al.  Decitabine in combination with donor lymphocyte infusions can induce remissions in relapsed myeloid malignancies with higher leukemic burden after allogeneic hematopoietic cell transplantation. , 2018, Leukemia research : a Forum for Studies on Leukemia and Normal Hemopoiesis.

[6]  L. Obéric,et al.  Efficacy of chemotherapy or chemo‐anti‐PD‐1 combination after failed anti‐PD‐1 therapy for relapsed and refractory hodgkin lymphoma: A series from lysa centers , 2018, American journal of hematology.

[7]  M. Davids,et al.  Tyrosine kinase inhibitors and immune checkpoint blockade in allogeneic hematopoietic cell transplantation. , 2018, Blood.

[8]  P. Hari,et al.  PD-1 blockade for relapsed lymphoma post-allogeneic hematopoietic cell transplant: high response rate but frequent GVHD. , 2017, Blood.

[9]  R. Houot,et al.  Efficacy and tolerability of nivolumab after allogeneic transplantation for relapsed Hodgkin lymphoma. , 2017, Blood.

[10]  R. Advani,et al.  Refinement of the Lugano Classification lymphoma response criteria in the era of immunomodulatory therapy. , 2016, Blood.

[11]  Catherine J. Wu,et al.  Ipilimumab for Patients with Relapse after Allogeneic Transplantation. , 2016, The New England journal of medicine.

[12]  J. Aster,et al.  Validation and Implementation of a Custom Next-Generation Sequencing Clinical Assay for Hematologic Malignancies. , 2016, The Journal of molecular diagnostics : JMD.

[13]  G. Freeman,et al.  Programmed death ligand-1 expression on donor T cells drives graft-versus-host disease lethality. , 2016, The Journal of clinical investigation.

[14]  J. Ritz,et al.  Unbalanced recovery of regulatory and effector T cells after allogeneic stem cell transplantation contributes to chronic GVHD. , 2016, Blood.

[15]  P. Bousso,et al.  The PD-1 Axis Enforces an Anatomical Segregation of CTL Activity that Creates Tumor Niches after Allogeneic Hematopoietic Stem Cell Transplantation. , 2016, Immunity.

[16]  S. Devine,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. , 2015, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[17]  P. Armand,et al.  Outcome and prognostic factors for patients who relapse after allogeneic hematopoietic stem cell transplantation. , 2013, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[18]  G. Freeman,et al.  Host programmed death ligand 1 is dominant over programmed death ligand 2 expression in regulating graft-versus-host disease lethality. , 2013, Blood.

[19]  A. Korman,et al.  PD-1/PD-L1 interactions contribute to functional T-cell impairment in patients who relapse with cancer after allogeneic stem cell transplantation. , 2011, Cancer research.

[20]  E John Wherry,et al.  T cell exhaustion , 2011 .

[21]  C. Klein,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. , 2011, Blood.

[22]  W. Shannon,et al.  In vivo administration of hypomethylating agents mitigate graft-versus-host disease without sacrificing graft-versus-leukemia. , 2010, Blood.

[23]  J. Radich,et al.  Chronic myeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[24]  I. Lowy,et al.  CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. , 2009, Blood.

[25]  Michael Hallek,et al.  Guidelines for the diagnosis and treatment of chronic lymphocytic leukemia: a report from the International Workshop on Chronic Lymphocytic Leukemia updating the National Cancer Institute-Working Group 1996 guidelines. , 2008, Blood.

[26]  Sigrid Stroobants,et al.  Revised response criteria for malignant lymphoma. , 2007, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[27]  B. Barlogie,et al.  International uniform response criteria for multiple myeloma , 2006, Leukemia.

[28]  B. Cheson,et al.  Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. , 2006, Blood.

[29]  C. Bloomfield,et al.  Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. , 2003, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[30]  A. Panoskaltsis-Mortari,et al.  Blockade of Programmed Death-1 Engagement Accelerates Graft-Versus-Host Disease Lethality by an IFN-γ-Dependent Mechanism1 , 2003, The Journal of Immunology.

[31]  A. Sharpe,et al.  Opposing roles of CD28:B7 and CTLA-4:B7 pathways in regulating in vivo alloresponses in murine recipients of MHC disparate T cells. , 1999, Journal of immunology.