A network approach to define the predictive role of immune profile on tumor response and toxicity of anti PD-1 single agent immunotherapy in patients with solid tumors

Background The immune profile of each patient could be considered as a portrait of the fitness of his/her own immune system. The predictive role of the immune profile in immune-related toxicities (irAEs) development and tumour response to treatment was investigated. Methods A prospective, multicenter study evaluating, through a multiplex assay, the soluble immune profile at the baseline of 53 patients with advanced cancer, treated with immunotherapy as single agent was performed. Four connectivity heat maps and networks were obtained by calculating the Spearman correlation coefficients for each group: responder patients who developed cumulative toxicity (R-T), responders who did not develop cumulative toxicity (R-NT), non-responders who developed cumulative toxicity (NR-T), non-responders who did not develop cumulative toxicity (NR-NT). Results A statistically significant up-regulation of IL-17A, sCTLA4, sCD80, I-CAM-1, sP-Selectin and sEselectin in NR-T was detected. A clear loss of connectivity of most of the soluble immune checkpoints and cytokines characterized the immune profile of patients with toxicity, while an inversion of the correlation for ICAM-1 and sP-selectin was observed in NR-T. Four connectivity networks were built for each group. The highest number of connections characterized the NR-T. Conclusions A connectivity network of immune dysregulation was defined for each subgroup of patients, regardless of tumor type. In patients with the worst prognosis (NR-T) the peculiar connectivity model could facilitate their early and timely identification, as well as the design of a personalized treatment approach to improve outcomes or prevent irAEs.

[1]  L. Farina,et al.  Immune-related toxicity and soluble profile in patients affected by solid tumors: a network approach , 2023, Cancer Immunology, Immunotherapy.

[2]  G. Tortora,et al.  The role of immune profile in predicting outcomes in cancer patients treated with immunotherapy , 2022, Frontiers in Immunology.

[3]  P. Marchetti,et al.  Genomic and Immune Approach in Platinum Refractory HPV-Negative Head and Neck Squamous Cell Carcinoma Patients Treated with Immunotherapy: A Novel Combined Profile , 2022, Biomedicines.

[4]  M. Suarez‐Almazor,et al.  Management of Immune-Related Adverse Events in Patients Treated With Chimeric Antigen Receptor T-Cell Therapy: ASCO Guideline , 2021, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[5]  M. Suarez‐Almazor,et al.  Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update , 2021, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[6]  Xudan Wang,et al.  Potentiality of α-fetoprotein (AFP) and soluble intercellular adhesion molecule-1 (sICAM-1) in prognosis prediction and immunotherapy response for patients with hepatocellular carcinoma , 2021, Bioengineered.

[7]  R. Sullivan,et al.  Immune-related toxicities of checkpoint inhibitors: mechanisms and mitigation strategies , 2021, Nature Reviews Drug Discovery.

[8]  L. Strigari,et al.  The Role of Soluble LAG3 and Soluble Immune Checkpoints Profile in Advanced Head and Neck Cancer: A Pilot Study , 2021, Journal of personalized medicine.

[9]  P. Libby,et al.  Interleukins in cancer: from biology to therapy , 2021, Nature Reviews Cancer.

[10]  A. Tafreshi,et al.  Five-Year Outcomes With Pembrolizumab Versus Chemotherapy for Metastatic Non–Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50% , 2021, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[11]  A. Luster,et al.  Chemokines and the immune response to cancer. , 2021, Immunity.

[12]  D. Cella,et al.  Health‐related quality‐of‐life assessment of patients with solid tumors on immuno‐oncology therapies , 2021, Cancer.

[13]  C. Porta,et al.  Lenvatinib plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. , 2021, The New England journal of medicine.

[14]  J. Loscalzo,et al.  Gene co-expression in the interactome: moving from correlation toward causation via an integrated approach to disease module discovery , 2021, NPJ systems biology and applications.

[15]  Yanqiao Zhang,et al.  Blocking IL-17A enhances tumor response to anti-PD-1 immunotherapy in microsatellite stable colorectal cancer , 2021, Journal for ImmunoTherapy of Cancer.

[16]  M. Milella,et al.  Infections and Immunotherapy in Lung Cancer: A Bad Relationship? , 2020, International journal of molecular sciences.

[17]  M. Filetti,et al.  Effect of concomitant medications with immune-modulatory properties on the outcomes of patients with advanced cancer treated with immune checkpoint inhibitors: development and validation of a novel prognostic index. , 2020, European journal of cancer.

