Checkpoint blockade toxicities: Insights into autoimmunity and treatment

Seminars in Immunology - Tập 52 - Trang 101473 - 2021
Michael J. Walsh1,2,3, Michael Dougan4,5
1Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
2Harvard Program in Virology, MA, USA
3Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
4Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
5Harvard Medical School, Boston, MA, USA

Tài liệu tham khảo

Ribas, 2018, Cancer immunotherapy using checkpoint blockade, Science, 359, 1350, 10.1126/science.aar4060 Baumeister, 2016, Coinhibitory pathways in immunotherapy for cancer, Annu. Rev. Immunol., 34, 539, 10.1146/annurev-immunol-032414-112049 Sade-Feldman, 2018, Defining T cell states associated with response to checkpoint immunotherapy in melanoma, Cell, 175, 998, 10.1016/j.cell.2018.10.038 Wang, 2018, Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis, JAMA Oncol., 4, 1721, 10.1001/jamaoncol.2018.3923 Dougan, 2017, Checkpoint blockade toxicity and immune homeostasis in the gastrointestinal tract, Front. Immunol., 8, 1547, 10.3389/fimmu.2017.01547 Chang, 2019, Endocrine toxicity of Cancer immunotherapy targeting immune checkpoints, Endocr. Rev., 40, 17, 10.1210/er.2018-00006 Cappelli, 2017, Rheumatic and musculoskeletal immune-related adverse events due to immune checkpoint inhibitors: a systematic review of the literature, Arthritis Care Res. (Hoboken), 69, 1751, 10.1002/acr.23177 Hodi, 2010, Improved survival with ipilimumab in patients with metastatic melanoma, N. Engl. J. Med., 363, 711, 10.1056/NEJMoa1003466 Eggermont, 2016, Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy, N. Engl. J. Med., 375, 1845, 10.1056/NEJMoa1611299 Robert, 2015, Pembrolizumab versus ipilimumab in advanced melanoma, N. Engl. J. Med., 372, 2521, 10.1056/NEJMoa1503093 Larkin, 2015, Combined nivolumab and ipilimumab or monotherapy in untreated melanoma, N. Engl. J. Med., 373, 23, 10.1056/NEJMoa1504030 Paz-Ares, 2018, Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer, N. Engl. J. Med., 379, 2040, 10.1056/NEJMoa1810865 Rini, 2019, Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma, N. Engl. J. Med., 380, 1116, 10.1056/NEJMoa1816714 Schmid, 2018, Atezolizumab and nab-paclitaxel in advanced triple-negative breast Cancer, N. Engl. J. Med., 379, 2108, 10.1056/NEJMoa1809615 Hellmann, 2018, Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden, N. Engl. J. Med., 378, 2093, 10.1056/NEJMoa1801946 Dougan, 2019, Cancer immunotherapy: beyond checkpoint blockade, Annu. Rev. Cancer Biol., 3, 55, 10.1146/annurev-cancerbio-030518-055552 van der Merwe, 1997, CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics, J. Exp. Med., 185, 393, 10.1084/jem.185.3.393 Schwartz, 2001, Structural basis for co-stimulation by the human CTLA-4/B7-2 complex, Nature, 410, 604, 10.1038/35069112 Qureshi, 2011, Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4, Science, 332, 600, 10.1126/science.1202947 Krummel, 1995, CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation, J. Exp. Med., 182, 459, 10.1084/jem.182.2.459 Tivol, 1995, Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4, Immunity, 3, 541, 10.1016/1074-7613(95)90125-6 Kuehn, 2014, Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4, Science, 345, 1623, 10.1126/science.1255904 Wing, 2008, CTLA-4 control over Foxp3+ regulatory T cell function, Science, 322, 271, 10.1126/science.1160062 Keir, 2008, PD-1 and its ligands in tolerance and immunity, Annu. Rev. Immunol., 26, 677, 10.1146/annurev.immunol.26.021607.090331 Wang, 2017, Molecular dissection of CD8(+) T-Cell dysfunction, Trends Immunol., 38, 567, 10.1016/j.it.2017.05.008 Keir, 2006, Tissue expression of PD-L1 mediates peripheral T cell tolerance, J. Exp. Med., 203, 883, 10.1084/jem.20051776 Latchman, 2001, PD-L2 is a second ligand for PD-1 and inhibits T cell activation, Nat. Immunol., 2, 261, 10.1038/85330 Freeman, 2000, Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation, J. Exp. Med., 192, 1027, 10.1084/jem.192.7.1027 Nishimura, 1999, Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor, Immunity, 11, 141, 10.1016/S1074-7613(00)80089-8 Luoma, 