Randomized, open-label, phase 2 study of andecaliximab plus nivolumab versus nivolumab alone in advanced gastric cancer identifies biomarkers associated with survival

Manish A Shah, David Cunningham, Jean-Philippe Metges, Eric Van Cutsem, Zev Wainberg, Emon Elboudwarej, Kai-Wen Lin, Scott Turner, Marianna Zavodovskaya, David Inzunza, Jinfeng Liu, Scott D Patterson, Jingzhu Zhou, Jing He, Dung Thai, Pankaj Bhargava, Carrie Baker Brachmann, Daniel V T Cantenacci, Manish A Shah, David Cunningham, Jean-Philippe Metges, Eric Van Cutsem, Zev Wainberg, Emon Elboudwarej, Kai-Wen Lin, Scott Turner, Marianna Zavodovskaya, David Inzunza, Jinfeng Liu, Scott D Patterson, Jingzhu Zhou, Jing He, Dung Thai, Pankaj Bhargava, Carrie Baker Brachmann, Daniel V T Cantenacci

Abstract

Background: Matrix metalloproteinase-9 (MMP9) selectively cleaves extracellular matrix proteins contributing to tumor growth and an immunosuppressive microenvironment. This study evaluated andecaliximab (ADX), an inhibitor of MMP9, in combination with nivolumab (NIVO), for the treatment of advanced gastric cancer.

Methods: Phase 2, open-label, randomized multicenter study evaluating the efficacy, safety, and pharmacodynamics of ADX+NIVO versus NIVO in patients with pretreated metastatic gastric or gastroesophageal junction (GEJ) adenocarcinoma. The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS), overall survival (OS), and adverse events (AEs). We explored the correlation of efficacy outcomes with biomarkers.

Results: 144 patients were randomized; 141 were treated: 81% white, 69% male, median age was 61 years in the ADX+NIVO group and 62 years in the NIVO-alone group. The ORR was 10% (95% CI 4 to 19) in the ADX+NIVO group and 7% (95% CI 2 to 16) in the NIVO-alone group (OR: 1.5 (95% CI 0.4 to 6.1; p=0.8)). There was no response or survival benefit associated with adding ADX. AE rates were comparable in both treatment groups; the most common AEs were fatigue, decreased appetite, nausea, and vomiting. Programmed cell death ligand 1, interferon-γ (IFN), and intratumoral CD8+ cell density were not associated with treatment response or survival. The gene signature most correlated with shorter survival was the epithelial-to-mesenchymal gene signature; high transforming growth factor (TGF)-β fibrosis score was negatively associated with OS (p=0.036). Gene expression analysis of baseline tumors comparing long-(1+ years) and short-term (<1 year) survivors showed that GRB7 was associated with survival beyond 1 year. Human epidermal growth factor receptor 2 (HER2)-positive disease was associated with significantly longer survival (p=0.0077). Median tumor mutation burden (TMB) was 2.01; patients with TMB ≥median had longer survival (p=0.0025) and improved PFS (p=0.016). Based on a model accounting for TMB, TGF-β fibrosis, and HER2, TMB was the main driver of survival in this patient population.

Conclusion: Combination of ADX+NIVO had a favorable safety profile but did not improve efficacy compared with NIVO alone in patients with pretreated metastatic gastric or GEJ adenocarcinoma. HER2 positivity, higher TMB or GRB7, and lower TGF-β were associated with improved outcomes.

Trial registration number: NCT02864381 or GS-US-296--2013.

Keywords: Biomarkers; Clinical Trials; Phase II as Topic; Tumor; antibodies; neoplasm.

