RANKL blockade improves efficacy of PD1-PD-L1 blockade or dual PD1-PD-L1 and CTLA4 blockade in mouse models of cancer

ABSTRACT Receptor activator of NF-κB ligand (RANKL) and its receptor RANK, are members of the tumor necrosis factor and receptor superfamilies, respectively. Antibodies targeting RANKL have recently been evaluated in combination with anti-CTLA4 in case reports of human melanoma and mouse models of cancer. However, the efficacy of anti-RANKL in combination with antibodies targeting other immune checkpoint receptors such as PD1 has not been reported. In this study, we demonstrated that blockade of RANKL improves anti-metastatic activity of antibodies targeting PD1/PD-L1 and improves subcutaneous growth suppression in mouse models of melanoma, prostate and colon cancer. Suppression of experimental lung metastasis following combination anti-RANKL with anti-PD1 requires NK cells and IFN-γ, whereas subcutaneous tumor growth suppression with this combination therapy is attenuated in the absence of T cells and IFN-γ. Furthermore, addition of anti-RANKL to anti-PD1 and anti-CTLA4 resulted in superior anti-tumor responses, irrespective of the ability of anti-CTLA4 isotype to engage activating FcR, and concurrent or delayed RANKL blockade was most effective. Early-during-treatment assessment reveals this triple combination therapy compared to dual anti-PD1 and anti-CTLA4 combination therapy further increased the proportion of tumor-infiltrating CD4+ and CD8+ T cells that can produce both IFN-γ and TNF. Finally, RANKL expression appears to identify tumor-specific CD8+ T cells expressing higher levels of PD1 which can be modulated by anti-PD1. These data set the scene for clinical evaluation of denosumab use in patients receiving contemporary immune checkpoint blockade.

[1]  N. Waddell,et al.  Interleukin-12 from CD103+ Batf3-Dependent Dendritic Cells Required for NK-Cell Suppression of Metastasis , 2017, Cancer Immunology Research.

[2]  D. Schadendorf,et al.  Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma , 2017, The New England journal of medicine.

[3]  B. Fox,et al.  Timing of PD-1 Blockade Is Critical to Effective Combination Immunotherapy with Anti-OX40 , 2017, Clinical Cancer Research.

[4]  J. Wilmott,et al.  Negative immune checkpoint regulation by VISTA: a mechanism of acquired resistance to anti-PD-1 therapy in metastatic melanoma patients , 2017, Modern Pathology.

[5]  Deborah S. Barkauskas,et al.  Co-administration of RANKL and CTLA4 Antibodies Enhances Lymphocyte-Mediated Antitumor Immunity in Mice , 2017, Clinical Cancer Research.

[6]  J. Madore,et al.  Dynamic Changes in PD-L1 Expression and Immune Infiltrates Early During Treatment Predict Response to PD-1 Blockade in Melanoma , 2017, Clinical Cancer Research.

[7]  Y. Shentu,et al.  Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. , 2016, The New England journal of medicine.

[8]  N. Munshi,et al.  Osteoclasts promote immune suppressive microenvironment in multiple myeloma: therapeutic implication. , 2016, Blood.

[9]  Deborah S. Barkauskas,et al.  Co-inhibition of CD73 and A2AR Adenosine Signaling Improves Anti-tumor Immune Responses. , 2016, Cancer cell.

[10]  A. Korman,et al.  Preclinical Development of Ipilimumab and Nivolumab Combination Immunotherapy: Mouse Tumor Models, In Vitro Functional Studies, and Cynomolgus Macaque Toxicology , 2016, PloS one.

[11]  L. Chin,et al.  Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. , 2016, Cancer discovery.

[12]  J. Lunceford,et al.  Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): an open-label, multicentre, phase 1b trial. , 2016, The Lancet. Oncology.

[13]  A. Ribas,et al.  Combination cancer immunotherapies tailored to the tumour microenvironment , 2016, Nature Reviews Clinical Oncology.

[14]  John J Miles,et al.  Suppression of Metastases Using a New Lymphocyte Checkpoint Target for Cancer Immunotherapy. , 2016, Cancer discovery.

[15]  A. Ravaud,et al.  Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. , 2015, The New England journal of medicine.

[16]  M. Valsecchi Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. , 2015, The New England journal of medicine.

[17]  L. Zitvogel,et al.  A Threshold Level of Intratumor CD8+ T-cell PD1 Expression Dictates Therapeutic Response to Anti-PD1. , 2015, Cancer research.

