Peripheral blood immune cell-based biomarkers in anti-PD-1/PD-L1 therapy

Immune checkpoint blockade targeting PD-1 and PD-L1 has resulted in unprecedented clinical benefit for cancer patients. Anti-PD-1/PD-L1 therapy has become the standard treatment for diverse cancer types as monotherapy or in combination with other anti-cancer therapies, and its indications are expanding. However, many patients do not benefit from anti-PD-1/PD-L1 therapy due to primary and/or acquired resistance, which is a major obstacle to broadening the clinical applicability of anti-PD-1/PD-L1 therapy. In addition, hyperprogressive disease, an acceleration of tumor growth following anti-PD-1/PD-L1 therapy, has been proposed as a new response pattern associated with deleterious prognosis. Anti-PD-1/PD-L1 therapy can also cause a unique pattern of adverse events termed immune-related adverse events, sometimes leading to treatment discontinuation and fatal outcomes. Investigations have been carried out to predict and monitor treatment outcomes using peripheral blood as an alternative to tissue biopsy. This review summarizes recent studies utilizing peripheral blood immune cells to predict various outcomes in cancer patients treated with anti-PD-1/PD-L1 therapy.

[1]  J. Ahn,et al.  Immune-related adverse events are clustered into distinct subtypes by T-cell profiling before and early after anti-PD-1 treatment , 2020, Oncoimmunology.

[2]  Jeffrey E. Lee,et al.  B cells and tertiary lymphoid structures promote immunotherapy response , 2020, Nature.

[3]  J. Wargo,et al.  B cells are associated with survival and immunotherapy response in sarcoma , 2020, Nature.

[4]  D. Schadendorf,et al.  Tertiary lymphoid structures improve immunotherapy and survival in melanoma , 2020, Nature.

[5]  Shuhang Wang,et al.  TCR Repertoire Diversity of Peripheral PD-1+CD8+ T Cells Predicts Clinical Outcomes after Immunotherapy in Patients with Non–Small Cell Lung Cancer , 2019, Cancer Immunology Research.

[6]  J. Gartner,et al.  Recognition of human gastrointestinal cancer neoantigens by circulating PD-1+ lymphocytes. , 2019, The Journal of clinical investigation.

[7]  E. Wherry,et al.  Defining ‘T cell exhaustion’ , 2019, Nature Reviews Immunology.

[8]  M. Rizzo,et al.  Peripheral changes in immune cell populations and soluble mediators after anti-PD-1 therapy in non-small cell lung cancer and renal cell carcinoma patients , 2019, Cancer Immunology, Immunotherapy.

[9]  Ho Yun Lee,et al.  Comprehensive Clinical and Genetic Characterization of Hyperprogression Based on Volumetry in Advanced Non-Small Cell Lung Cancer Treated With Immune Checkpoint Inhibitor. , 2019, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[10]  David L Rimm,et al.  Comparison of Biomarker Modalities for Predicting Response to PD-1/PD-L1 Checkpoint Blockade: A Systematic Review and Meta-analysis. , 2019, JAMA oncology.

[11]  B. Cho,et al.  Hyperprogressive disease during PD-1/PD-L1 blockade in patients with non-small-cell lung cancer. , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[12]  G. Kochan,et al.  Functional systemic CD4 immunity is required for clinical responses to PD‐L1/PD‐1 blockade therapy , 2019, EMBO molecular medicine.

[13]  S. Mallal,et al.  A case report of clonal EBV-like memory CD4+ T cell activation in fatal checkpoint inhibitor-induced encephalitis , 2019, Nature Medicine.

[14]  Hiromasa Morikawa,et al.  PD-1+ regulatory T cells amplified by PD-1 blockade promote hyperprogression of cancer , 2019, Proceedings of the National Academy of Sciences.

[15]  E. Wherry,et al.  CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. , 2019, Annual review of immunology.

[16]  P. Hwu,et al.  IL17A Blockade Successfully Treated Psoriasiform Dermatologic Toxicity from Immunotherapy , 2019, Cancer Immunology Research.

[17]  J. Sugisaka,et al.  Profiling Preexisting Antibodies in Patients Treated With Anti–PD-1 Therapy for Advanced Non–Small Cell Lung Cancer , 2019, JAMA oncology.