[18]  P. Marchetti,et al.  Soluble Immune Checkpoints, Gut Metabolites and Performance Status as Parameters of Response to Nivolumab Treatment in NSCLC Patients , 2020, Journal of personalized medicine.

[19]  C. Porta,et al.  Nivolumab plus ipilimumab versus sunitinib for first-line treatment of advanced renal cell carcinoma: extended 4-year follow-up of the phase III CheckMate 214 trial , 2020, ESMO Open.

[20]  S. Patel,et al.  Nivolumab and Ipilimumab in Metastatic Uveal Melanoma: Results From a Single-Arm Phase II Study. , 2020, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  T. Powles,et al.  Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. , 2020, The Lancet. Oncology.

[22]  L. Del Mastro,et al.  New emerging targets in cancer immunotherapy: the role of GITR , 2020, ESMO open.

[23]  R. Motzer,et al.  Nivolumab versus everolimus in patients with advanced renal cell carcinoma: Updated results with long-term follow-up of the randomized, open-label, phase 3 CheckMate 025 trial. , 2020, Cancer.

[24]  M. Suarez‐Almazor,et al.  Immune-related adverse events of checkpoint inhibitors , 2020, Nature Reviews Disease Primers.

[25]  Fengchun Zhang,et al.  Are immune-related adverse events associated with the efficacy of immune checkpoint inhibitors in patients with cancer? A systematic review and meta-analysis , 2020, BMC Medicine.

[26]  Marco Angelini,et al.  Molecular networks in Network Medicine: Development and applications , 2020, Wiley interdisciplinary reviews. Systems biology and medicine.

[27]  K. Reynolds,et al.  Immune-Related Adverse Events (irAEs): Diagnosis, Management, and Clinical Pearls , 2020, Current Oncology Reports.

[28]  H. Schwarz,et al.  The relevance of soluble CD137 in the regulation of immune responses and for immunotherapeutic intervention , 2020, Journal of leukocyte biology.

[29]  S. Lee,et al.  Clinical Characteristics and Treatment of Immune-Related Adverse Events of Immune Checkpoint Inhibitors , 2020, Immune network.

[30]  P. Vidal Interferon α in cancer immunoediting: From elimination to escape , 2020, Scandinavian journal of immunology.

[31]  J. Utikal,et al.  Elevated baseline serum PD-1 or PD-L1 predicts poor outcome of PD-1 inhibition therapy in metastatic melanoma. , 2020, Annals of oncology : official journal of the European Society for Medical Oncology.

[32]  Chul Kim,et al.  Safety and efficacy of immune checkpoint inhibitors (ICIs) in cancer patients with HIV, hepatitis B, or hepatitis C viral infection , 2019, Journal of Immunotherapy for Cancer.

[33]  C. Crowson,et al.  Family History of Rheumatic, Autoimmune, and Nonautoimmune Diseases and Risk of Rheumatoid Arthritis , 2019, Arthritis care & research.

[34]  A. Adler,et al.  IL-17 inhibits CXCL9/10-mediated recruitment of CD8+ cytotoxic T cells and regulatory T cells to colorectal tumors , 2019, Journal of Immunotherapy for Cancer.

[35]  Hung-Ming Wang,et al.  Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study , 2019, The Lancet.

[36]  N. Rezaei,et al.  IL‐17 and colorectal cancer: From carcinogenesis to treatment , 2019, Cytokine.

[37]  Hannah A. Blair Secukinumab: A Review in Ankylosing Spondylitis , 2019, Drugs.

[38]  C. Lin,et al.  Immune Checkpoint Immunotherapy for Non-Small Cell Lung Cancer: Benefits and Pulmonary Toxicities. , 2018, Chest.

[39]  R. Sullivan,et al.  Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis , 2018, JAMA oncology.

[40]  P. Ascierto,et al.  Soluble CTLA-4 as a favorable predictive biomarker in metastatic melanoma patients treated with ipilimumab: an Italian melanoma intergroup study , 2018, Cancer Immunology, Immunotherapy.

[41]  L. Macconaill,et al.  Frameshift events predict anti-PD-1/L1 response in head and neck cancer. , 2018, JCI insight.

[42]  Matthew D. Hellmann,et al.  Immune‐Related Adverse Events Associated with Immune Checkpoint Blockade , 2018, The New England journal of medicine.