2020, Molecular pathways of Colon inflammation induced by Cancer immunotherapy, Cell, 182, 655, 10.1016/j.cell.2020.06.001 Bluestone, 2020, Tolerance in the age of immunotherapy, N. Engl. J. Med., 383, 1156, 10.1056/NEJMra1911109 Anderson, 2016, Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation, Immunity, 44, 989, 10.1016/j.immuni.2016.05.001 Ingram, 2018, Anti-CTLA-4 therapy requires an Fc domain for efficacy, Proc. Natl. Acad. Sci. U. S. A., 115, 3912, 10.1073/pnas.1801524115 Simpson, 2013, Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma, J. Exp. Med., 210, 1695, 10.1084/jem.20130579 Selby, 2013, Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells, Cancer Immunol. Res., 1, 32, 10.1158/2326-6066.CIR-13-0013 Wildin, 2001, X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy, Nat. Genet., 27, 18, 10.1038/83707 Sage, 2013, The receptor PD-1 controls follicular regulatory T cells in the lymph nodes and blood, Nat. Immunol., 14, 152, 10.1038/ni.2496 Weber, 2009, A randomized, double-blind, placebo-controlled, phase II study comparing the tolerability and efficacy of ipilimumab administered with or without prophylactic budesonide in patients with unresectable stage III or IV melanoma, Clin. Cancer Res., 15, 5591, 10.1158/1078-0432.CCR-09-1024 Hasan Ali, 2019, Human leukocyte antigen variation is associated with adverse events of checkpoint inhibitors, Eur. J. Cancer, 107, 8, 10.1016/j.ejca.2018.11.009 Stamatouli, 2018, Collateral damage: insulin-dependent diabetes induced with checkpoint inhibitors, Diabetes, 67, 1471, 10.2337/dbi18-0002 Abu-Sbeih, 2019, Immune checkpoint inhibitor therapy in patients with preexisting inflammatory bowel disease, J. Clin. Oncol. Akturk, 2018, PD-1 inhibitor immune-related adverse events in patients with preexisting endocrine autoimmunity, J. Clin. Endocrinol. Metab., 103, 3589, 10.1210/jc.2018-01430 Menzies, 2017, Anti-PD-1 therapy in patients with advanced melanoma and preexisting autoimmune disorders or major toxicity with ipilimumab, Ann. Oncol., 28, 368, 10.1093/annonc/mdw443 Leonardi, 2018, Safety of programmed Death-1 pathway inhibitors among patients with non-small-Cell lung Cancer and preexisting autoimmune disorders, J. Clin. Oncol., 36, 1905, 10.1200/JCO.2017.77.0305 Johnson, 2016, Ipilimumab therapy in patients with advanced melanoma and preexisting autoimmune disorders, JAMA Oncol., 2, 234, 10.1001/jamaoncol.2015.4368 Badran, 2020, Immune checkpoint inhibitor-associated celiac disease, J. Immunother. Cancer, 8, 10.1136/jitc-2020-000958 Safa, 2019, Immune checkpoint inhibitor related myasthenia gravis: single center experience and systematic review of the literature, J. Immunother. Cancer, 7, 319, 10.1186/s40425-019-0774-y Martins, 2019, Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance, Nat. Rev. Clin. Oncol., 16, 563, 10.1038/s41571-019-0218-0 Dougan, 2020, AGA clinical practice update on diagnosis and management of immune checkpoint inhibitor (ICI) colitis and hepatitis: expert review, Gastroenterology Seethapathy, 2020, Incidence and clinical features of immune-related acute kidney injury in patients receiving programmed cell death Ligand-1 inhibitors, Kidney Int. Rep., 5, 1700, 10.1016/j.ekir.2020.07.011 Correale, 2006, The risk of relapses in multiple sclerosis during systemic infections, Neurology, 67, 652, 10.1212/01.wnl.0000233834.09743.3b Brahmer, 2018, Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: american society of clinical oncology clinical practice guideline, J. Clin. Oncol., 10.1200/JCO.2017.77.6385 Haanen, 2017, Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up, Ann. Oncol., 28, iv119, 10.1093/annonc/mdx225 Puzanov, 2017, Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group, J. Immunother. Cancer, 5, 95, 10.1186/s40425-017-0300-z Acharya, 2020, Endogenous glucocorticoid signaling regulates CD8(+) t cell differentiation and development of dysfunction in the tumor microenvironment, Immunity, 53, 658, 10.1016/j.immuni.2020.08.005 Arbour, 2018, Impact of baseline steroids on efficacy of programmed cell Death-1 and programmed death-ligand 1 blockade in patients with non-small-Cell lung