Combination anti–CTLA-4 plus anti–PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies

Significance Immune checkpoint blockade therapy has become a critical pillar of cancer therapy. Here, we characterize the cellular mechanisms of monotherapy and combination anti–cytotoxic T lymphocyte antigen-4 plus anti–programmed cell death-1 therapy. Using high-dimensional single-cell profiling, we determine that combination therapy elicits cellular responses that are partially distinct from those induced by either monotherapy. In particular, combination therapy mediates a switch from expansion of phenotypically exhausted cluster of differentiation 8 (CD8) T cells to expansion of activated effector CD8 T cells. In addition, we systematically compare T cell subsets present in matched peripheral blood and tumor tissues to define what aspects of antitumor responses can be observed peripherally. These findings have significant implications for both the cellular mechanisms of action and biomarkers of response to monotherapies and combination therapy. Immune checkpoint blockade therapy targets T cell-negative costimulatory molecules such as cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 (PD-1). Combination anti–CTLA-4 and anti–PD-1 blockade therapy has enhanced efficacy, but it remains unclear through what mechanisms such effects are mediated. A critical question is whether combination therapy targets and modulates the same T cell populations as monotherapies. Using a mass cytometry-based systems approach, we comprehensively profiled the response of T cell populations to monotherapy and combination anti–CTLA-4 plus anti–PD-1 therapy in syngeneic murine tumors and clinical samples. Most effects of monotherapies were additive in the context of combination therapy; however, multiple combination therapy-specific effects were observed. Highly phenotypically exhausted cluster of differentiation 8 (CD8) T cells expand in frequency following anti–PD-1 monotherapy but not combination therapy, while activated terminally differentiated effector CD8 T cells expand only following combination therapy. Combination therapy also led to further increased frequency of T helper type 1 (Th1)-like CD4 effector T cells even though anti–PD-1 monotherapy is not sufficient to do so. Mass cytometry analyses of peripheral blood from melanoma patients treated with immune checkpoint blockade therapies similarly revealed mostly additive effects on the frequencies of T cell subsets along with unique modulation of terminally differentiated effector CD8 T cells by combination ipilimumab plus nivolumab therapy. Together, these findings indicate that dual blockade of CTLA-4 and PD-1 therapy is sufficient to induce unique cellular responses compared with either monotherapy.

[1]  D. Pe’er,et al.  Negative Co‐stimulation Constrains T Cell Differentiation by Imposing Boundaries on Possible Cell States , 2019, Immunity.

[2]  M. Atkins,et al.  First-line pembrolizumab (pembro) monotherapy for advanced non-clear cell renal cell carcinoma (nccRCC): Results from KEYNOTE-427 cohort B. , 2019, Journal of Clinical Oncology.

[3]  T. Powles,et al.  Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal‐Cell Carcinoma , 2019, The New England journal of medicine.

[4]  J. Madore,et al.  Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy. , 2019, Cancer cell.

[5]  Daniel E. Speiser,et al.  Intratumoral Tcf1+PD‐1+CD8+ T Cells with Stem‐like Properties Promote Tumor Control in Response to Vaccination and Checkpoint Blockade Immunotherapy , 2019, Immunity.

[6]  Maxim N. Artyomov,et al.  High-Dimensional Analysis Delineates Myeloid and Lymphoid Compartment Remodeling during Successful Immune-Checkpoint Cancer Therapy , 2018, Cell.

[7]  J. Allison,et al.  Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. , 2018, Cancer discovery.

[8]  J. Wargo,et al.  Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3+ Regulatory T Cells (Tregs) in Human Cancers , 2018, Clinical Cancer Research.

[9]  S. Loi,et al.  Dual PD-1 and CTLA-4 Checkpoint Blockade Promotes Antitumor Immune Responses through CD4+Foxp3− Cell–Mediated Modulation of CD103+ Dendritic Cells , 2018, Cancer Immunology Research.

[10]  L. Swiech,et al.  Selective FcγR Co-engagement on APCs Modulates the Activity of Therapeutic Antibodies Targeting T Cell Antigens. , 2018, Cancer cell.

[11]  Evan W. Newell,et al.  Epigenomic‐Guided Mass Cytometry Profiling Reveals Disease‐Specific Features of Exhausted CD8 T Cells , 2018, Immunity.

[12]  Angela E. Leek,et al.  Fc Effector Function Contributes to the Activity of Human Anti-CTLA-4 Antibodies , 2018, Cancer cell.

[13]  Bohuslav Melichar,et al.  Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal‐Cell Carcinoma , 2018, The New England journal of medicine.

[14]  M. Sawyer,et al.  Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[15]  Aaron M. Newman,et al.  Early B cell changes predict autoimmunity following combination immune checkpoint blockade , 2018, The Journal of clinical investigation.

