Resistance to immune checkpoint inhibitors in non-small cell lung cancer: biomarkers and therapeutic strategies

Robert J Walsh, Ross A Soo, Robert J Walsh, Ross A Soo

Abstract

The treatment landscape for patients with advanced non-small cell lung cancer has evolved greatly with the advent of immune checkpoint inhibitors. However, many patients do not derive benefit from checkpoint blockade, developing either primary or secondary resistance, highlighting a need for alternative approaches to modulate immune function. In this review, we highlight the absence of a common definition of primary and secondary resistance and summarize their frequency and clinical characteristics. Furthermore, we provide an overview of the biomarkers and mechanisms of resistance involving the tumor, the tumor microenvironment and the host, and suggest treatment strategies to overcome these mechanisms and improve clinical outcomes.

Keywords: PD-L1; acquired resistance; immune checkpoint inhibitors; primary resistance.

Conflict of interest statement

Conflict of interest statement: RAS has received honoraria from Astra-Zeneca, BMS, Boehringer Ingelheim, Celgene, Lilly, Merck, Novartis, Pfizer, Roche, Taiho, Takeda, and Yuhan; and research funding from Astra-Zeneca and Boehringer Ingelheim. RJW reports no conflict of interest.

© The Author(s), 2020.

Figures

Figure 1.
Figure 1.
A spider plot representing examples of resistance to immune checkpoint inhibition with (P) primary resistance, defined as best response being disease progression; (AR1) acquired resistance, defined as initial stable disease and subsequent disease progression; and (AR2) acquired resistance, defined as initial response and subsequent disease progression.
Figure 2.
Figure 2.
Biomarkers of primary and acquired resistance occurring in the (A) tumor microenvironment (TME), (B) in the tumor, and (C) host factors. Within the TME, factors involved in primary resistance (blue font) includes the presence of (a) immunosuppressive cells including cancer associated fibroblasts (CAFs), myeloid derived suppressor cells (MDSCs), regulatory T cells (T reg), M2 macrophages, and reduced tumor infiltrating lymphocytes (TILs), (b) immune-suppressive molecules such as indoleamine 2,3-dioxygenase (IDO), adenosine, vascular endothelial growth factor (VEGF), glucose, and (c) tumoral factors such as reduced PD-L1 expression, tumor mutation burden (TMB), intra-tumoral heterogeneity (ITH), genetic loss of HLA class I, dysregulated IFN/JAK pathway, aberrant oncologic signaling pathways (PTEN loss, mutations in WNT/b-catenin, LKB1, c-myc). Biomarkers associated with acquired resistance (red font) include (d) loss of B2m and MHC-I, neoantigen evolution with loss of neoepitopes; the IFN/JAK escape pathway with loss of function JAK-1 and JAK-2 mutations and (e) the upregulation of other immune checkpoints such as T-cell immunoglobulin, mucin domain-3 protein (TIM-3), lymphocyte-activation gene 3 (LAG-3), B and T lymphocyte attenuator (BTLA), T-cell immunoreceptor tyrosine-based inhibition motif domain (TIGIT), and V-domain immunoglobulin-containing suppressor of T-cell activation (VISTA). Host factors affecting resistance includes (e) gut microbiome and antibiotic use.