Conflict of interest statement

Competing interests: MAS has received institutional funding from Bristol Meyers Squibb, Oncolys Biopharma, and Merck, DCu has received grants from MedImmune, Clovis, Eli Lilly, 4SC, Bayer, Celgene, Leap, and Roche, and has participated in a scientific advisory board for OVIBIO. J-PM has received honoraria from Merck Sharp & Dohme. J-PM, reports honoraria from MSD, EVC reports grants from Amgen, Bayer, Boehringer Ingelheim, Bristol-Meyers Squibb, Celgene, Ipsen, Lily, Merck Sharp & Dohme, Merck KGaA, Novartis, Roche, and Servier, and has served as a consultant for Array, Astellas, AstraZeneca, Bayer, Beigene, Biocartis, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Daiichi, Halozyme, GSK, Incyte, Ipsen, Lilly, Merck Sharp & Dohme, Merck KGaA, Novartis, Pierre Fabre, Roche, Servier, Sirtex, Taiho. ZW reports research support from BMS, Gilead Sciences, Novartis, Plexxikon, and has served as a consultant for AstraZeneca, Bayer, Daiichi, Five Prime, Gilead, Lily, Macrogenic, and Merck. MZ has nothing to disclose. DI has nothing to disclose. JL is an employee of Gilead Sciences, and owns stock in Gilead Sciences, and Roche. SDP is an employee of Gilead Sciences, Inc., and owns stock in Gilead Science, and Amgen. CBB, DT, EE, JH, JZ, K-WL, PB, ST are employees of and own stock in Gilead Sciences. DCa has received consulting fees from Genentech, Roche, Eli Lilly, Merck, Daiichi Sankyo, BMS, Ono Pharma USA, Five Prime, Seattle Genetics, Amgen, Taiho Pharmaceutical, Astellas Pharma, Gritstone Bio, Pieris Pharmaceutical, Zymeworks, Basilea Pharmaceutica, QED Group, Arcus Biosciences, Foundation Medicine, Pierian Biosciences, Silverback Therapeutics, Servier Pharmaceuticals, Blueprint Medicines, Tempus, Guardant Health, Archer Biosciences, and Natera, and has received honoraria from Genentech, Roche, Eli Lilly, Merck, Daiichi Sankyo, AstraZeneca, Tempus, and Guardant Health.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
OS (A) and PFS (B) by treatment arm and change from baseline in CD8+ cell density (Wilcoxon signed-rank test) (C) and gene signature scores (D). (A)*Since the primary endpoint was not met, OS was not analyzed per protocol but for exploratory purposes. (B)*Since the primary endpoint was not met, PFS was not analyzed per protocol but for exploratory purposes. BL, baseline; CR, complete response; IFN, interferon; n.s., not significant; OS, overall survival; PD-L1, programmed death ligand 1; PFS, progression-free survival; PR, partial response; TC+IC, tumor and associated immune cell-positive.
Figure 2
Figure 2
OS (A) and PFS (B) of total treated population, overall survival in all treated patients with PD-L1 (TC+IC)-positive tumors (C) and PD-L1 (TC-only)-positive tumors (D), OS analyses in select subgroups (E). Gray shading denotes probability of survival between upper and lower 95% CI. ADX, andecaliximab; IFN, interferon; OS, overall survival; PBO, placebo; PD-L1, programmed death ligand 1; PFS, progression-free survival; TC+IC, tumor and associated immune cell-positive.
Figure 3
Figure 3
Associations between hallmark EMT ssGSEA score and TGF-β gene signatures in all archival tumors with gene expression (n=80) (A), and between TGF-β fibrosis gene signature scores and Hallmark EMT ssGSEA score (B). Correlation between desmoplasia and TGF-β (C), between desmoplasia and EMT (D). OS for EMT ssGSEA score high versus low (median cut) patients (E), and OS for TGF-β ssGSEA score high versus low (median cut) patients (F). (A)* In all archival tumors with gene expression data (n=80). EMT, epithelial-to-mesenchymal transition; OS, overall survival; ssGSEA, single-sample gene set enrichment analysis; TGF, transforming growth factor.
Figure 4
Figure 4
Plot analysis of gene signatures associated with survival in ERBB2 expression according to documented HER2 status (A), OS by HER2 status (B), chromosome instability signature (almac) according to HER2 status (C)57 58, and OS by TMB high versus low (median cut) (D). HER2, human epidermal growth factor receptor 2; OS, overall survival; TMB, tumor mutation burden.
Figure 5
Figure 5
Association between documented HER2 status and TGF-β fibrosis signature according to HER2 status (A), TMB according to HER2 status (B), correlation between ERBB2 gene expression and TGF-β fibrosis ssGSEA score (C), and a Cox model of OS incorporating TMB, TGF-β fibrosis, and HER2 (D). (C)*All HRs refer to biomarker level above median versus below median. HER2, human epidermal growth factor receptor 2; OS, overall survival; ssGSEA, single-sample gene set enrichment analysis; TGF, transforming growth factor; TMB, tumor mutation burden.