[16]  B. Becher,et al.  High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy , 2018, Nature Medicine.

[17]  Peter K. Sorger,et al.  Combination Cancer Therapy Can Confer Benefit via Patient-to-Patient Variability without Drug Additivity or Synergy , 2017, Cell.

[18]  Dana Pe’er,et al.  Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade , 2017, Cell.

[19]  A. Eggermont,et al.  Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab , 2017, Annals of oncology : official journal of the European Society for Medical Oncology.

[20]  C. Leslie,et al.  Chromatin states define tumor-specific T cell dysfunction and reprogramming , 2017, Nature.

[21]  Michael B. Stadler,et al.  An Immune Atlas of Clear Cell Renal Cell Carcinoma , 2017, Cell.

[22]  Benjamin D. Greenbaum,et al.  Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses , 2017, Cell.

[23]  G. Sica,et al.  Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1–targeted therapy in lung cancer patients , 2017, Proceedings of the National Academy of Sciences.

[24]  Jedd D. Wolchok,et al.  T-cell invigoration to tumour burden ratio associated with anti-PD-1 response , 2017, Nature.

[25]  Sean C. Bendall,et al.  Systemic Immunity Is Required for Effective Cancer Immunotherapy , 2017, Cell.

[26]  R. Melchiotti,et al.  Cluster stability in the analysis of mass cytometry data , 2017, Cytometry. Part A : the journal of the International Society for Analytical Cytology.

[27]  S. Berger,et al.  Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade , 2016, Science.

[28]  Ronald D. Vale,et al.  T cell costimulatory receptor CD28 is a primary target for PD-1–mediated inhibition , 2016, Science.

[29]  Sandra P. Calderon-Copete,et al.  T Cell Factor 1-Expressing Memory-like CD8(+) T Cells Sustain the Immune Response to Chronic Viral Infections. , 2016, Immunity.

[30]  Matheus C. Bürger,et al.  Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy , 2016, Nature.

[31]  A. Iwasaki,et al.  Access of protective antiviral antibody to neuronal tissues requires CD4 T cell help , 2016, Nature.

[32]  P. Klenerman,et al.  CD39 Expression Identifies Terminally Exhausted CD8+ T Cells , 2015, PLoS pathogens.

[33]  Sean C. Bendall,et al.  Data-Driven Phenotypic Dissection of AML Reveals Progenitor-like Cells that Correlate with Prognosis , 2015, Cell.

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

[35]  C. Drake,et al.  Immune checkpoint blockade: a common denominator approach to cancer therapy. , 2015, Cancer cell.

[36]  P. Sharma,et al.  The future of immune checkpoint therapy , 2015, Science.

[37]  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.

[38]  Eli R. Zunder,et al.  Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm , 2015, Nature Protocols.

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

[40]  H. Koblish,et al.  Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8+ T cells directly within the tumor microenvironment , 2014, Journal of Immunotherapy for Cancer.

[41]  G. Dranoff,et al.  Activating Fc γ receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies , 2013, The Journal of experimental medicine.

[42]  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.

[43]  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.

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

[45]  P. LoRusso,et al.  Tremelimumab in Combination with Exemestane in Patients with Advanced Breast Cancer and Treatment-Associated Modulation of Inducible Costimulator Expression on Patient T Cells , 2010, Clinical Cancer Research.

[46]  R. Blasberg,et al.  Tumor-reactive CD4+ T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts , 2010, The Journal of experimental medicine.

[47]  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.

[48]  A. Iwasaki,et al.  CTL mobilization to virus-infected tissue requires CD4+ T cell help , 2009, Nature.

[49]  John F. Ward,et al.  Anti-CTLA-4 therapy results in higher CD4+ICOShi T cell frequency and IFN-γ levels in both nonmalignant and malignant prostate tissues , 2009, Proceedings of the National Academy of Sciences.

[50]  P. Sharma,et al.  CTLA-4 blockade increases IFNγ-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients , 2008, Proceedings of the National Academy of Sciences.

[51]  J. Allison,et al.  Combination Immunotherapy of B16 Melanoma Using Anti–Cytotoxic T Lymphocyte–Associated Antigen 4 (Ctla-4) and Granulocyte/Macrophage Colony-Stimulating Factor (Gm-Csf)-Producing Vaccines Induces Rejection of Subcutaneous and Metastatic Tumors Accompanied by Autoimmune Depigmentation , 1999, The Journal of experimental medicine.

[52]  Vanessa M. Hubbard-Lucey,et al.  Comprehensive analysis of the clinical immuno-oncology landscape , 2018, Annals of oncology : official journal of the European Society for Medical Oncology.