Agonistic CD40 mAb-Driven IL12 Reverses Resistance to Anti-PD1 in a T-cell-Rich Tumor.

The durability and efficacy of anti-human PD1 monoclonal antibodies (PD1 mAb) vary across different malignancies. Although an absence of tumor-infiltrating cytotoxic T lymphocytes has been identified as a cause for resistance to PD1 mAb, the presence of intratumor exhausted PD1hi T cells also contributes to insensitivity to this immune checkpoint therapy. In this study, we used mouse tumor models of PD1 mAb resistance that harbored PD1hi T cells and flow cytometry analysis of tumor-infiltrating leukocytes immediately post-therapy as a screening platform to identify agents that could resensitize T cells to PD1 blockade. We showed that an agonistic anti-CD40 mAb converted PD1hi T cells into PD1lo T cells, reversing phenotypic T-cell exhaustion and allowing the anti-PD1 refractory tumors to respond to anti-PD1 therapy. PD1 downmodulation by anti-CD40 mAb relied upon IL12 but not IL23, CD80/CD86/CD28, or CD70/CD27. Consistent with a role for regulatory T cells (Treg) in promoting T-cell exhaustion, we also showed that intratumor Treg presented with a less activated and attenuated suppressive phenotype, marked by reductions in CTLA4 and PD1. Similar to anti-CD40 mAb, anti-CTLA4 mAb also lowered intratumor T-cell PD1 expression. Our study provides a proof-of-principle framework to systematically identify immune conditioning agents able to convert PD1hi T cells to PD1lo T cells, with clinical implications in the management of anti-PD1 refractory patients. Cancer Res; 76(21); 6266-77. ©2016 AACR.

[1]  Jedd D. Wolchok,et al.  The future of cancer treatment: immunomodulation, CARs and combination immunotherapy , 2016, Nature Reviews Clinical Oncology.

[2]  Drew M. Pardoll,et al.  PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. , 2016, The New England journal of medicine.

[3]  Nicolai J. Birkbak,et al.  Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade , 2016, Science.

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

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

[6]  Jens Schreiner,et al.  Progression of Lung Cancer Is Associated with Increased Dysfunction of T Cells Defined by Coexpression of Multiple Inhibitory Receptors , 2015, Cancer Immunology Research.

[7]  E. Wherry,et al.  Molecular and cellular insights into T cell exhaustion , 2015, Nature Reviews Immunology.

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

[9]  Bert Vogelstein,et al.  PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. , 2015, The New England journal of medicine.

[10]  T. Aparicio,et al.  PD-1 blockade in tumors with mismatch-repair deficiency , 2015 .

[11]  Lieping Chen,et al.  PD-1 Upregulated on Regulatory T Cells during Chronic Virus Infection Enhances the Suppression of CD8+ T Cell Immune Response via the Interaction with PD-L1 Expressed on CD8+ T Cells , 2015, The Journal of Immunology.

[12]  Antoni Ribas,et al.  Classifying Cancers Based on T-cell Infiltration and PD-L1. , 2015, Cancer research.

[13]  C. Meyer,et al.  Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients , 2015, Proceedings of the National Academy of Sciences.

[14]  P. Sharma,et al.  Immune Checkpoint Targeting in Cancer Therapy: Toward Combination Strategies with Curative Potential , 2015, Cell.

[15]  Martin L. Miller,et al.  Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer , 2015, Science.

[16]  E. Wherry,et al.  Overcoming T cell exhaustion in infection and cancer. , 2015, Trends in immunology.

[17]  P. Herzig,et al.  Induced PD-L1 Expression Mediates Acquired Resistance to Agonistic Anti-CD40 Treatment , 2015, Cancer Immunology Research.

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

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

[20]  J. Wolchok,et al.  Anticancer immunotherapy by CTLA-4 blockade: obligatory contribution of IL-2 receptors and negative prognostic impact of soluble CD25 , 2015, Cell Research.

[21]  Maxim N. Artyomov,et al.  Checkpoint Blockade Cancer Immunotherapy Targets Tumour-Specific Mutant Antigens , 2014, Nature.

[22]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[23]  A. Rudensky,et al.  Interplay between regulatory T cells and PD-1 in modulating T cell exhaustion and viral control during chronic LCMV infection , 2014, The Journal of experimental medicine.