References

    1. Ford HER, Marshall A, Bridgewater JA, et al. . Docetaxel versus active symptom control for refractory oesophagogastric adenocarcinoma (COUGAR-02): an open-label, phase 3 randomised controlled trial. Lancet Oncol 2014;15:78–86. 10.1016/S1470-2045(13)70549-7
    1. Kang JH, Lee SI, Lim DH, et al. . Salvage chemotherapy for pretreated gastric cancer: a randomized phase III trial comparing chemotherapy plus best supportive care with best supportive care alone. J Clin Oncol 2012;30:1513–8. 10.1200/JCO.2011.39.4585
    1. Fuchs CS, Tomasek J, Yong CJ, et al. . Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (regard): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2014;383:31–9. 10.1016/S0140-6736(13)61719-5
    1. Wilke H, Muro K, Van Cutsem E, et al. . Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 2014;15:1224–35. 10.1016/S1470-2045(14)70420-6
    1. Appleby TC, Greenstein AE, Hung M, et al. . Biochemical characterization and structure determination of a potent, selective antibody inhibitor of human MMP9. J Biol Chem 2017;292:6810–20. 10.1074/jbc.M116.760579
    1. Farina AR, Mackay AR. Gelatinase B/MMP-9 in tumour pathogenesis and progression. Cancers (Basel) 2014;6:240–96. 10.3390/cancers6010240
    1. Yang Q, Ye Z-Y, Zhang J-X, et al. . Expression of matrix metalloproteinase-9 mRNA and vascular endothelial growth factor protein in gastric carcinoma and its relationship to its pathological features and prognosis. Anat Rec (Hoboken) 2010;293:2012–9. 10.1002/ar.21071
    1. Al-Batran S-E, Pauligk C, Wirtz R, et al. . The validation of matrix metalloproteinase-9 mRNA gene expression as a predictor of outcome in patients with metastatic gastric cancer. Ann Oncol 2012;23:1699–705. 10.1093/annonc/mdr552
    1. Iniesta P, Morán A, De Juan C, et al. . Biological and clinical significance of MMP-2, MMP-9, TIMP-1 and TIMP-2 in non-small cell lung cancer. Oncol Rep 2007;17:217–23. 10.3892/or.17.1.217
    1. Lukaszewicz-Zając M, Mroczko B, Szmitkowski M. Gastric cancer - The role of matrix metalloproteinases in tumor progression. Clin Chim Acta 2011;412:1725–30. 10.1016/j.cca.2011.06.003
    1. Ogata Y, Matono K, Sasatomi T, et al. . The MMP-9 expression determined the efficacy of postoperative adjuvant chemotherapy using oral fluoropyrimidines in stage II or III colorectal cancer. Cancer Chemother Pharmacol 2006;57:577–83. 10.1007/s00280-005-0081-9
    1. Shao W, Wang W, Xiong X-G, et al. . Prognostic impact of MMP-2 and MMP-9 expression in pathologic stage Ia non-small cell lung cancer. J Surg Oncol 2011;104:841–6. 10.1002/jso.22001
    1. Yousef EM, Tahir MR, St-Pierre Y, et al. . MMP-9 expression varies according to molecular subtypes of breast cancer. BMC Cancer 2014;14:609. 10.1186/1471-2407-14-609
    1. Zhao S, Ma W, Zhang M, et al. . High expression of CD147 and MMP-9 is correlated with poor prognosis of triple-negative breast cancer (TNBC) patients. Med Oncol 2013;30:335. 10.1007/s12032-012-0335-4
    1. Shah MA, Starodub A, Sharma S, et al. . Andecaliximab/GS-5745 alone and combined with mFOLFOX6 in advanced gastric and gastroesophageal junction adenocarcinoma: results from a phase I study. Clin Cancer Res 2018;24:3829–37. 10.1158/1078-0432.CCR-17-2469
    1. Shah MA, Yanez Ruiz EP, Bodoky G, et al. . A phase III, randomized, double-blind, placebo-controlled study to evaluate the efficacy and safety of andecaliximab combined with mFOLFOX6 as first-line treatment in patients with advanced gastric or gastroesophageal junction adenocarcinoma (GAMMA-1). JCO 2019;37:4. 10.1200/JCO.2019.37.4_suppl.4
    1. Vandooren J, Van den Steen PE, Opdenakker G. Biochemistry and molecular biology of gelatinase B or matrix metalloproteinase-9 (MMP-9): the next decade. Crit Rev Biochem Mol Biol 2013;48:222–72. 10.3109/10409238.2013.770819