References

    1. Kazandjian D, Keegan P, Suzman DL, et al. Characterization of outcomes in patients with metastatic non-small cell lung cancer treated with programmed cell death protein 1 inhibitors past RECIST version 1.1-defined disease progression in clinical trials. Semin Oncol 2017; 44: 3–7.
    1. Ricciuti B, Genova C, Bassanelli M, et al. Safety and efficacy of nivolumab in patients with advanced non-small-cell lung cancer treated beyond progression. Clin Lung Cancer 2019; 20: 178–185.e172.
    1. Gandara DR, von Pawel J, Mazieres J, et al. Atezolizumab treatment beyond progression in advanced NSCLC: results from the randomized, phase III OAK study. J Thorac Oncol 2018; 13: 1906–1918.
    1. Kim C, Hoang CD, Kesarwala AH, et al. Role of local ablative therapy in patients with oligometastatic and oligoprogressive non-small cell lung cancer. J Thorac Oncol 2017; 12: 179–193.
    1. Ning MS, Gomez DR, Heymach JV, et al. Stereotactic ablative body radiation for oligometastatic and oligoprogressive disease. Transl Lung Cancer Res 2019; 8: 97–106.
    1. Laurie SA, Banerji S, Blais N, et al. Canadian consensus: oligoprogressive, pseudoprogressive, and oligometastatic non-small-cell lung cancer. Curr Oncol 2019; 26: e81–e93.
    1. Tumati V, Iyengar P. The current state of oligometastatic and oligoprogressive non-small cell lung cancer. J Thorac Dis 2018; 10: S2537–S2544.
    1. Chen Daniel S, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 2013; 39: 1–10.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252–264.
    1. Low JL, Walsh RJ, Ang Y, et al. The evolving immuno-oncology landscape in advanced lung cancer: first-line treatment of non-small cell lung cancer. Ther Adv Med Oncol 2019; 11: 1758835919870360.
    1. Doroshow DB, Sanmamed MF, Hastings K, et al. Immunotherapy in non-small cell lung cancer: facts and hopes. Clin Cancer Res 2019; 25: 4592–4602.
    1. Syn NL, Teng MWL, Mok TSK, et al. De-novo and acquired resistance to immune checkpoint targeting. Lancet Oncol 2017; 18: e731–e741.
    1. Jenkins RW, Barbie DA, Flaherty KT. Mechanisms of resistance to immune checkpoint inhibitors. Br J Cancer 2018; 118: 9–16.
    1. Shah S, Wood K, Labadie B, et al. Clinical and molecular features of innate and acquired resistance to anti-PD-1/PD-L1 therapy in lung cancer. Oncotarget 2017; 9: 4375–4384.
    1. Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med 2016; 375: 1823–1833.
    1. Mok TSK, Wu Y-L, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial. Lancet 2019; 393: 1819–1830.
    1. Carbone DP, Reck M, Paz-Ares L, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med 2017; 376: 2415–2426.
    1. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 2018; 378: 2078–2092.
    1. Paz-Ares L, Luft A, Vicente D, et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N Engl J Med 2018; 379: 2040–2051.
    1. West H, McCleod M, Hussein M, et al. Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 2019; 20: 924–937.
    1. Socinski MA, Koynov KD, Berard H, et al. LBA65 IMpower131: progression-free survival (PFS) and overall survival (OS) analysis of a randomised phase III study of atezolizumab + carboplatin + paclitaxel or nab-paclitaxel vs carboplatin + nab-paclitaxel in 1L advanced squamous NSCLC. Ann Oncol 2018; 29(Suppl. 8): mdy424-077.
    1. Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med 2018; 378: 2288–2301.
    1. Hellmann MD, Ciuleanu T-E, Pluzanski A, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med 2018; 378: 2093–2104.
    1. Rizvi NA, Chul Cho B, Reinmuth N, et al. LBA6 Durvalumab with or without tremelimumab vs platinum-based chemotherapy as first-line treatment for metastatic non-small cell lung cancer: MYSTIC. Ann Oncol 2018; 29(Suppl. 10): mdy511-005.
    1. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015; 373: 123–135.
    1. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015; 373: 1627–1639.
    1. Herbst RS, Baas P, Kim D-W, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016; 387: 1540–1550.
    1. Rittmeyer A, Barlesi F, Waterkamp D, 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. Lancet 2017; 389: 255–265.