[24]  M. Smyth,et al.  A balance of interleukin-12 and -23 in cancer. , 2013, Trends in immunology.

[25]  Michael Y. Gerner,et al.  Cutting Edge: IL-12 and Type I IFN Differentially Program CD8 T Cells for Programmed Death 1 Re-expression Levels and Tumor Control , 2013, The Journal of Immunology.

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

[27]  G. Freeman,et al.  PD-L1 blockade synergizes with IL-2 therapy in reinvigorating exhausted T cells. , 2013, The Journal of clinical investigation.

[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]  Jenna M. Sullivan,et al.  TIM3+FOXP3+ regulatory T cells are tissue-specific promoters of T-cell dysfunction in cancer , 2013, Oncoimmunology.

[30]  W. Rosenberg,et al.  The Third Signal Cytokine IL-12 Rescues the Anti-Viral Function of Exhausted HBV-Specific CD8 T Cells , 2013, PLoS pathogens.

[31]  Burton E. Barnett,et al.  Progenitor and Terminal Subsets of CD8+ T Cells Cooperate to Contain Chronic Viral Infection , 2012, Science.

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

[33]  A. Rudensky,et al.  Regulatory T cells: mechanisms of differentiation and function. , 2012, Annual review of immunology.

[34]  Drew M. Pardoll,et al.  The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.

[35]  M. Smyth,et al.  Biology and clinical observations of regulatory T cells in cancer immunology. , 2010, Current topics in microbiology and immunology.

[36]  M. Willson,et al.  CD40 Agonists Alter Tumor Stroma and Show Efficacy Against Pancreatic Carcinoma in Mice and Humans , 2011 .

[37]  M. Smyth,et al.  Microenvironment and Immunology Anti-TIM 3 Antibody Promotes T Cell IFN-g – Mediated Antitumor Immunity and Suppresses Established Tumors , 2011 .

[38]  M. Glennie,et al.  Control of Established Melanoma by CD27 Stimulation Is Associated With Enhanced Effector Function and Persistence, and Reduced PD-1 Expression of Tumor Infiltrating CD8+ T Cells , 2010, Journal of immunotherapy.

[39]  J. Curtsinger,et al.  Inflammatory cytokines as a third signal for T cell activation. , 2010, Current opinion in immunology.

[40]  A. Möller,et al.  IL-23 suppresses innate immune response independently of IL-17A during carcinogenesis and metastasis , 2010, Proceedings of the National Academy of Sciences.

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

[42]  R. Noelle,et al.  Molecular mechanism and function of CD40/CD40L engagement in the immune system , 2009, Immunological reviews.

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

[44]  W. Leonard,et al.  Development of regulatory T cells requires IL-7Ralpha stimulation by IL-7 or TSLP. , 2008, Blood.

[45]  G. Freeman,et al.  Selective expansion of a subset of exhausted CD8 T cells by αPD-L1 blockade , 2008, Proceedings of the National Academy of Sciences.

[46]  E. Wherry,et al.  Cutting Edge: IL-12 Inversely Regulates T-bet and Eomesodermin Expression during Pathogen-Induced CD8+ T Cell Differentiation1 , 2006, The Journal of Immunology.

[47]  H. Yagita,et al.  Induction of CD70 on Dendritic Cells through CD40 or TLR Stimulation Contributes to the Development of CD8+ T Cell Responses in the Absence of CD4+ T Cells1 , 2005, The Journal of Immunology.

[48]  H. Schneider,et al.  Unifying concepts in CD28, ICOS and CTLA4 co-receptor signalling , 2003, Nature Reviews Immunology.

[49]  P. Blair,et al.  ICOS Costimulation Requires IL-2 and Can Be Prevented by CTLA-4 Engagement1 , 2001, The Journal of Immunology.

[50]  F. Belardelli,et al.  IFN‐α and IL‐18 exert opposite regulatory effects on the IL‐12 receptor expression and IL‐12‐induced IFN‐γ production in mouse macrophages: novel pathways in the regulation of the inflammatory response of macrophages , 2000, Journal of leukocyte biology.

[51]  A. Lanzavecchia,et al.  Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation , 1996, The Journal of experimental medicine.