    1. Hijova E. Matrix metalloproteinases: their biological functions and clinical implications. Bratisl Lek Listy 2005;106:127–32.
    1. Perng D-W, Chang K-T, Su K-C, et al. . Matrix metalloprotease-9 induces transforming growth factor-β(1) production in airway epithelium via activation of epidermal growth factor receptors. Life Sci 2011;89:204–12. 10.1016/j.lfs.2011.06.008
    1. Van den Steen PE, Proost P, Wuyts A, et al. . Neutrophil gelatinase B potentiates interleukin-8 tenfold by aminoterminal processing, whereas it degrades CTAP-III, PF-4, and Gro-alpha and leaves RANTES and MCP-2 intact. Blood 2000;96:2673–81. 10.1182/blood.V96.8.2673
    1. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 2010;141:52–67. 10.1016/j.cell.2010.03.015
    1. Ardi VC, Van den Steen PE, Opdenakker G, et al. . Neutrophil MMP-9 proenzyme, unencumbered by TIMP-1, undergoes efficient activation in vivo and catalytically induces angiogenesis via a basic fibroblast growth factor (FGF-2)/FGFR-2 pathway. J Biol Chem 2009;284:25854–66. 10.1074/jbc.M109.033472
    1. Deryugina EI, Quigley JP. Pleiotropic roles of matrix metalloproteinases in tumor angiogenesis: contrasting, overlapping and compensatory functions. Biochim Biophys Acta 2010;1803:103–20. 10.1016/j.bbamcr.2009.09.017
    1. Denney H, Clench MR, Woodroofe MN. Cleavage of chemokines CCL2 and CXCL10 by matrix metalloproteinases-2 and -9: implications for chemotaxis. Biochem Biophys Res Commun 2009;382:341–7. 10.1016/j.bbrc.2009.02.164
    1. Cox JH, Dean RA, Roberts CR, et al. . Matrix metalloproteinase processing of CXCL11/I-TAC results in loss of chemoattractant activity and altered glycosaminoglycan binding. J Biol Chem 2008;283:19389–99. 10.1074/jbc.M800266200
    1. Van den Steen PE, Husson SJ, Proost P, et al. . Carboxyterminal cleavage of the chemokines MIG and IP-10 by gelatinase B and neutrophil collagenase. Biochem Biophys Res Commun 2003;310:889–96. 10.1016/j.bbrc.2003.09.098
    1. Juric V, O'Sullivan C, Stefanutti E, et al. . MMP-9 inhibition promotes anti-tumor immunity through disruption of biochemical and physical barriers to T-cell trafficking to tumors. PLoS One 2018;13:e0207255. 10.1371/journal.pone.0207255
    1. Shitara K, Özgüroğlu M, Bang Y-J, et al. . Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial. Lancet 2018;392:123–33. 10.1016/S0140-6736(18)31257-1
    1. Kang Y-K, Boku N, Satoh T, et al. . Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390:2461–71. 10.1016/S0140-6736(17)31827-5
    1. Bang Y-J, Ruiz EY, Van Cutsem E, et al. . Phase III, randomised trial of avelumab versus physician’s choice of chemotherapy as third-line treatment of patients with advanced gastric or gastro-oesophageal junction cancer: primary analysis of JAVELIN Gastric 300. Ann Oncol 2018;29:2052–60. 10.1093/annonc/mdy264
    1. Muro K, Chung HC, Shankaran V, et al. . Pembrolizumab for patients with PD-L1-positive advanced gastric cancer (KEYNOTE-012): a multicentre, open-label, phase 1B trial. Lancet Oncol 2016;17:717–26. 10.1016/S1470-2045(16)00175-3
    1. Fuchs CS, Doi T, Jang RW, et al. . 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 2018;4:e180013. 10.1001/jamaoncol.2018.0013
    1. Yoshihara K, Shahmoradgoli M, Martínez E, et al. . Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun 2013;4:2612. 10.1038/ncomms3612
    1. Janjigian YY, Bendell J, Calvo E, et al. . CheckMate-032 study: efficacy and safety of nivolumab and nivolumab plus ipilimumab in patients with metastatic esophagogastric cancer. J Clin Oncol 2018;36:2836–44. 10.1200/JCO.2017.76.6212
    1. Andrews S. FastQC: a quality control tool for high throughput sequence data 2010.
