Immunological effects of the anti-programmed death-1 antibody on human peripheral blood mononuclear cells.

Immune checkpoint antibody-mediated blockade has gained attention as a new cancer immunotherapy strategy. Accumulating evidence suggests that this therapy imparts a survival benefit to metastatic melanoma and non-small cell lung cancer patients. A substantial amount of data on immune checkpoint antibodies has been collected from clinical trials; however, the direct effect of the antibodies on human peripheral blood mononuclear cells (PBMCs) has not been exclusively investigated. In this study, we developed an anti-programmed death-1 (PD-1) antibody (with biosimilarity to nivolumab) and examined the effects of the antibody on PBMCs derived from cancer patients. Specifically, we investigated the effects of the anti-PD-1 antibody on proliferation, cytokine production, cytotoxic T lymphocytes (CTL) and regulatory T cells. These investigations yielded several important results. First, the anti-PD-1 antibody had no obvious effect on resting PBMCs; however, high levels of the anti-PD-1 antibody partly stimulated PBMC proliferation when accompanied by an anti-CD3 antibody. Second, the anti-PD-1 antibody restored the growth inhibition of anti-CD3 Ab-stimulated PBMCs mediated by PD-L1. Third, the anti-PD-1 antibody exhibited a moderate inhibitory effect on the induction of myeloid-derived suppressor cells (MDSCs) by anti-CD3 antibody stimulation. Additionally, the presence of the anti-PD-1 antibody promoted antigen-specific CTL induction, which suggests that combining anti-PD-1 antibody and conventional immunotherapy treatments may have beneficial effects. These results indicate that specific cellular immunological mechanisms are partly responsible for the antitumor effect exhibited by the anti-PD-1 antibody against advanced cancers in clinical trials.

[1]  L. Crinò,et al.  Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. , 2015, The New England journal of medicine.

[2]  Dirk Schadendorf,et al.  Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. , 2015, The New England journal of medicine.

[3]  G. Linette,et al.  Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. , 2015, The Lancet. Oncology.

[4]  G. Freeman,et al.  The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in Melanoma. , 2015, Clinical therapeutics.

[5]  F. Cappuzzo,et al.  Activity and safety of nivolumab, an anti-PD-1 immune checkpoint inhibitor, for patients with advanced, refractory squamous non-small-cell lung cancer (CheckMate 063): a phase 2, single-arm trial. , 2015, The Lancet. Oncology.

[6]  J. Wolchok,et al.  Combination Therapy with Anti–CTLA-4 and Anti–PD-1 Leads to Distinct Immunologic Changes In Vivo , 2015, The Journal of Immunology.

[7]  Zachary B. Abrams,et al.  Patients with pancreatic adenocarcinoma exhibit elevated levels of myeloid-derived suppressor cells upon progression of disease , 2015, Cancer Immunology, Immunotherapy.

[8]  Jing-wei Jiang,et al.  Phenotypes, accumulation, and functions of myeloid-derived suppressor cells and associated treatment strategies in cancer patients. , 2014, Human immunology.

[9]  A. Korman,et al.  In Vitro Characterization of the Anti-PD-1 Antibody Nivolumab, BMS-936558, and In Vivo Toxicology in Non-Human Primates , 2014, Cancer Immunology Research.

[10]  T. Ashizawa,et al.  Anti-vascular endothelial growth factor receptor (VEGFR) 2 autoantibody identification in glioblastoma patient using single B cell-based antibody gene cloning. , 2014, Immunology letters.

[11]  F. Fulciniti,et al.  Immunological and biological changes during ipilimumab treatment and their potential correlation with clinical response and survival in patients with advanced melanoma , 2014, Cancer Immunology, Immunotherapy.

[12]  David C. Smith,et al.  Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[13]  T. Okazaki,et al.  A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application , 2013, Nature Immunology.

[14]  C. Horak,et al.  Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[15]  M. Smyth,et al.  Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors By Gene-Modified T Cells , 2013, Clinical Cancer Research.

[16]  C. Horak,et al.  Nivolumab plus ipilimumab in advanced melanoma. , 2013, The New England journal of medicine.

[17]  M. Sznol,et al.  Antagonist Antibodies to PD-1 and B7-H1 (PD-L1) in the Treatment of Advanced Human Cancer , 2013, Clinical Cancer Research.

[18]  Paolo A Ascierto,et al.  Biomarkers for Immunostimulatory Monoclonal Antibodies in Combination Strategies for Melanoma and Other Tumor Types , 2013, Clinical Cancer Research.

[19]  P. Ascierto,et al.  Clinical Development of Immunostimulatory Monoclonal Antibodies and Opportunities for Combination , 2013, Clinical Cancer Research.

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

[21]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[22]  T. Ashizawa,et al.  Identification of novel MAGE-A6- and MAGE-A12-derived HLA-A24-restricted cytotoxic T lymphocyte epitopes using an in silico peptide-docking assay , 2012, Cancer Immunology, Immunotherapy.

[23]  T. Ashizawa,et al.  Analysis of HLA‐A24‐restricted peptides of carcinoembryonic antigen using a novel structure‐based peptide‐HLA docking algorithm , 2011, Cancer science.

[24]  J. Allison,et al.  Single dose of anti–CTLA-4 enhances CD8+ T-cell memory formation, function, and maintenance , 2010, Proceedings of the National Academy of Sciences.

[25]  J. Wolchok,et al.  Anti-CTLA-4 antibody therapy: immune monitoring during clinical development of a novel immunotherapy. , 2010, Seminars in oncology.

[26]  P. Ascierto,et al.  Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. , 2010, Seminars in oncology.

[27]  D. Schadendorf,et al.  Improved survival with ipilimumab in patients with metastatic melanoma. , 2010, The New England journal of medicine.

[28]  A. Korman,et al.  Enhanced Tumor Eradication by Combining CTLA-4 or PD-1 Blockade With CpG Therapy , 2010, Journal of immunotherapy.

[29]  F. Marincola,et al.  Dendritic Cell Vaccination Combined with CTLA4 Blockade in Patients with Metastatic Melanoma , 2009, Clinical Cancer Research.

[30]  S. Senju,et al.  An in vivo model of priming of antigen-specific human CTL by Mo-DC in NOD/Shi-scid IL2rgamma(null) (NOG) mice. , 2009, Immunology letters.

[31]  Daohai Yu,et al.  PD1 blockade reverses the suppression of melanoma antigen-specific CTL by CD4+ CD25(Hi) regulatory T cells. , 2009, International immunology.

[32]  Lisa M. Ebert,et al.  Melan-A–specific Cytotoxic T Cells Are Associated with Tumor Regression and Autoimmunity Following Treatment with Anti-CTLA-4 , 2009, Clinical Cancer Research.

[33]  S. O’Day,et al.  Phase I/II study of ipilimumab for patients with metastatic melanoma. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[34]  A. Korman,et al.  Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs. , 2007, International immunology.

[35]  K. Flaherty,et al.  Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.