Leukemia vaccine overcomes limitations of checkpoint blockade by evoking clonal T-cell responses in a murine acute myeloid leukemia model

We have developed a personalized vaccine in which patientderived leukemia cells are fused to autologous dendritic cells, evoking a polyclonal T-cell response against shared antigens and neoantigens. We postulated that the dendritic cell/acute myeloid leukemia fusion vaccine would demonstrate synergy with checkpoint blockade by expanding tumor antigen-specific lymphocytes that would provide a critical substrate for checkpoint blockade-mediated activation. Using an immunocompetent murine leukemia model, we examined the immunological response to and therapeutic efficacy of vaccination in conjunction with checkpoint blockade with respect to leukemia engraftment, disease burden, survival and the induction of tumor-specific immunity. Mice treated with checkpoint blockade alone had rapid progression of leukemia and only a modest extension of survival. Vaccination with dendritic cell/acute myeloid leukemia fusions resulted in the expansion of tumor-specific lymphocytes and disease eradication in a subset of animals, while the combination of vaccination and checkpoint blockade induced a fully protective tumor-specific immune response in all treated animals. Vaccination followed by checkpoint blockade resulted in upregulation of genes regulating activation and proliferation in memory and effector T cells. Long-term survivors exhibited increased T-cell clonal diversity and were resistant to subsequent tumor challenge. The combination of dendritic cell/acute myeloid leukemia fusion vaccine and checkpoint blockade treatment offers unique synergy, inducing durable activation of leukemia-specific immunity, protection from lethal tumor challenge and the selective expansion of tumor-reactive clones.

[1]  G. Kochan,et al.  Functional systemic CD4 immunity is required for clinical responses to PD‐L1/PD‐1 blockade therapy , 2019, EMBO molecular medicine.

[2]  J. Clohessy,et al.  Vulnerabilities in mIDH2 AML confer sensitivity to APL-like targeted combination therapy , 2019, Cell Research.

[3]  A. Goldberg,et al.  Immune Checkpoint Inhibitors in Acute Myeloid Leukemia: Novel Combinations and Therapeutic Targets , 2019, Current Oncology Reports.

[4]  F. Slack,et al.  Hypomethylating agent alters the immune microenvironment in acute myeloid leukaemia (AML) and enhances the immunogenicity of a dendritic cell/AML vaccine , 2019, British journal of haematology.

[5]  Lai Guan Ng,et al.  Dimensionality reduction for visualizing single-cell data using UMAP , 2018, Nature Biotechnology.

[6]  P. Sharma,et al.  Efficacy, Safety, and Biomarkers of Response to Azacitidine and Nivolumab in Relapsed/Refractory Acute Myeloid Leukemia: A Nonrandomized, Open-Label, Phase II Study. , 2018, Cancer discovery.

[7]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

[8]  A. Levine,et al.  A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy , 2017, Nature.

[9]  Shao-Cong Sun,et al.  NF-κB signaling in inflammation , 2017, Signal Transduction and Targeted Therapy.

[10]  D. Neuberg,et al.  Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions , 2016, Science Translational Medicine.

[11]  F. Ramos,et al.  Acute myeloid leukemia in the older adults , 2016, Leukemia research reports.

[12]  H. Zarour Reversing T-cell Dysfunction and Exhaustion in Cancer , 2016, Clinical Cancer Research.

[13]  K. Akashi,et al.  A TIM-3/Gal-9 Autocrine Stimulatory Loop Drives Self-Renewal of Human Myeloid Leukemia Stem Cells and Leukemic Progression. , 2015, Cell stem cell.

[14]  G. Freeman,et al.  Combination cancer immunotherapy and new immunomodulatory targets , 2015, Nature Reviews Drug Discovery.

[15]  T. Whiteside,et al.  Programmed death-1 checkpoint blockade in acute myeloid leukemia , 2015, Expert opinion on biological therapy.

[16]  Mikhail Shugay,et al.  MiXCR: software for comprehensive adaptive immunity profiling , 2015, Nature Methods.

[17]  A. Regev,et al.  Spatial reconstruction of single-cell gene expression data , 2015 .

[18]  M. Guindani,et al.  A role for BMP-induced homeobox gene MIXL1 in acute myelogenous leukemia and identification of type I BMP receptor as a potential target for therapy , 2014, Oncotarget.

[19]  Loise M. Francisco,et al.  RGMb is a novel binding partner for PD-L2 and its engagement with PD-L2 promotes respiratory tolerance , 2014, The Journal of experimental medicine.

[20]  R. Champlin Reduced intensity allogeneic hematopoietic transplantation is an established standard of care for treatment of older patients with acute myeloid leukemia. , 2013, Best practice & research. Clinical haematology.

[21]  Nikhil C. Munshi,et al.  Vaccination with Dendritic Cell/Tumor Fusions following Autologous Stem Cell Transplant Induces Immunologic and Clinical Responses in Multiple Myeloma Patients , 2013, Clinical Cancer Research.

[22]  C. Drake,et al.  Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. , 2012, The New England journal of medicine.

[23]  Joshua F. McMichael,et al.  Clonal evolution in relapsed acute myeloid leukemia revealed by whole genome sequencing , 2011, Nature.

[24]  D. Munn,et al.  Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. , 2011, Blood.

[25]  N. Munshi,et al.  Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma. , 2011, Blood.

[26]  H. Waldmann,et al.  mTOR signalling and metabolic regulation of T cell differentiation. , 2010, Current opinion in immunology.

[27]  Jenna M. Sullivan,et al.  Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity , 2010, The Journal of experimental medicine.

[28]  P. Rotwein,et al.  Molecular biology, genetics and biochemistry of the repulsive guidance molecule family. , 2009, The Biochemical journal.

[29]  R. Ahmed,et al.  mTOR regulates memory CD8 T cell differentiation , 2009, Nature.

[30]  S. L. Petersen Alloreactivity as therapeutic principle in the treatment of hematologic malignancies. Studies of clinical and immunologic aspects of allogeneic hematopoietic cell transplantation with nonmyeloablative conditioning. , 2007, Danish medical bulletin.

[31]  Marie-Paule Lefranc,et al.  IMGT , the international ImMunoGeneTics information system , 2003 .

[32]  J. Allison,et al.  ICOS co-stimulatory receptor is essential for T-cell activation and function , 2001, Nature.

[33]  G. Peoples,et al.  Arming the Immune System Through Vaccination to Prevent Cancer Recurrence. , 2016, American Society of Clinical Oncology educational book. American Society of Clinical Oncology. Annual Meeting.