    1. Okonechnikov K, Conesa A, García-Alcalde F. Qualimap 2: advanced multi-sample quality control for high-throughput sequencing data. Bioinformatics 2016;32:292–4. 10.1093/bioinformatics/btv566
    1. Patro R, Duggal G, Love MI, et al. . Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods 2017;14:417–9. 10.1038/nmeth.4197
    1. Dobin A, Davis CA, Schlesinger F, et al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21. 10.1093/bioinformatics/bts635
    1. Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol 2010;11:R25. 10.1186/gb-2010-11-3-r25
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 2010;26:139–40. 10.1093/bioinformatics/btp616
    1. Law CW, Chen Y, Shi W, et al. . voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol 2014;15:R29. 10.1186/gb-2014-15-2-r29
    1. Subramanian A, Tamayo P, Mootha VK, et al. . Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 2005;102:15545–50. 10.1073/pnas.0506580102
    1. Ayers M, Lunceford J, Nebozhyn M, et al. . IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 2017;127:2930–40. 10.1172/JCI91190
    1. Fehrenbacher L, Spira A, Ballinger M, et al. . Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016;387:1837–46. 10.1016/S0140-6736(16)00587-0
    1. Charoentong P, Finotello F, Angelova M, et al. . Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep 2017;18:248–62. 10.1016/j.celrep.2016.12.019
    1. Tan TZ, Miow QH, Miki Y, et al. . Epithelial-mesenchymal transition spectrum quantification and its efficacy in deciphering survival and drug responses of cancer patients. EMBO Mol Med 2014;6:1279–93. 10.15252/emmm.201404208
    1. Mariathasan S, Turley SJ, Nickles D, et al. . TGF-beta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018;554:544–8. 10.1038/nature25501
    1. Liberzon A, Birger C, Thorvaldsdóttir H, et al. . The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–25. 10.1016/j.cels.2015.12.004
    1. Barbie DA, Tamayo P, Boehm JS, et al. . Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 2009;462:108–12. 10.1038/nature08460
    1. Van der Auwera GA, Carneiro MO, Hartl C, et al. . From FastQ data to high confidence variant calls: the genome analysis toolkit best practices pipeline. Curr Protoc Bioinformatics 2013;43:1101–33. 10.1002/0471250953.bi1110s43
    1. Li H, Durbin R. Fast and accurate long-read alignment with Burrows-Wheeler transform. Bioinformatics 2010;26:589–95. 10.1093/bioinformatics/btp698
    1. Chalmers ZR, Connelly CF, Fabrizio D, et al. . Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 2017;9:34. 10.1186/s13073-017-0424-2
    1. Peinado H, Quintanilla M, Cano A. Transforming growth factor beta-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions. J Biol Chem 2003;278:21113–23. 10.1074/jbc.M211304200
    1. Carter SL, Eklund AC, Kohane IS, et al. . A signature of chromosomal instability inferred from gene expression profiles predicts clinical outcome in multiple human cancers. Nat Genet 2006;38:1043–8. 10.1038/ng1861
    1. Szász AM, Li Q, Eklund AC, et al. . The CIN4 chromosomal instability qPCR classifier defines tumor aneuploidy and stratifies outcome in grade 2 breast cancer. PLoS One 2013;8:e56707. 10.1371/journal.pone.0056707
    1. Mouw KW, Goldberg MS, Konstantinopoulos PA, et al. . DNA damage and repair biomarkers of immunotherapy response. Cancer Discov 2017;7:675–93. 10.1158/-17-0226
    1. Konstantinopoulos PA, Spentzos D, Karlan BY, et al. . Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer. J Clin Oncol 2010;28:3555–61. 10.1200/JCO.2009.27.5719
    1. Severson TM, Wolf DM, Yau C, et al. . The BRCA1ness signature is associated significantly with response to PARP inhibitor treatment versus control in the I-SPY 2 randomized neoadjuvant setting. Breast Cancer Res 2017;19:99. 10.1186/s13058-017-0861-2
