The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in Melanoma.

PURPOSE Blocking the interaction between the programmed cell death (PD)-1 protein and one of its ligands, PD-L1, has been reported to have impressive antitumor responses. Therapeutics targeting this pathway are currently in clinical trials. Pembrolizumab and nivolumab are the first of this anti-PD-1 pathway family of checkpoint inhibitors to gain accelerated approval from the US Food and Drug Administration (FDA) for the treatment of ipilimumab-refractory melanoma. Nivolumab has been associated with improved overall survival compared with dacarbazine in patients with previously untreated wild-type serine/threonine-protein kinase B-raf proto-oncogene BRAF melanoma. Although the most mature data are in the treatment of melanoma, the FDA has granted approval of nivolumab for squamous cell lung cancer and the breakthrough therapy designation to immune- checkpoint inhibitors for use in other cancers: nivolumab, an anti-PD-1 monoclonal antibody, for Hodgkin lymphoma, and MPDL-3280A, an anti-PD-L1 monoclonal antibody, for bladder cancer and non-small cell lung cancer. Here we review the literature on PD-1 and PD-L1 blockade and focus on the reported clinical studies that have included patients with melanoma. METHODS PubMed was searched to identify relevant clinical studies of PD-1/PD-L1-targeted therapies in melanoma. A review of data from the current trials on clinicaltrial.gov was incorporated, as well as data presented in abstracts at the 2014 annual meeting of the American Society of Clinical Oncology, given the limited number of published clinical trials on this topic. FINDINGS The anti-PD-1 and anti-PD-L1 agents have been reported to have impressive antitumor effects in several malignancies, including melanoma. The greatest clinical activity in unselected patients has been seen in melanoma. Tumor expression of PD-L1 is a suggestive, but inadequate, biomarker predictive of response to immune-checkpoint blockade. However, tumors expressing little or no PD-L1 are less likely to respond to PD-1 pathway blockade. Combination checkpoint blockade with PD-1 plus cytotoxic T-lymphocyte antigen (CTLA)-4 blockade appears to improve response rates in patients who are less likely to respond to single-checkpoint blockade. Toxicity with PD-1 blocking agents is less than the toxicity with previous immunotherapies (eg, interleukin 2, CTLA-4 blockade). Certain adverse events can be severe and potentially life threatening, but most can be prevented or reversed with close monitoring and appropriate management. IMPLICATIONS This family of immune-checkpoint inhibitors benefits not only patients with metastatic melanoma but also those with historically less responsive tumor types. Although a subset of patients responds to single-agent blockade, the initial trial of checkpoint-inhibitor combinations has reported a potential to improve response rates. Combination therapies appear to be a means of increasing response rates, albeit with increased immune-related adverse events. As these treatments become available to patients, education regarding the recognition and management of immune-related effects of immune-checkpoint blockade will be essential for maximizing clinical benefit.

[1]  J. Altman,et al.  Viral Immune Evasion Due to Persistence of Activated T Cells Without Effector Function , 1998, The Journal of experimental medicine.

[2]  F. Traganos,et al.  Activation-induced expression of human programmed death-1 gene in T-lymphocytes. , 1997, Experimental cell research.

[3]  G. Zhu,et al.  B7‐H1 (CD274) inhibits the development of herpetic stromal keratitis (HSK) , 2005, FEBS letters.

[4]  T. Honjo,et al.  PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. , 2004, International immunology.

[5]  Axel Hoos,et al.  Guidelines for the Evaluation of Immune Therapy Activity in Solid Tumors: Immune-Related Response Criteria , 2009, Clinical Cancer Research.

[6]  D. Schadendorf,et al.  Pooled Analysis of Long-Term Survival Data From Phase II and Phase III Trials of Ipilimumab in Unresectable or Metastatic Melanoma. , 2015, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[7]  T. Okazaki,et al.  Tumor cell expression of programmed cell death‐1 ligand 1 is a prognostic factor for malignant melanoma , 2010, Cancer.

[8]  CTLA-4 and PD-1 Receptors Inhibit T-Cell Activation by Distinct Mechanisms. , 2004 .

[9]  A. Korman,et al.  Anti-CTLA-4 Antibodies of IgG2a Isotype Enhance Antitumor Activity through Reduction of Intratumoral Regulatory T Cells , 2013, Cancer Immunology Research.

[10]  P. Linsley,et al.  CD80 (B7-1) Binds Both CD28 and CTLA-4 with a Low Affinity and Very Fast Kinetics , 1997, The Journal of experimental medicine.

[11]  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.

[12]  D. Schadendorf,et al.  Nivolumab in previously untreated melanoma without BRAF mutation. , 2015, The New England journal of medicine.

[13]  Drew M Pardoll,et al.  Differential binding properties of B7-H1 and B7-DC to programmed death-1. , 2003, Biochemical and biophysical research communications.

[14]  G. Freeman,et al.  Endothelial expression of PD‐L1 and PD‐L2 down‐regulates CD8+ T cell activation and cytolysis , 2003, European journal of immunology.

