Tumor immune profiling predicts response to anti-PD-1 therapy in human melanoma.

BACKGROUND Immune checkpoint blockade is revolutionizing therapy for advanced cancer, but many patients do not respond to treatment. The identification of robust biomarkers that predict clinical response to specific checkpoint inhibitors is critical in order to stratify patients and to rationally select combinations in the context of an expanding array of therapeutic options. METHODS We performed multiparameter flow cytometry on freshly isolated metastatic melanoma samples from 2 cohorts of 20 patients each prior to treatment and correlated the subsequent clinical response with the tumor immune phenotype. RESULTS Increasing fractions of programmed cell death 1 high/cytotoxic T lymphocyte-associated protein 4 high (PD-1hiCTLA-4hi) cells within the tumor-infiltrating CD8+ T cell subset strongly correlated with response to therapy (RR) and progression-free survival (PFS). Functional analysis of these cells revealed a partially exhausted T cell phenotype. Assessment of metastatic lesions during anti-PD-1 therapy demonstrated a release of T cell exhaustion, as measured by an accumulation of highly activated CD8+ T cells within tumors, with no effect on Tregs. CONCLUSIONS Our data suggest that the relative abundance of partially exhausted tumor-infiltrating CD8+ T cells predicts response to anti-PD-1 therapy. This information can be used to appropriately select patients with a high likelihood of achieving a clinical response to PD-1 pathway inhibition. FUNDING This work was funded by a generous gift provided by Inga-Lill and David Amoroso as well as a generous gift provided by Stephen Juelsgaard and Lori Cook.

[1]  Dana Pe'er,et al.  PD-1 Blockade Expands Intratumoral Memory T Cells , 2016, Cancer Immunology Research.

[2]  G. Freeman,et al.  Differential Expression of PD-L1 between Primary and Metastatic Sites in Clear-Cell Renal Cell Carcinoma , 2015, Cancer Immunology Research.

[3]  J. Madore,et al.  PD‐L1 expression in melanoma shows marked heterogeneity within and between patients: implications for anti‐PD‐1/PD‐L1 clinical trials , 2015, Pigment cell & melanoma research.

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

[5]  H. An,et al.  Clinicopathological and prognostic significance of programmed cell death ligand1 (PD-L1) expression in patients with non-small cell lung cancer: a meta-analysis. , 2015, Journal of thoracic disease.

[6]  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.

[7]  Razelle Kurzrock,et al.  PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy , 2015, Molecular Cancer Therapeutics.

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

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

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

[11]  D. Douek,et al.  PD-1 identifies the patient-specific CD8⁺ tumor-reactive repertoire infiltrating human tumors. , 2014, The Journal of clinical investigation.

[12]  S. Arron,et al.  Memory regulatory T cells reside in human skin. , 2014, The Journal of clinical investigation.

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

[14]  E John Wherry,et al.  T cell exhaustion , 2011 .

[15]  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.

[16]  S. Rosenberg,et al.  Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. , 2009, Blood.

[17]  J. Altman,et al.  Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. , 2008, Immunity.

[18]  G. Freeman,et al.  Reinvigorating exhausted HIV-specific T cells via PD-1–PD-1 ligand blockade , 2006, The Journal of experimental medicine.

[19]  L. Austin,et al.  CD69, HLA-DR and the IL-2R identify persistently activated T cells in psoriasis vulgaris lesional skin: blood and skin comparisons by flow cytometry. , 2000, Journal of autoimmunity.

[20]  G. Vanham,et al.  Expression of activation antigens, HLA‐DR and CD38, on CD8 lymphocytes during HIV‐1 infection , 1992, AIDS.