    1. Gettinger SN, Wurtz A, Goldberg SB, et al. Clinical features and management of acquired resistance to PD-1 axis inhibitors in 26 patients with advanced non-small cell lung cancer. J Thorac Oncol 2018; 13: 831–839.
    1. Chiou VL, Burotto M. Pseudoprogression and immune-related response in solid tumors. J Clin Oncol 2015; 33: 3541–3543.
    1. Hodi FS, Ballinger M, Lyons B, et al. Immune-modified response evaluation criteria in solid tumors (imRECIST): refining guidelines to assess the clinical benefit of cancer immunotherapy. J Clin Oncol 2018; 36: 850–858.
    1. Tazdait M, Mezquita L, Lahmar J, et al. Patterns of responses in metastatic NSCLC during PD-1 or PDL-1 inhibitor therapy: comparison of RECIST 1.1, irRECIST and iRECIST criteria. Eur J Cancer 2018; 88: 38–47.
    1. Nishino M, Giobbie-Hurder A, Gargano M, et al. Developing a common language for tumor response to immunotherapy: immune-related response criteria using unidimensional measurements. Clin Cancer Res 2013; 19: 3936–3943.
    1. Borcoman E, Nandikolla A, Long G, et al. Patterns of response and progression to immunotherapy. Am Soc Clin Oncol Educ Book 2018; 38: 169–178.
    1. Antonia SJ, Borghaei H, Ramalingam SS, et al. Four-year survival with nivolumab in patients with previously treated advanced non-small-cell lung cancer: a pooled analysis. Lancet Oncol 2019; 20: 1395–1408.
    1. Topalian SL, Taube JM, Anders RA, et al. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer 2016; 16: 275.
    1. Shen X, Zhao B. Efficacy of PD-1 or PD-L1 inhibitors and PD-L1 expression status in cancer: meta-analysis. BMJ 2018; 362: k3529.
    1. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015; 348: 124–128.
    1. Gandara DR, Paul SM, Kowanetz M, et al. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med 2018; 24: 1441–1448.
    1. Kim ES, Velcheti V, Mekhail T, et al. LBA55 primary efficacy results from B-F1RST, a prospective phase II trial evaluating blood-based tumour mutational burden (bTMB) as a predictive biomarker for atezolizumab (atezo) in 1L non-small cell lung cancer (NSCLC). Ann Oncol 2018; 29(Suppl. 8): mdy424-067.
    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med 2017; 377: 2500–2501.
    1. Hellmann MD, Nathanson T, Rizvi H, et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell 2018; 33: 843–852.e844.
    1. McGranahan N, Furness AJS, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016; 351: 1463–1469.
    1. Schalper KA, Brown J, Carvajal-Hausdorf D, et al. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J Natl Cancer Inst 2015; 107: dju435.
    1. Soo RA, Chen Z, Yan Teng RS, et al. Prognostic significance of immune cells in non-small cell lung cancer: meta-analysis. Oncotarget 2018; 9: 24801–24820.
    1. Herbst RS, Soria J-C, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014; 515: 563–567.
    1. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014; 515: 568–571.
    1. Perea F, Sánchez-Palencia A, Gómez-Morales M, et al. HLA class I loss and PD-L1 expression in lung cancer: impact on T-cell infiltration and immune escape. Oncotarget 2017; 9: 4120–4133.
    1. Chowell D, Morris LGT, Grigg CM, et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018; 359: 582–587.
    1. Negrao MV, Lam VK, Reuben A, et al. PD-L1 expression, tumor mutational burden, and cancer gene mutations are stronger predictors of benefit from immune checkpoint blockade than HLA class I genotype in non-small cell lung cancer. J Thorac Oncol 2019; 14: 1021–1031.
    1. Shukla SA, Rooney MS, Rajasagi M, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol 2015; 33: 1152–1158.
    1. McGranahan N, Rosenthal R, Hiley CT, et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 2017; 171: 1259–1271.e1211.
    1. Restifo NP, Marincola FM, Kawakami Y, et al. Loss of functional beta 2-microglobulin in metastatic melanomas from five patients receiving immunotherapy. J Natl Cancer Inst 1996; 88: 100–108.
    1. Zaretsky JM, Garcia-Diaz A, Shin DS, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med 2016; 375: 819–829.
    1. Anagnostou V, Smith KN, Forde PM, et al. Evolution of neoantigen landscape during immune checkpoint blockade in non-small cell lung cancer. Cancer Discov 2017; 7: 264–276.