[15]  R. Sullivan,et al.  Characterizing the Clinical Benefit of Ipilimumab in Patients Who Progressed on High-dose IL-2 , 2012, Journal of immunotherapy.

[16]  Philip Smith,et al.  Schistosoma mansoni Worms Induce Anergy of T Cells via Selective Up-Regulation of Programmed Death Ligand 1 on Macrophages1 , 2004, The Journal of Immunology.

[17]  C. Mu,et al.  High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation , 2011, Medical oncology.

[18]  L. Gordon,et al.  Disabling immune tolerance by programmed death-1 blockade with pidilizumab after autologous hematopoietic stem-cell transplantation for diffuse large B-cell lymphoma: results of an international phase II trial. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[19]  T. Honjo,et al.  Induced expression of PD‐1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. , 1992, The EMBO journal.

[20]  C. Horak,et al.  Peripheral and tumor immune correlates in patients with advanced melanoma treated with combination nivolumab (anti-PD-1, BMS-936558, ONO-4538) and ipilimumab. , 2013 .

[21]  J. Lunceford,et al.  Abstract CT105: MK-3475 (anti-PD-1 monoclonal antibody) for non-small cell lung cancer (NSCLC): Antitumor activity and association with tumor PD-L1 expression , 2014 .

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

[23]  J. Cheville,et al.  618: Costimulatory B7-H1 In Renal Cell Carcinoma Patients: Indicator of Tumor Aggressiveness and Potential Therapeutic Target , 2005 .

[24]  J. Lunceford,et al.  Abstract CT104: Antitumor activity of the anti-PD-1 monoclonal antibody MK-3475 in melanoma(MEL): Correlation of tumor PD-L1 expression with outcome , 2014 .

[25]  J. Lunceford,et al.  Clinical efficacy and correlation with tumor PD-L1 expression in patients (pts) with melanoma (MEL) treated with the anti-PD-1 monoclonal antibody MK-3475. , 2014 .

[26]  Alison P. Klein,et al.  Colocalization of Inflammatory Response with B7-H1 Expression in Human Melanocytic Lesions Supports an Adaptive Resistance Mechanism of Immune Escape , 2012, Science Translational Medicine.

[27]  A. Ribas,et al.  The evolution of checkpoint blockade as a cancer therapy: what's here, what's next? , 2015, Current opinion in immunology.

[28]  H. V. van Boven,et al.  Overall survival and PD‐L1 expression in metastasized malignant melanoma , 2011, Cancer.

[29]  T. Honjo,et al.  Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. , 1999, Immunity.

[30]  J. Sosman,et al.  Biomarkers and associations with the clinical activity of PD-L1 blockade in a MPDL3280A study. , 2013 .

[31]  R. Berger,et al.  Phase I Safety and Pharmacokinetic Study of CT-011, a Humanized Antibody Interacting with PD-1, in Patients with Advanced Hematologic Malignancies , 2008, Clinical Cancer Research.

[32]  G. Freeman,et al.  Structures of PD-1 with its ligands: Sideways and dancing cheek to cheek , 2008, Proceedings of the National Academy of Sciences.

[33]  G. Freeman,et al.  Blockade of Programmed Death-1 Ligands on Dendritic Cells Enhances T Cell Activation and Cytokine Production 1 , 2003, The Journal of Immunology.

[34]  C. Drake,et al.  Molecular Pathways Molecular Pathways : Coexpression of Immune Checkpoint Molecules : Signaling Pathways and Implications for Cancer Immunotherapy , 2013 .

[35]  G. Freeman,et al.  The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection , 2007, Nature Immunology.

[36]  J. Cheville,et al.  Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[37]  R. Davis,et al.  Safety and activity of PD1 blockade by pidilizumab in combination with rituximab in patients with relapsed follicular lymphoma: a single group, open-label, phase 2 trial. , 2014, The Lancet. Oncology.

[38]  G. Freeman,et al.  PD-1 and its ligands in tolerance and immunity. , 2008, Annual review of immunology.

[39]  Yoshimasa Tanaka,et al.  Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[40]  Luc J. Smink,et al.  Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease , 2003, Nature.

[41]  A. Korman,et al.  Activity of Anti-PD-1 in Murine Tumor Models: Role of “Host” PD-L1 and Synergistic Effect of Anti-PD-1 and Anti-CTLA-4 (48.37) , 2007, The Journal of Immunology.

[42]  M. Dolled-Filhart,et al.  A phase Ib study of MK-3475 in patients with human papillomavirus (HPV)-associated and non-HPV–associated head and neck (H/N) cancer. , 2014 .

[43]  G. Freeman,et al.  PD‐L1 and PD‐L2 have distinct roles in regulating host immunity to cutaneous leishmaniasis , 2006, European journal of immunology.

[44]  J. Wolchok,et al.  Efficacy and safety of the anti-PD-1 monoclonal antibody MK-3475 in 411 patients (pts) with melanoma (MEL). , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[45]  G. Freeman,et al.  Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. , 2007, Immunity.

[46]  G. Freeman,et al.  A new therapeutic strategy for malaria: targeting T cell exhaustion , 2012, Nature Immunology.