[18]  G. Linette,et al.  A Single Dose of Neoadjuvant PD-1 Blockade Predicts Clinical Outcomes in Resectable Melanoma , 2019, Nature Medicine.

[19]  J. Ahn,et al.  The First-week Proliferative Response of Peripheral Blood PD-1+CD8+ T Cells Predicts the Response to Anti-PD-1 Therapy in Solid Tumors , 2019, Clinical Cancer Research.

[20]  S. Ha,et al.  The Ratio of Peripheral Regulatory T Cells to Lox-1+ Polymorphonuclear Myeloid-derived Suppressor Cells Predicts the Early Response to Anti-PD-1 Therapy in Patients with Non-Small Cell Lung Cancer. , 2019, American journal of respiratory and critical care medicine.

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

[22]  Edmond J. Breen,et al.  Circulating Cytokines Predict Immune-Related Toxicity in Melanoma Patients Receiving Anti-PD-1–Based Immunotherapy , 2018, Clinical Cancer Research.

[23]  D. Planchard,et al.  Hyperprogressive Disease in Patients With Advanced Non–Small Cell Lung Cancer Treated With PD-1/PD-L1 Inhibitors or With Single-Agent Chemotherapy , 2018, JAMA oncology.

[24]  J. Soria,et al.  Hyperprogressive disease: recognizing a novel pattern to improve patient management , 2018, Nature Reviews Clinical Oncology.

[25]  J. Lunceford,et al.  Pan-tumor genomic biomarkers for PD-1 checkpoint blockade–based immunotherapy , 2018, Science.

[26]  A. Mammen,et al.  Pre-existing antiacetylcholine receptor autoantibodies and B cell lymphopaenia are associated with the development of myositis in patients with thymoma treated with avelumab, an immune checkpoint inhibitor targeting programmed death-ligand 1 , 2018, Annals of the rheumatic diseases.

[27]  K. Hargadon,et al.  Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. , 2018, International immunopharmacology.

[28]  E. Jaffee,et al.  T cell receptor repertoire features associated with survival in immunotherapy-treated pancreatic ductal adenocarcinoma. , 2018, JCI insight.

[29]  Tao Jiang,et al.  Heterogeneity of PD‐L1 Expression Among the Different Histological Components and Metastatic Lymph Nodes in Patients With Resected Lung Adenosquamous Carcinoma , 2018, Clinical lung cancer.

[30]  F. Prósper,et al.  Anti-PD1 associated fulminant myocarditis after a single pembrolizumab dose: the role of occult pre-existing autoimmunity , 2018, Haematologica.

[31]  C. Klein,et al.  A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small cell lung cancer treated with PD-1 blockade , 2018, Nature Network Boston.

[32]  E. Wherry,et al.  Non-conventional Inhibitory CD4+Foxp3-PD-1hi T Cells as a Biomarker of Immune Checkpoint Blockade Activity. , 2018, Cancer cell.

[33]  M. Fehlings,et al.  Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates , 2018, Nature.

[34]  Jedd D. Wolchok,et al.  Cancer immunotherapy using checkpoint blockade , 2018, Science.

[35]  S. Baxi,et al.  Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis , 2018, British Medical Journal.

[36]  P. Hegde,et al.  Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial , 2018, The Lancet.

[37]  A. Kamphorst,et al.  CD8 T Cell Exhaustion in Chronic Infection and Cancer: Opportunities for Interventions. , 2018, Annual review of medicine.

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

[39]  Reinhard Dummer,et al.  High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy , 2018, Nature Network Boston.

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

[41]  J. Cho Immunotherapy for Non-small-cell Lung Cancer: Current Status and Future Obstacles , 2017, Immune network.

[42]  P. Hofman PD-L1 immunohistochemistry for non-small cell lung carcinoma: which strategy should be adopted? , 2017, Expert review of molecular diagnostics.

[43]  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.

[44]  V. Servois,et al.  Hyperprogression during anti-PD-1/PD-L1 therapy in patients with recurrent and/or metastatic head and neck squamous cell carcinoma , 2017, Annals of oncology : official journal of the European Society for Medical Oncology.