[43]  T. Uede,et al.  Implication of Soluble Forms of Cell Adhesion Molecules in Infectious Disease and Tumor: Insights from Transgenic Animal Models , 2018, International journal of molecular sciences.

[44]  N. Girard,et al.  Immune-checkpoint inhibitors associated with interstitial lung disease in cancer patients , 2017, European Respiratory Journal.

[45]  K. Kerr,et al.  Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. , 2017, Annals of oncology : official journal of the European Society for Medical Oncology.

[46]  J. Lang,et al.  Soluble PD-1 and PD-L1: predictive and prognostic significance in cancer. , 2017, Oncotarget.

[47]  K. Shen,et al.  Recent progress in GM-CSF-based cancer immunotherapy. , 2017, Immunotherapy.

[48]  R. Iacovelli,et al.  Prognostic role of the cumulative toxicity in patients affected by metastatic renal cells carcinoma and treated with first-line tyrosine kinase inhibitors , 2017, Anti-cancer drugs.

[49]  Carlos Barrios,et al.  Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial , 2017, The Lancet.

[50]  R. Sullivan,et al.  Clinical outcomes in metastatic uveal melanoma treated with PD‐1 and PD‐L1 antibodies , 2016, Cancer.

[51]  K. Harrington,et al.  Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. , 2016, The New England journal of medicine.

[52]  R. McWilliams,et al.  The use of pembrolizumab for the treatment of metastatic uveal melanoma , 2016, Melanoma research.

[53]  Y. Shentu,et al.  Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial , 2016, The Lancet.

[54]  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.

[55]  J. Taube,et al.  Antagonists of PD-1 and PD-L1 in Cancer Treatment. , 2015, Seminars in oncology.

[56]  L. Crinò,et al.  Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. , 2015, The New England journal of medicine.

[57]  B. Eynde,et al.  Tryptophan-Degrading Enzymes in Tumoral Immune Resistance , 2015, Front. Immunol..

[58]  Huaxi Xu,et al.  Th17/Treg Cells Imbalance and GITRL Profile in Patients with Hashimoto’s Thyroiditis , 2014, International journal of molecular sciences.

[59]  F. Qiu,et al.  γδT17 cells promote the accumulation and expansion of myeloid-derived suppressor cells in human colorectal cancer. , 2014, Immunity.

[60]  Xiaoxuan Sun,et al.  Correlation of Increased Blood Levels of GITR and GITRL with Disease Severity in Patients with Primary Sjögren's Syndrome , 2013, Clinical & developmental immunology.

[61]  Y. Min,et al.  The presence of high level soluble herpes virus entry mediator in sera of gastric cancer patients , 2011, Experimental & Molecular Medicine.

[62]  J. Galon,et al.  Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. , 2011, Cancer research.

[63]  G. Pesce,et al.  The soluble CTLA-4 receptor and its role in autoimmune diseases: an update , 2010, Autoimmunity Highlights.

[64]  I. Campbell Chi‐squared and Fisher–Irwin tests of two‐by‐two tables with small sample recommendations , 2007, Statistics in medicine.

[65]  R. Ferris,et al.  Immune Escape Associated with Functional Defects in Antigen-Processing Machinery in Head and Neck Cancer , 2006, Clinical Cancer Research.

[66]  S. Quezada,et al.  Principles and use of anti-CTLA4 antibody in human cancer immunotherapy. , 2006, Current opinion in immunology.

[67]  C. Riccardi,et al.  GITR: a multifaceted regulator of immunity belonging to the tumor necrosis factor receptor superfamily , 2005, European journal of immunology.

[68]  J. Giddings Soluble adhesion molecules in inflammatory and vascular diseases. , 2005, Biochemical Society transactions.

[69]  M. Volin Soluble adhesion molecules in the pathogenesis of rheumatoid arthritis. , 2005, Current pharmaceutical design.

[70]  H. Waldmann,et al.  Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[71]  J. Kirkwood,et al.  Interferon-alpha in tumor immunity and immunotherapy. , 2002, Cytokine & growth factor reviews.

[72]  J. Shimizu,et al.  Stimulation of CD25+CD4+ regulatory T cells through GITR breaks immunological self-tolerance , 2002, Nature Immunology.

[73]  D. Cook The role of MIP‐1α in Inflammation and hematopoiesis , 1996, Journal of leukocyte biology.

[74]  A. Barabasi,et al.  Network medicine : a network-based approach to human disease , 2010 .

[75]  A. Molina,et al.  Soluble adhesion molecules in renal transplantation. , 1996, Renal failure.