    1. Kang J, D'Andrea AD, Kozono D. A DNA repair pathway-focused score for prediction of outcomes in ovarian cancer treated with platinum-based chemotherapy. J Natl Cancer Inst 2012;104:670–81. 10.1093/jnci/djs177
    1. Kruhøffer M, Jensen JL, Laiho P, et al. . Gene expression signatures for colorectal cancer microsatellite status and HNPCC. Br J Cancer 2005;92:2240–8. 10.1038/sj.bjc.6602621
    1. Peng G, Chun-Jen Lin C, Mo W, et al. . Genome-wide transcriptome profiling of homologous recombination DNA repair. Nat Commun 2014;5:3361. 10.1038/ncomms4361
    1. McGrail DJ, Lin CC-J, Garnett J, et al. . Improved prediction of PARP inhibitor response and identification of synergizing agents through use of a novel gene expression signature generation algorithm. NPJ Syst Biol Appl 2017;3:8. 10.1038/s41540-017-0011-6
    1. Richard C, Fumet J-D, Chevrier S, et al. . Exome analysis reveals genomic markers associated with better efficacy of nivolumab in lung cancer patients. Clin Cancer Res 2019;25:957–66. 10.1158/1078-0432.CCR-18-1940
    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. 10.1126/scitranslmed.aah3560
    1. Koh J, Ock C-Y, Kim JW, et al. . Clinicopathologic implications of immune classification by PD-L1 expression and CD8-positive tumor-infiltrating lymphocytes in stage II and III gastric cancer patients. Oncotarget 2017;8:26356–67. 10.18632/oncotarget.15465
    1. Satoh T, Kang Y-K, Chao Y, et al. . Exploratory subgroup analysis of patients with prior trastuzumab use in the ATTRACTION-2 trial: a randomized phase III clinical trial investigating the efficacy and safety of nivolumab in patients with advanced gastric/gastroesophageal junction cancer. Gastric Cancer 2020;23:143–53. 10.1007/s10120-019-00970-8
    1. Grillo F, Fassan M, Sarocchi F, et al. . HER2 heterogeneity in gastric/gastroesophageal cancers: from benchside to practice. World J Gastroenterol 2016;22:5879–87. 10.3748/wjg.v22.i26.5879
    1. Janjigian YY, Maron SB, Chatila WK, et al. . First-line pembrolizumab and trastuzumab in HER2-positive oesophageal, gastric, or gastro-oesophageal junction cancer: an open-label, single-arm, phase 2 trial. Lancet Oncol 2020;21:821–31. 10.1016/S1470-2045(20)30169-8
    1. Catenacci DVT, Kang Y-K, Park H, et al. . Margetuximab plus pembrolizumab in patients with previously treated, HER2-positive gastro-oesophageal adenocarcinoma (CP-MGAH22-05): a single-arm, phase 1b-2 trial. Lancet Oncol 2020;21:1066–76. 10.1016/S1470-2045(20)30326-0
    1. Janjigian YY, Kawazoe A, Yanez PE, et al. . Pembrolizumab plus trastuzumab and chemotherapy for HER2+ metastatic gastric or gastroesophageal junction (G/GEJ) cancer: initial findings of the global phase 3 KEYNOTE-811 study. JCO 2021;39:abstr 4013:4013. 10.1200/JCO.2021.39.15_suppl.4013
    1. Massagué J. TGF-beta in cancer. Cell 2008;134:215–30. 10.1016/j.cell.2008.07.001
    1. Kitisin K, Saha T, Blake T, et al. . Tgf-beta signaling in development. Sci STKE 2007;2007:cm1. 10.1126/stke.3992007cm1
    1. Brabletz T, Kalluri R, Nieto MA, et al. . EMT in cancer. Nat Rev Cancer 2018;18:128–34. 10.1038/nrc.2017.118
    1. Stojnev S, Krstić M, Čukuranović Kokoris J, et al. . Prognostic impact of canonical TGF-β signaling in urothelial bladder cancer. Medicina (Kaunas) 2019;5510.3390/medicina55060302
    1. Samstein RM, Lee C-H, Shoushtari AN, et al. . Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 2019;51:202–6. 10.1038/s41588-018-0312-8
    1. Cai H, Jing C, Chang X, et al. . Mutational landscape of gastric cancer and clinical application of genomic profiling based on target next-generation sequencing. J Transl Med 2019;17:189. 10.1186/s12967-019-1941-0
    1. Marabelle A, Fakih M, Lopez J, et al. . Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353–65. 10.1016/S1470-2045(20)30445-9

Source: PubMed

3
Abonner