    1. Shin DS, Zaretsky JM, Escuin-Ordinas H, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov 2017; 7: 188–201.
    1. Gao J, Shi LZ, Zhao H, et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 2016; 167: 397–404.e399.
    1. Li SD, Ma M, Li H, et al. Cancer gene profiling in non-small cell lung cancers reveals activating mutations in JAK2 and JAK3 with therapeutic implications. Genome Med 2017; 9: 89.
    1. Spranger S, Gajewski TF. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat Rev Cancer 2018; 18: 139–147.
    1. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015; 523: 231.
    1. Luke JJ, Bao R, Sweis RF, et al. WNT/β-catenin pathway activation correlates with immune exclusion across human cancers. Clin Cancer Res 2019; 25: 3074–3083.
    1. Peng W, Chen JQ, Liu C, et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy. Cancer Discov 2016; 6: 202–216.
    1. George S, Miao D, Demetri GD, et al. Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma. Immunity 2017; 46: 197–204.
    1. Roh W, Chen P-L, Reuben A, et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci Transl Med 2017; 9: eaah3560.
    1. Koyama S, Akbay EA, Li YY, et al. STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment. Cancer Res 2016; 76: 999–1008.
    1. Biton J, Mansuet-Lupo A, Pécuchet N, et al. TP53, STK11, and EGFR mutations predict tumor immune profile and the response to anti-PD-1 in lung adenocarcinoma. Clin Cancer Res 2018; 24: 5710–5723.
    1. Dong Z-Y, Zhong W-Z, Zhang X-C, et al. Potential predictive value of TP53 and KRAS mutation status for response to PD-1 blockade immunotherapy in lung adenocarcinoma. Clin Cancer Res 2017; 23: 3012–3024.
    1. Assoun S, Theou-Anton N, Nguenang M, et al. Association of TP53 mutations with response and longer survival under immune checkpoint inhibitors in advanced non-small-cell lung cancer. Lung Cancer 2019; 132: 65–71.
    1. Lisberg A, Cummings A, Goldman JW, et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor naïve patients with advanced NSCLC. J Thorac Oncol 2018; 13: 1138–1145.
    1. Gainor JF, Shaw AT, Sequist LV, et al. EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in non-small cell lung cancer: a retrospective analysis. Clin Cancer Res 2016; 22: 4585–4593.
    1. Lee CK, Man J, Lord S, et al. Clinical and molecular characteristics associated with survival among patients treated with checkpoint inhibitors for advanced non-small cell lung carcinoma: a systematic review and meta-analysis. JAMA Oncol 2018; 4: 210–216.
    1. Soo RA, Lim SM, Syn NL, et al. Immune checkpoint inhibitors in epidermal growth factor receptor mutant non-small cell lung cancer: current controversies and future directions. Lung Cancer 2018; 115: 12–20.
    1. Riera-Domingo C, Audigé A, Granja S, et al. Immunity, hypoxia, and metabolism–the Ménage à Trois of cancer: implications for immunotherapy. Physiol Rev 2020; 100: 1–102.
    1. Damgaci S, Ibrahim-Hashim A, Enriquez-Navas PM, et al. Hypoxia and acidosis: immune suppressors and therapeutic targets. Immunology 2018; 154: 354–362.
    1. Huber V, Camisaschi C, Berzi A, et al. Cancer acidity: an ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol 2017; 43: 74–89.
    1. Erra Díaz F, Dantas E, Geffner J. Unravelling the interplay between extracellular acidosis and immune cells. Mediators Inflamm 2018; 2018: 1218297.
    1. Noman MZ, Janji B, Berchem G, et al. Hypoxia-induced autophagy. Autophagy 2012; 8: 704–706.
    1. Jeong H, Kim S, Hong BJ, et al. Tumor-associated macrophages enhance tumor hypoxia and aerobic glycolysis. Cancer Res 2019; 79: 795–806.
    1. Yuan J, Zhou J, Dong Z, et al. Pretreatment serum VEGF is associated with clinical response and overall survival in advanced melanoma patients treated with ipilimumab. Cancer Immunol Res 2014; 2: 127–132.
    1. Chen P-L, Roh W, Reuben A, et al. Analysis of immune signatures in longitudinal tumor samples yields insight into biomarkers of response and mechanisms of resistance to immune checkpoint blockade. Cancer Discov 2016; 6: 827–837.
    1. Sabatino M, Kim-Schulze S, Panelli MC, et al. Serum vascular endothelial growth factor and fibronectin predict clinical response to high-dose interleukin-2 therapy. J Clin Oncol 2009; 27: 2645–2652.