[18]  J. Larkin,et al.  Pembrolizumab versus Ipilimumab in Advanced Melanoma. , 2015, The New England journal of medicine.

[19]  A. Salama,et al.  Rapid complete response of metastatic melanoma in a patient undergoing ipilimumab immunotherapy in the setting of active ulcerative colitis , 2015, Journal of Immunotherapy for Cancer.

[20]  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.

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

[22]  M. Millenson,et al.  PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. , 2015, The New England journal of medicine.

[23]  J. Hackney,et al.  The immunoreceptor TIGIT regulates antitumor and antiviral CD8(+) T cell effector function. , 2014, Cancer cell.

[24]  Seema A. Khan,et al.  RANKL expression in normal and malignant breast tissue responds to progesterone and is up-regulated during the luteal phase , 2014, Breast Cancer Research and Treatment.

[25]  S. Quezada,et al.  Impact of tumour microenvironment and Fc receptors on the activity of immunomodulatory antibodies. , 2014, Trends in immunology.

[26]  I. Holen,et al.  Targeting RANKL in metastasis , 2014, BoneKEy reports.

[27]  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.

[28]  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.

[29]  G. Scagliotti,et al.  Overall Survival Improvement in Patients with Lung Cancer and Bone Metastases Treated with Denosumab Versus Zoledronic Acid: Subgroup Analysis from a Randomized Phase 3 Study , 2012, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[30]  P. Kostenuik,et al.  Bench to bedside: elucidation of the OPG–RANK–RANKL pathway and the development of denosumab , 2012, Nature Reviews Drug Discovery.

[31]  F. Saad,et al.  Denosumab and bone-metastasis-free survival in men with castration-resistant prostate cancer: results of a phase 3, randomised, placebo-controlled trial , 2012, The Lancet.

[32]  E. Wherry T cell exhaustion , 2011, Nature Immunology.

[33]  G. Scagliotti,et al.  Randomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myeloma. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[34]  Y. Fujiwara,et al.  Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[35]  S. Quezada,et al.  Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti–CTLA-4 antibodies , 2009, The Journal of experimental medicine.

[36]  X. Mariette,et al.  Randomized phase II trial of denosumab in patients with bone metastases from prostate cancer, breast cancer, or other neoplasms after intravenous bisphosphonates. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[37]  P. Gascón,et al.  Randomized active-controlled phase II study of denosumab efficacy and safety in patients with breast cancer-related bone metastases. , 2007, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[38]  K. Aoki,et al.  Amelioration of bone loss in collagen-induced arthritis by neutralizing anti-RANKL monoclonal antibody. , 2006, Biochemical and biophysical research communications.

[39]  S. Anderton,et al.  Kinetics of costimulatory molecule expression by T cells and dendritic cells during the induction of tolerance versus immunity in vivo , 2005, European journal of immunology.

[40]  Y. Kong,et al.  Role of RANKL and RANK in bone loss and arthritis , 2002, Annals of the rheumatic diseases.

[41]  Yufang Shi,et al.  Regulation of activation‐induced receptor activator of NF‐κB ligand (RANKL) expression in T cells , 2002, European journal of immunology.

[42]  B. Foster,et al.  The TRAMP Mouse as a Model for Prostate Cancer , 2001, Current protocols in immunology.

[43]  N. Schreiber-Agus,et al.  Mouse models of prostate cancer , 1999, Oncogene.

[44]  Brian R. Wong,et al.  TRANCE Is a Novel Ligand of the Tumor Necrosis Factor Receptor Family That Activates c-Jun N-terminal Kinase in T Cells* , 1997, The Journal of Biological Chemistry.

[45]  B. Foster,et al.  Characterization of prostatic epithelial cell lines derived from transgenic adenocarcinoma of the mouse prostate (TRAMP) model. , 1997, Cancer research.

[46]  W. Qian,et al.  Progression to androgen insensitivity in a novelin vitro mouse model for prostate cancer , 1995, The Journal of Steroid Biochemistry and Molecular Biology.

[47]  R. Matusik,et al.  Prostate cancer in a transgenic mouse. , 1995, Proceedings of the National Academy of Sciences of the United States of America.

[48]  R. Scolyer,et al.  Resistance to PD1/PDL1 checkpoint inhibition. , 2017, Cancer treatment reviews.

[49]  S. Turley,et al.  Stromal infrastructure of the lymph node and coordination of immunity. , 2015, Trends in immunology.

[50]  Antonio Polley,et al.  Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection , 2009, Nature Immunology.