Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors
Tài liệu tham khảo
Dighe, 1994, Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors, Immunity, 1, 447, 10.1016/1074-7613(94)90087-6
Levitsky, 1994, In vivo priming of two distinct antitumor effector populations: the role of MHC class I expression, J. Exp. Med., 179, 1215, 10.1084/jem.179.4.1215
van den Broek, 1996, Decreased tumor surveillance in perforin-deficient mice, J. Exp. Med., 184, 1781, 10.1084/jem.184.5.1781
Kaplan, 1998, Demonstration of an interferon gamma-dependent tumor surveillance system in immunocompetent mice, Proc. Natl. Acad. Sci. U. S. A., 95, 7556, 10.1073/pnas.95.13.7556
Hung, 1998, The central role of CD4(+) T cells in the antitumor immune response, J. Exp. Med., 188, 2357, 10.1084/jem.188.12.2357
Smyth, 2000, Perforin-mediated cytotoxicity is critical for surveillance of spontaneous lymphoma, J. Exp. Med., 192, 755, 10.1084/jem.192.5.755
Shankaran, 2001, IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity, Nature, 410, 1107, 10.1038/35074122
Smith, 1965, Spontaneous regression of primary malignant melanomas with regional metastases, Cancer, 18, 1399, 10.1002/1097-0142(196511)18:11<1399::AID-CNCR2820181104>3.0.CO;2-R
Saleh, 2001, Primary melanoma tumour regression associated with an immune response to the tumour-associated antigen melan-A/MART-1, Int. J. Cancer, 94, 551, 10.1002/ijc.1491
Deng, 2010, Accumulation of foxp3+ T regulatory cells in draining lymph nodes correlates with disease progression and immune suppression in colorectal cancer patients, Clin. Cancer Res., 16, 4105, 10.1158/1078-0432.CCR-10-1073
Severson, 2015, PD-1+Tim-3+ CD8+ T lymphocytes display varied degrees of functional exhaustion in patients with regionally metastatic differentiated thyroid cancer, Cancer Immunol. Res., 3, 620, 10.1158/2326-6066.CIR-14-0201
Shahinian, 1993, Differential T cell costimulatory requirements in CD28-deficient mice, Science, 261, 609, 10.1126/science.7688139
Joffre, 2009, Inflammatory signals in dendritic cell activation and the induction of adaptive immunity, Immunol. Rev., 227, 234, 10.1111/j.1600-065X.2008.00718.x
Chen, 2013, Molecular mechanisms of T cell co-stimulation and co-inhibition, Nat. Rev. Immunol., 13, 227, 10.1038/nri3405
Walunas, 1994, CTLA-4 can function as a negative regulator of T cell activation, Immunity, 1, 405, 10.1016/1074-7613(94)90071-X
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
Greenwald, 2001, CTLA-4 regulates induction of anergy in vivo, Immunity, 14, 145, 10.1016/S1074-7613(01)00097-8
Krummel, 1996, CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells, J. Exp. Med., 183, 2533, 10.1084/jem.183.6.2533
Chambers, 1998, Secondary but not primary T cell responses are enhanced in CTLA-4-deficient CD8+ T cells, Eur. J. Immunol., 28, 3137, 10.1002/(SICI)1521-4141(199810)28:10<3137::AID-IMMU3137>3.0.CO;2-X
Wing, 2008, CTLA-4 control over Foxp3+ regulatory T cell function, Science, 322, 271, 10.1126/science.1160062
Peggs, 2009, Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti–CTLA-4 antibodies, J. Exp. Med., 206, 1717, 10.1084/jem.20082492
Wei, 2017, Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade, Cell, 170, 1120, 10.1016/j.cell.2017.07.024
Hodi, 2010, Improved survival with ipilimumab in patients with metastatic melanoma, N. Engl. J. Med., 363, 711, 10.1056/NEJMoa1003466
Schadendorf, 2015, Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma, J. Clin. Oncol., 33, 1889, 10.1200/JCO.2014.56.2736
Robert, 2011, Ipilimumab plus dacarbazine for previously untreated metastatic melanoma, N. Engl. J. Med., 364, 2517, 10.1056/NEJMoa1104621
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