    1. Wang J, Chen J, Guo Y, et al. Strategies targeting angiogenesis in advanced non-small cell lung cancer. Oncotarget 2017; 8: 53854–53872.
    1. Chen DS, Hurwitz H. Combinations of bevacizumab with cancer immunotherapy. Cancer J 2018; 24: 193–204.
    1. Voron T, Colussi O, Marcheteau E, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med 2015; 212: 139–148.
    1. Shiono A, Kaira K, Mouri A, et al. Improved efficacy of ramucirumab plus docetaxel after nivolumab failure in previously treated non-small cell lung cancer patients. Thorac Cancer 2019; 10: 775–781.
    1. Brochez L, Chevolet I, Kruse V. The rationale of indoleamine 2,3-dioxygenase inhibition for cancer therapy. Eur J Cancer 2017; 76: 167–182.
    1. Botticelli A, Cerbelli B, Lionetto L, et al. Can IDO activity predict primary resistance to anti-PD-1 treatment in NSCLC? J Transl Med 2018; 16: 219.
    1. Shayan G, Srivastava R, Li J, et al. Adaptive resistance to anti-PD1 therapy by Tim-3 upregulation is mediated by the PI3K-Akt pathway in head and neck cancer. Oncoimmunology 2016; 6: e1261779.
    1. Huang R-Y, Francois A, McGray AR, et al. Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. Oncoimmunology 2016; 6: e1249561.
    1. Koyama S, Akbay EA, Li YY, et al. Adaptive resistance to therapeutic PD-1 blockade is associated with upregulation of alternative immune checkpoints. Nat Commun 2016; 7: 10501.
    1. Gao J, Ward JF, Pettaway CA, et al. VISTA is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med 2017; 23: 551–555.
    1. Thommen DS, Schreiner J, Müller P, et al. Progression of lung cancer is associated with increased dysfunction of T cells defined by coexpression of multiple inhibitory receptors. Cancer Immunol Res 2015; 3: 1344–1355.
    1. Vétizou M, Pitt JM, Daillère R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015; 350: 1079–1084.
    1. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018; 359: 97–103.
    1. Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015; 350: 1084–1089.
    1. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018; 359: 91–97.
    1. Derosa L, Hellmann MD, Spaziano M, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann Oncol 2018; 29: 1437–1444.
    1. Pinato DJ, Howlett S, Ottaviani D, et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol 2019; 5: 1774–1778.
    1. Elkrief A, El Raichani L, Richard C, et al. Antibiotics are associated with decreased progression-free survival of advanced melanoma patients treated with immune checkpoint inhibitors. Oncoimmunology 2019; 8: e1568812.
    1. Frankel AE, Coughlin LA, Kim J, et al. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia 2017; 19: 848–855.
    1. Schvartsman G, Peng SA, Bis G, et al. Response rates to single-agent chemotherapy after exposure to immune checkpoint inhibitors in advanced non-small cell lung cancer. Lung Cancer 2017; 112: 90–95.
    1. Park SE, Lee SH, Ahn JS, et al. Increased response rates to salvage chemotherapy administered after PD-1/PD-L1 inhibitors in patients with non-small cell lung cancer. J Thorac Oncol 2018; 13: 106–111.
    1. Grigg C, Reuland BD, Sacher AG, et al. Clinical outcomes of patients with non-small cell lung cancer (NSCLC) receiving chemotherapy after immune checkpoint blockade. J Clin Oncol 2017; 35: 9082.
    1. Leger PD, Rothschild S, Castellanos E, et al. Response to salvage chemotherapy following exposure to immune checkpoint inhibitors in patients with non-small cell lung cancer. J Clin Oncol 2017; 35: 9084.
    1. Grohe C, Gleiber W, Haas S, et al. 119O Efficacy and safety of nintedanib + docetaxel in lung adenocarcinoma patients (pts) following treatment with immune checkpoint inhibitors (ICIs): first results of the ongoing non-interventional study (NIS) VARGADO. Ann Oncol 2019; 30.
    1. Corral J, Majem M, Rodríguez-Abreu D, et al. Efficacy of nintedanib and docetaxel in patients with advanced lung adenocarcinoma treated with first-line chemotherapy and second-line immunotherapy in the nintedanib NPU program. Clin Transl Oncol 2019; 21: 1270–1279.