Cancer, J. Clin. Oncol., 36, 2872, 10.1200/JCO.2018.79.0006 Faje, 2018, High-dose glucocorticoids for the treatment of ipilimumab-induced hypophysitis is associated with reduced survival in patients with melanoma, Cancer, 10.1002/cncr.31629 Keskin, 2019, Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial, Nature, 565, 234, 10.1038/s41586-018-0792-9 Tawbi, 2018, Nivolumab and ipilimumab in melanoma metastatic to the brain, N. Engl. J. Med., 379, 2178, 10.1056/NEJMoa1805453 Badran, 2019, Concurrent therapy with immune checkpoint inhibitors and TNFalpha blockade in patients with gastrointestinal immune-related adverse events, J. Immunother. Cancer, 7, 226, 10.1186/s40425-019-0711-0 Hughes, 2019, Budesonide treatment for microscopic colitis from immune checkpoint inhibitors, J. Immunother. Cancer, 7, 292, 10.1186/s40425-019-0756-0 Cohen, 2020, Liver biopsy findings in patients on immune checkpoint inhibitors, Mod. Pathol. Johnson, 2016, Fulminant myocarditis with combination immune checkpoint blockade, N. Engl. J. Med., 375, 1749, 10.1056/NEJMoa1609214 Phillips, 2019, Treatment outcomes of immune-related cutaneous adverse events, J. Clin. Oncol., 37, 2746, 10.1200/JCO.18.02141 Salem, 2019, Abatacept for severe immune checkpoint inhibitor-associated myocarditis, N. Engl. J. Med., 380, 2377, 10.1056/NEJMc1901677 Bishu, 2020, Efficacy and outcome of Tofacitinib in Immune checkpoint inhibitor colitis, Gastroenterology Spankuch, 2017, Severe hepatitis under combined immunotherapy: resolution under corticosteroids plus anti-thymocyte immunoglobulins, Eur. J. Cancer, 81, 203, 10.1016/j.ejca.2017.05.018 Abu-Sbeih, 2018, Outcomes of vedolizumab therapy in patients with immune checkpoint inhibitor-induced colitis: a multi-center study, J. Immunother. Cancer, 6, 142, 10.1186/s40425-018-0461-4 Teachey, 2016, Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia, Cancer Discov., 6, 664, 10.1158/2159-8290.CD-16-0040 Abu-Sbeih, 2019, Early introduction of selective immunosuppressive therapy associated with favorable clinical outcomes in patients with immune checkpoint inhibitor-induced colitis, J. Immunother. Cancer, 7, 93, 10.1186/s40425-019-0577-1 Mooradian, 2020, Mucosal inflammation predicts response to systemic steroids in immune checkpoint inhibitor colitis, J. Immunother. Cancer, 8, 10.1136/jitc-2019-000451 Ridker, 2017, Effect of interleukin-1beta inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial, Lancet, 390, 1833, 10.1016/S0140-6736(17)32247-X Bertrand, 2017, TNFalpha blockade overcomes resistance to anti-PD-1 in experimental melanoma, Nat. Commun., 8, 2256, 10.1038/s41467-017-02358-7 Perez-Ruiz, 2019, Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy, Nature, 569, 428, 10.1038/s41586-019-1162-y Chen, 2016, Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade, Cancer Discov., 6, 827, 10.1158/2159-8290.CD-15-1545 Gao, 2016, Loss of IFN-gamma pathway genes in tumor cells as a mechanism of resistance to Anti-CTLA-4 therapy, Cell, 167, 397, 10.1016/j.cell.2016.08.069 Gettinger, 2017, Impaired HLA class I antigen processing and presentation as a mechanism of acquired resistance to immune checkpoint inhibitors in lung Cancer, Cancer Discov., 7, 1420, 10.1158/2159-8290.CD-17-0593 Manguso, 2017, In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target, Nature, 547, 413, 10.1038/nature23270 McGranahan, 2017, Allele-specific HLA loss and immune escape in lung cancer evolution, Cell, 171, 1259, 10.1016/j.cell.2017.10.001 Sade-Feldman, 2017, Resistance to checkpoint blockade therapy through inactivation of antigen presentation, Nat. Commun., 8, 1136, 10.1038/s41467-017-01062-w Shin, 2017, Primary resistance to PD-1 blockade mediated by JAK1/2 mutations, Cancer Discov., 7, 188, 10.1158/2159-8290.CD-16-1223 Zaretsky, 2016, Mutations associated with acquired resistance to PD-1 blockade in melanoma, N. Engl. J. Med., 375, 819, 10.1056/NEJMoa1604958 Verheijden, 2020, Association of Anti-TNF with decreased survival in steroid refractory ipilimumab and Anti-PD1-Treated patients in the dutch melanoma treatment registry, Clin. Cancer Res., 26, 2268, 10.1158/1078-0432.CCR-19-3322