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
Latchman, 2001, PD-L2 is a second ligand for PD-1 and inhibits T cell activation, Nat. Immunol., 2, 261, 10.1038/85330
Francisco, 2009, PD-L1 regulates the development, maintenance, and function of induced regulatory T cells, J. Exp. Med., 206, 3015, 10.1084/jem.20090847
Blank, 2004, PD-L1/B7H-1 inhibits the effector phase of tumor rejection by T cell receptor (TCR) transgenic CD8+ T cells, Cancer Res., 64, 1140, 10.1158/0008-5472.CAN-03-3259
Iwai, 2003, PD-1 inhibits antiviral immunity at the effector phase in the liver, J. Exp. Med., 198, 39, 10.1084/jem.20022235
Barber, 2006, Restoring function in exhausted CD8 T cells during chronic viral infection, Nature, 439, 682, 10.1038/nature04444
Ahmadzadeh, 2009, Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired, Blood, 114, 1537, 10.1182/blood-2008-12-195792
Dong, 2002, Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion, Nat. Med., 8, 793, 10.1038/nm730
Weber, 2015, Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial, Lancet Oncol., 16, 375, 10.1016/S1470-2045(15)70076-8
Robert, 2015, Nivolumab in previously untreated melanoma without BRAF mutation, N. Engl. J. Med., 372, 320, 10.1056/NEJMoa1412082
Weber, 2017, CheckMate 238 collaborators, adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma, N. Engl. J. Med., 377, 1824, 10.1056/NEJMoa1709030
Brahmer, 2015, Nivolumab versus docetaxel in advanced squamous-cell non–small-cell lung cancer, N. Engl. J. Med., 373, 123, 10.1056/NEJMoa1504627
Borghaei, 2015, Nivolumab versus docetaxel in advanced nonsquamous non–small-cell lung cancer, N. Engl. J. Med., 373, 1627, 10.1056/NEJMoa1507643
Motzer, 2015, Nivolumab versus everolimus in advanced renal-cell carcinoma, N. Engl. J. Med., 373, 1803, 10.1056/NEJMoa1510665
Ferris, 2016, Nivolumab for recurrent squamous-cell carcinoma of the head and neck, N. Engl. J. Med., 375, 1856, 10.1056/NEJMoa1602252
El-Khoueiry, 2017, Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial, Lancet, 389, 2492, 10.1016/S0140-6736(17)31046-2
Overman, 2017, Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study, Lancet Oncol., 18, 1182, 10.1016/S1470-2045(17)30422-9
Ansell, 2015, PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma, N. Engl. J. Med., 372, 311, 10.1056/NEJMoa1411087
Robert, 2015, Pembrolizumab versus ipilimumab in advanced melanoma, N. Engl. J. Med., 372, 2521, 10.1056/NEJMoa1503093
Schachter, 2017, Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006), Lancet, 390, 1853, 10.1016/S0140-6736(17)31601-X
Reck, 2016, Pembrolizumab versus chemotherapy for PD-L1–positive non–small-cell lung cancer, N. Engl. J. Med., 375, 1823, 10.1056/NEJMoa1606774
Chen, 2017, Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma, J. Clin. Oncol., 35, 2125, 10.1200/JCO.2016.72.1316
Chow, 2016, Antitumor activity of pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort, J. Clin. Oncol., 34, 3838, 10.1200/JCO.2016.68.1478
Bellmunt, 2017, Pembrolizumab as second-line therapy for advanced urothelial carcinoma, N. Engl. J. Med., 376, 1015, 10.1056/NEJMoa1613683
Fuchs, 2018, Safety and efficacy of pembrolizumab monotherapy in patients with previously treated advanced gastric and gastroesophageal junction cancer: Phase 2 Clinical KEYNOTE-059 Trial, JAMA Oncol., 4, 10.1001/jamaoncol.2018.0013
Prasad, 2018, Cancer drugs approved based on biomarkers and not tumor type—FDA approval of pembrolizumab for mismatch repair-deficient solid cancers, JAMA Oncol., 4, 157, 10.1001/jamaoncol.2017.4182
Rittmeyer, 2017, Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial, Lancet, 389, 255, 10.1016/S0140-6736(16)32517-X