    1. Capelletto E, Osman G, Morabito A, et al. NSCLC survival expectancy for patients treated with docetaxel/nintedanib in the SENECA trial and previous immunotherapy. WCLC 2019. Abstract.
    1. Molife C, Hess LM, Cui ZL, et al. Sequential therapy with ramucirumab and/or checkpoint inhibitors for non-small-cell lung cancer in routine practice. Future Oncol 2019; 15: 2915–2931.
    1. Reck M, Kaiser R, Mellemgaard A, et al. Docetaxel plus nintedanib versus docetaxel plus placebo in patients with previously treated non-small-cell lung cancer (LUME-Lung 1): a phase 3, double-blind, randomised controlled trial. Lancet Oncol 2014; 15: 143–155.
    1. Garon EB, Ciuleanu T-E, Arrieta O, et al. Ramucirumab plus docetaxel versus placebo plus docetaxel for second-line treatment of stage IV non-small-cell lung cancer after disease progression on platinum-based therapy (REVEL): a multicentre, double-blind, randomised phase 3 trial. Lancet 2014; 384: 665–673.
    1. Mazzone R, Zwergel C, Artico M, et al. The emerging role of epigenetics in human autoimmune disorders. Clin Epigenetics 2019; 11: 34.
    1. Orillion A, Hashimoto A, Damayanti N, et al. Entinostat neutralizes myeloid-derived suppressor cells and enhances the antitumor effect of PD-1 inhibition in murine models of lung and renal cell carcinoma. Clin Cancer Res 2017; 23: 5187–5201.
    1. Hellmann M, Jänne P, Opyrchal M, et al. OA05.01 efficacy/safety of entinostat (ENT) and pembrolizumab (PEMBRO) in NSCLC patients previously treated with anti-PD-(L)1 therapy. J Thorac Oncol 2018; 13: S330.
    1. Ramalingam SS, Hellmann MD, Awad MM, et al. Abstract CT078: tumor mutational burden (TMB) as a biomarker for clinical benefit from dual immune checkpoint blockade with nivolumab (nivo) + ipilimumab (ipi) in first-line (1L) non-small cell lung cancer (NSCLC): identification of TMB cutoff from CheckMate 568. Cancer Res 2018; 78: CT078.
    1. Theelen WSME, Peulen HMU, Lalezari F, et al. Effect of pembrolizumab after stereotactic body radiotherapy vs pembrolizumab alone on tumor response in patients with advanced non-small cell lung cancer: results of the PEMBRO-RT phase 2 randomized clinical trial. JAMA Oncol 2019; 5: 1276–1282.
    1. van Willigen WW, Bloemendal M, Gerritsen WR, et al. Dendritic cell cancer therapy: vaccinating the right patient at the right time. Front Immunol 2018; 9: 2265.
    1. Ott PA, Govindan R, Naing A, et al. 1127O A personal neoantigen vaccine, NEO-PV-01, with anti-PD1 induces broad de novo anti-tumor immunity in patients with metastatic melanoma, NSCLC, and bladder cancer. Ann Oncol 2018; 29(Suppl. 8): mdy288.
    1. Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov 2015; 14: 642.
    1. Pol J, Kroemer G, Galluzzi L. First oncolytic virus approved for melanoma immunotherapy. Oncoimmunology 2015; 5: e1115641.
    1. Rudin CM, Pandha HS, Gupta S, et al. LBA40 Phase Ib KEYNOTE-200: a study of an intravenously delivered oncolytic virus, coxsackievirus A21 in combination with pembrolizumab in advanced NSCLC and bladder cancer patients. Ann Oncol 2018; 29(Suppl. 8): mdy424-050.
    1. Flood BA, Higgs EF, Li S, et al. STING pathway agonism as a cancer therapeutic. Immunol Rev 2019; 290: 24–38.
    1. Harrington KJ, Brody J, Ingham M, et al. LBA15 Preliminary results of the first-in-human (FIH) study of MK-1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumors or lymphomas. Ann Oncol 2018; 29(Suppl. 8): mdy424-015.
    1. Ribas A, Lawrence D, Atkinson V, et al. Combined BRAF and MEK inhibition with PD-1 blockade immunotherapy in BRAF-mutant melanoma. Nat Med 2019; 25: 936–940.
    1. Ascierto PA, Ferrucci PF, Fisher R, et al. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat Med 2019; 25: 941–946.
    1. Sullivan RJ, Hamid O, Gonzalez R, et al. Atezolizumab plus cobimetinib and vemurafenib in BRAF-mutated melanoma patients. Nat Med 2019; 25: 929–935.
    1. Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med 2019; 380: 1116–1127.
    1. Motzer RJ, Penkov K, Haanen J, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med 2019; 380: 1103–1115.
    1. Yang JC-H, Gadgeel SM, Sequist LV, et al. Pembrolizumab in combination with erlotinib or gefitinib as first-line therapy for advanced NSCLC with sensitizing EGFR mutation. J Thorac Oncol 2019; 14: 553–559.