[47]  J. Bluestone,et al.  Lymphoproliferative disorder in CTLA-4 knockout mice is characterized by CD28-regulated activation of Th2 responses. , 1999, Journal of immunology.

[48]  G. Freeman,et al.  The B7 family revisited. , 2005, Annual review of immunology.

[49]  G. Freeman,et al.  PD-L2 is a second ligand for PD-1 and inhibits T cell activation , 2001, Nature Immunology.

[50]  T. Okazaki,et al.  Establishment of NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[51]  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.

[52]  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.

[53]  M. Atkins,et al.  Prognostic and predictive markers for the new immunotherapies. , 2014, Oncology.

[54]  Philip J. R. Goulder,et al.  PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression , 2006, Nature.

[55]  P. Hegde,et al.  MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer , 2014, Nature.

[56]  Shohei Koyama,et al.  Loss of Lkb1 and Pten leads to lung squamous cell carcinoma with elevated PD-L1 expression. , 2014, Cancer cell.

[57]  S. Rosenberg,et al.  Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. , 1994, JAMA.

[58]  G. Freeman,et al.  Engagement of the Pd-1 Immunoinhibitory Receptor by a Novel B7 Family Member Leads to Negative Regulation of Lymphocyte Activation , 2000, The Journal of experimental medicine.

[59]  J. Fay,et al.  Phase 2, multicenter, safety and efficacy study of pidilizumab in patients with metastatic melanoma. , 2014 .

[60]  V. Kuchroo,et al.  Studies in B7-Deficient Mice Reveal a Critical Role for B7 Costimulation in Both Induction and Effector Phases of Experimental Autoimmune Encephalomyelitis , 1999, The Journal of experimental medicine.

[61]  Ronald N Germain,et al.  Tuning of antigen sensitivity by T cell receptor-dependent negative feedback controls T cell effector function in inflamed tissues. , 2014, Immunity.

[62]  Tasuku Honjo,et al.  Anti-Programmed Cell Death 1 Antibody Reduces CD4+PD-1+ T Cells and Relieves the Lupus-Like Nephritis of NZB/W F1 Mice , 2010, The Journal of Immunology.

[63]  J. Wolchok,et al.  Hepatotoxicity with combination of vemurafenib and ipilimumab. , 2013, The New England journal of medicine.

[64]  D. Olive,et al.  The butyrophilin (BTN) gene family: from milk fat to the regulation of the immune response , 2012, Immunogenetics.

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

[66]  J. Allison Immune Checkpoint Blockade in Cancer Therapy , 2008 .

[67]  J. Cheville,et al.  Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. , 2006, Cancer research.

[68]  Israel Lowy,et al.  Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[69]  Yoshimasa Tanaka,et al.  Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice , 2003, Nature Medicine.

[70]  S. Husby,et al.  Alternative splice variants of the human PD-1 gene. , 2005, Cellular immunology.

[71]  Gareth Griffiths,et al.  Interferon-alpha and survival in metastatic renal carcinoma: early results of a randomised controlled trial. Medical Research Council Renal Cancer Collaborators. , 1999, Lancet.

[72]  R. Sullivan,et al.  Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM). , 2013 .

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

[74]  Clive R. Taylor,et al.  Association of tumor PD-L1 expression and immune biomarkers with clinical activity in patients (pts) with advanced solid tumors treated with nivolumab (anti-PD-1; BMS-936558; ONO-4538). , 2013 .

[75]  Antoni Ribas,et al.  Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. , 2013, The New England journal of medicine.

[76]  T. Okazaki,et al.  PD-1 Inhibits Antiviral Immunity at the Effector Phase in the Liver , 2003, The Journal of experimental medicine.

[77]  Michael Loran Dustin,et al.  PD-1 promotes immune exhaustion by inducing antiviral T cell motility paralysis , 2013, The Journal of experimental medicine.

[78]  R. Lufkin,et al.  Durable responses and reversible toxicity of high-dose interleukin-2 treatment of melanoma and renal cancer in a Community Hospital Biotherapy Program , 2014, Journal of Immunotherapy for Cancer.

[79]  J. Taube,et al.  Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy , 2014, Clinical Cancer Research.

[80]  H. Nie,et al.  Aberrant Regulation of Synovial T Cell Activation by Soluble Costimulatory Molecules in Rheumatoid Arthritis1 , 2006, The Journal of Immunology.

[81]  J. Allison,et al.  PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors , 2010, Proceedings of the National Academy of Sciences.

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

[83]  G. Freeman,et al.  PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation , 2015, Nature Communications.

[84]  H. Kohrt,et al.  Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients , 2014, Nature.

[85]  R. Arceci Intratumor Heterogeneity and Branched Evolution Revealed by Multiregion Sequencing , 2012 .

[86]  J. Wolchok,et al.  Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti–CTLA-4 therapy against melanoma , 2013, The Journal of experimental medicine.

[87]  Michael R. Green,et al.  Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. , 2010, Blood.

[88]  C. Thompson,et al.  Regulation of surface and intracellular expression of CTLA4 on mouse T cells. , 1996, Journal of immunology.