Patel, 2018, Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial, Lancet Oncol., 19, 51, 10.1016/S1470-2045(17)30900-2
Antonia, 2017, Durvalumab after chemoradiotherapy in stage III non–small-cell lung cancer, N. Engl. J. Med., 377, 1919, 10.1056/NEJMoa1709937
Wolchok, 2017, Overall survival with combined nivolumab and ipilimumab in advanced melanoma, N. Engl. J. Med., 377, 1345, 10.1056/NEJMoa1709684
Motzer, 2018, Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma, N. Engl. J. Med., 378, 1277, 10.1056/NEJMoa1712126
Long, 2017, Standard-dose pembrolizumab in combination with reduced-dose ipilimumab for patients with advanced melanoma (KEYNOTE-029): an open-label, phase 1b trial, Lancet Oncol., 18, 1202, 10.1016/S1470-2045(17)30428-X
Antonia, 2016, Safety and antitumour activity of durvalumab plus tremelimumab in non-small cell lung cancer: a multicentre, phase 1b study, Lancet Oncol., 17, 299, 10.1016/S1470-2045(15)00544-6
Kumar, 2017, Current diagnosis and management of immune related adverse events (irAEs) induced by immune checkpoint inhibitor therapy, Front. Pharmacol., 8, 49, 10.3389/fphar.2017.00049
Larkin, 2015, Combined nivolumab and ipilimumab or monotherapy in untreated melanoma, N. Engl. J. Med., 373, 23, 10.1056/NEJMoa1504030
Nishijima, 2017, Safety and tolerability of PD-1/PD-L1 inhibitors compared with chemotherapy in patients with advanced cancer: a meta-analysis, Oncologist, 22, 470, 10.1634/theoncologist.2016-0419
Man, 2018, Treatment-related toxicities of immune checkpoint inhibitors in advanced cancers: a meta-analysis, Asia Pac. J. Clin. Oncol., 10.1111/ajco.12838
Pitt, 2016, Resistance mechanisms to immune-checkpoint blockade in cancer: tumor-intrinsic and -extrinsic factors, Immunity, 44, 1255, 10.1016/j.immuni.2016.06.001
Kvistborg, 2014, Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response, Sci. Transl. Med., 6, 10.1126/scitranslmed.3008918
Ayers, 2017, IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade, J. Clin. Invest., 127, 2930, 10.1172/JCI91190
Tumeh, 2014, PD-1 blockade induces responses by inhibiting adaptive immune resistance, Nature, 515, 568, 10.1038/nature13954
Hargadon, 2017, Strategies to improve the efficacy of dendritic cell-based immunotherapy for melanoma, Front. Immunol., 8, 1594, 10.3389/fimmu.2017.01594
Daud, 2016, Tumor immune profiling predicts response to anti–PD-1 therapy in human melanoma, J. Clin. Invest., 126, 3447, 10.1172/JCI87324
Topalian, 2012, Safety, activity, and immune correlates of anti–PD-1 antibody in cancer, N. Engl. J. Med., 366, 2443, 10.1056/NEJMoa1200690
Rizvi, 2015, Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer, Science, 348, 124, 10.1126/science.aaa1348
McGranahan, 2016, Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade, Science, 351, 1463, 10.1126/science.aaf1490
Routy, 2018, Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors, Science, 359, 91, 10.1126/science.aan3706
Ahn, 2016, Demethylation of the PD-1 promoter is imprinted during the effector phase of CD8 T cell exhaustion, J. Virol., 90, 8934, 10.1128/JVI.00798-16
Pauken, 2016, Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade, Science, 354, 1160, 10.1126/science.aaf2807
Blackburn, 2008, Selective expansion of a subset of exhausted CD8 T cells by PD-L1 blockade, Proc. Natl. Acad. Sci., 105, 15016, 10.1073/pnas.0801497105
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
Zaretsky, 2016, Mutations associated with acquired resistance to PD-1 blockade in melanoma, N. Engl. J. Med., 375, 819, 10.1056/NEJMoa1604958
Hargadon, 2016, Dysregulation of TGFβ1 activity in cancer and its influence on the quality of anti-tumor immunity, J. Clin. Med., 5, E76, 10.3390/jcm5090076