[45]  W. Miller,et al.  Reversal of Autoimmune Toxicity and Loss of Tumor Response by Interleukin-17 Blockade. , 2017, The New England journal of medicine.

[46]  J. Bluestone,et al.  Is autoimmunity the Achilles' heel of cancer immunotherapy? , 2017, Nature Medicine.

[47]  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.

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

[49]  Razelle Kurzrock,et al.  Hyperprogressors after Immunotherapy: Analysis of Genomic Alterations Associated with Accelerated Growth Rate , 2017, Clinical Cancer Research.

[50]  S. Culine,et al.  Pembrolizumab as Second‐Line Therapy for Advanced Urothelial Carcinoma , 2017, The New England journal of medicine.

[51]  J. Wargo,et al.  Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy , 2017, Cell.

[52]  M. Millward,et al.  Dynamic versus static biomarkers in cancer immune checkpoint blockade: unravelling complexity , 2017, Nature Reviews Drug Discovery.

[53]  C. Rudin,et al.  Antibody-mediated thyroid dysfunction during T-cell checkpoint blockade in patients with non-small-cell lung cancer , 2016, Annals of oncology : official journal of the European Society for Medical Oncology.

[54]  J. Radford Nivolumab for recurrent squamous-cell carcinoma of the head and neck , 2016, BDJ.

[55]  Charles Ferté,et al.  Hyperprogressive Disease Is a New Pattern of Progression in Cancer Patients Treated by Anti-PD-1/PD-L1 , 2016, Clinical Cancer Research.

[56]  J. Seidman,et al.  Fulminant Myocarditis with Combination Immune Checkpoint Blockade. , 2016, The New England journal of medicine.

[57]  M. Vetizou,et al.  Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. , 2016, Immunity.

[58]  J. Wolchok,et al.  Targeting T Cell Co-receptors for Cancer Therapy. , 2016, Immunity.

[59]  Antoni Ribas,et al.  The “cancer immunogram” , 2016, Science.

[60]  J. Gartner,et al.  Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients , 2016, Nature Medicine.

[61]  T. Fleisher Immune Dysregulation in Human Subjects With Heterozygous Germline Mutations in CTLA4 , 2015, Pediatrics.

[62]  C. Rudin,et al.  Nivolumab versus Docetaxel in Advanced Nonsquamous Non-Small-Cell Lung Cancer. , 2015, The New England journal of medicine.

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

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

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

[66]  I. Mellman,et al.  Oncology meets immunology: the cancer-immunity cycle. , 2013, Immunity.

[67]  P. A. Futreal,et al.  Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. , 2012, The New England journal of medicine.

[68]  T. Nomura,et al.  Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. , 2009, Immunity.

[69]  Thomas Korn,et al.  IL-17 and Th17 Cells. , 2009, Annual review of immunology.

[70]  P. Tak,et al.  Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. , 2008, Pharmacology & therapeutics.

[71]  V. Kuchroo,et al.  TH-17 cells in the circle of immunity and autoimmunity , 2007, Nature Immunology.

[72]  P. Santamaria,et al.  CD8+ T cells in autoimmunity. , 2005, Current opinion in immunology.

[73]  P. Lipsky Systemic lupus erythematosus: an autoimmune disease of B cell hyperactivity , 2001, Nature Immunology.

[74]  J. Altman,et al.  Viral Immune Evasion Due to Persistence of Activated T Cells Without Effector Function , 1998, The Journal of experimental medicine.

[75]  R. Zinkernagel,et al.  Induction and Exhaustion of Lymphocytic Choriomeningitis Virus–specific Cytotoxic T Lymphocytes Visualized Using Soluble Tetrameric Major Histocompatibility Complex Class I–Peptide Complexes , 1998, The Journal of experimental medicine.

[76]  C. Marquette,et al.  Comparative study of the PD-L1 status between surgically resected specimens and matched biopsies of NSCLC patients reveal major discordances: a potential issue for anti-PD-L1 therapeutic strategies. , 2016, Annals of oncology : official journal of the European Society for Medical Oncology.

[77]  Shimon Sakaguchi,et al.  Regulatory T cells exert checks and balances on self tolerance and autoimmunity , 2010, Nature Immunology.