    1. Gettinger S, Hellmann MD, Chow LQM, et al. Nivolumab plus erlotinib in patients with EGFR-mutant advanced NSCLC. J Thorac Oncol 2018; 13: 1363–1372.
    1. Fukumura D, Kloepper J, Amoozgar Z, et al. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol 2018; 15: 325–340.
    1. Du W, Huang H, Sorrelle N, et al. Sitravatinib potentiates immune checkpoint blockade in refractory cancer models. JCI Insight 2018; 3: e124184.
    1. He K. Preliminary biomarker analysis of sitravatinib in combination with nivolumab in NSCLC patients progressing on prior checkpoint inhibitor. 33rd Annual meeting & pre-conference programs of the Society for Immunotherapy of Cancer (SITC 2018). J ImmunoTher Cancer 2018; 6: 114.
    1. Brose MS, Vogelzang NJ, DiSimone C, et al. A phase Ib/II trial of lenvatinib plus pembrolizumab in non-small cell lung cancer. J Clin Oncol 2019; 37: 16.
    1. Weiskopf K. Cancer immunotherapy targeting the CD47/SIRPα axis. Eur J Cancer 2017; 76: 100–109.
    1. Kauder SE, Kuo TC, Harrabi O, et al. ALX148 blocks CD47 and enhances innate and adaptive antitumor immunity with a favorable safety profile. PLoS One 2018; 13: e0201832.
    1. Chow LQM, Gainor JF, Lakhani NJ, et al. A phase I study of ALX148, a CD47 blocker, in combination with established anticancer antibodies in patients with advanced malignancy. J Clin Oncol 2019; 37: 2514.
    1. Sanmamed MF, Pastor F, Rodriguez A, et al. Agonists of co-stimulation in cancer immunotherapy directed against CD137, OX40, GITR, CD27, CD28, and ICOS. Semin Oncol 2015; 42: 640–655.
    1. Zappasodi R, Sirard C, Li Y, et al. Rational design of anti-GITR-based combination immunotherapy. Nat Med 2019; 25: 759–766.
    1. Papadopoulos KP, Autio KA, Golan T, et al. Phase 1 study of MK-4166, an anti-human glucocorticoid-induced tumor necrosis factor receptor (GITR) antibody, as monotherapy or with pembrolizumab (pembro) in patients (pts) with advanced solid tumors. J Clin Oncol 2019; 37: 9509.
    1. Hellmann MD, Paz-Ares L, Bernabe Caro R, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. N Engl J Med 2019; 381: 2020–2031.
    1. Anderson AC, Joller N, Kuchroo VK. Lag-3, Tim-3, and TIGIT: co-inhibitory receptors with specialized functions in immune regulation. Immunity 2016; 44: 989–1004.
    1. Ascierto PA, Bono P, Bhatia S, et al. LBA18 Efficacy of BMS-986016, a monoclonal antibody that targets lymphocyte activation gene-3 (LAG-3), in combination with nivolumab in pts with melanoma who progressed during prior anti-PD-1/PD-L1 therapy (mel prior IO) in all-comer and biomarker-enriched populations. Ann Oncol 2017; 28(Suppl. 5).
    1. Hong DS, Schoffski P, Calvo A, et al. Phase I/II study of LAG525 ± spartalizumab (PDR001) in patients (pts) with advanced malignancies. J Clin Oncol 2018; 36: 3012.
    1. Davar D. A phase 1 study of TSR-022, an anti-TIM-3 monoclonal antibody, in combination with TSR-042 (anti-PD-1) in patients with colorectal cancer and post-PD-1 NSCLC and melanoma. 33rd Annual meeting & pre-conference programs of the Society for Immunotherapy of Cancer (SITC 2018) J ImmunoTher Cancer 2018; 6: 114.
    1. Riaz N, Havel JJ, Makarov V, et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell 2017; 171: 934–949.e916.
    1. Tang J, Shalabi A, Hubbard-Lucey VM. Comprehensive analysis of the clinical immuno-oncology landscape. Ann Oncol 2017; 29: 84–91.
    1. Smoragiewicz M, Bogaerts J, Calvo E, et al. Design and conduct of early clinical studies of immunotherapy agent combinations: recommendations from the task force on methodology for the development of innovative cancer therapies. Ann Oncol 2018; 29: 2175–2182.
    1. Shin SH, Bode AM, Dong Z. Addressing the challenges of applying precision oncology. NPJ Precis Oncol 2017; 1: 28.
    1. Kim JM, Chen DS. Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure). Ann Oncol 2016; 27: 1492–1504.
    1. Jerby-Arnon L, Shah P, Cuoco MS, et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 2018; 175: 984–997.e924.
    1. Hugo W, Zaretsky JM, Sun L, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016; 165: 35–44.

Source: PubMed

3
Abonnere