Incorporating Immunotherapy in the Management of Gastric Cancer: Molecular and Clinical Implications
Abstract
:Simple Summary
Abstract
1. Introduction
2. Molecular Classification
3. The Role of Tumour-Infiltrating Immune Cells in the Gastric Cancer Microenvironment
4. Role of Chronic Infection with Helicobacter pylori and the Association with an Immunosuppressive Microenvironment
4.1. Contribution of H. pylori Virulence Factors to Chronic Inflammation
4.2. Host Immune Response to H. pylori Infection
4.3. H. pylori Infection Promotes an Immunosuppressive Tumour Microenvironment
5. Molecular Mechanisms Underlying the Pathology of Microsatellite Instable Gastric Cancer
6. EBV-Related Gastric Cancer
7. Potential Role and Relevance of POLE/D Mutations in Gastric Cancer
8. HER2 Overexpression in Gastric Cancer
9. Immunotherapy in the Clinical Management of Gastric Cancer
9.1. PD-L1 as a Prognostic Biomarker
9.2. Immunotherapy in Chemo-Resistant/Refractory Setting; PD-L1 Expression as a Predictive Biomarker
9.3. Immunotherapy in Previously Untreated Patients; PD-L1 Expression as a Predictive Biomarker
9.4. Immunotherapy in HER2-Positive Gastric Cancer
9.5. Immunotherapy Efficacy in Rare Subgroups: Role of MSI, EBV and TMB (Tumour Mutational Burden) Status as Predictive Biomarkers
10. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Oliveira, C.; Pinheiro, H.; Figueiredo, J.; Seruca, R.; Carneiro, F. Familial gastric cancer: Genetic susceptibility, pathology, and implications for management. Lancet Oncol. 2015, 16, e60–e70. [Google Scholar] [CrossRef]
- Rawla, P.; Barsouk, A. Epidemiology of gastric cancer: Global trends, risk factors and prevention. Prz. Gastroenterol. 2019, 14, 26–38. [Google Scholar] [CrossRef] [PubMed]
- Matsueda, S.; Graham, D.Y. Immunotherapy in gastric cancer. World J. Gastroenterol. 2014, 20, 1657–1666. [Google Scholar] [CrossRef] [PubMed]
- Couzin-Frankel, J. Breakthrough of the year 2013. Cancer immunotherapy. Science 2013, 342, 1432–1433. [Google Scholar] [CrossRef]
- Lauren, P. The two histological main types of gastric carcinoma: Diffuse and so-called intestinal-type carcinoma. An attempt at a histo-clinical classificaTION. Acta Pathol. Microbiol. Scand. 1965, 64, 31–49. [Google Scholar] [CrossRef] [PubMed]
- Milne, A.N.; Offerhaus, G.J. Early-onset gastric cancer: Learning lessons from the young. World J. Gastrointest. Oncol. 2010, 2, 59–64. [Google Scholar] [CrossRef]
- Ma, J.; Shen, H.; Kapesa, L.; Zeng, S. Lauren classification and individualized chemotherapy in gastric cancer. Oncol. Lett. 2016, 11, 2959–2964. [Google Scholar] [CrossRef]
- Hu, B.; El Hajj, N.; Sittler, S.; Lammert, N.; Barnes, R.; Meloni-Ehrig, A. Gastric cancer: Classification, histology and application of molecular pathology. J. Gastrointest. Oncol. 2012, 3, 251–261. [Google Scholar]
- Dicken, B.J.; Bigam, D.L.; Cass, C.; Mackey, J.R.; Joy, A.A.; Hamilton, S.M. Gastric adenocarcinoma: Review and considerations for future directions. Ann. Surg. 2005, 241, 27–39. [Google Scholar] [CrossRef]
- Li, Z.; Wu, J.; Ji, M.; Shi, L.; Xu, B.; Jiang, J.; Wu, C. Prognostic role of lemur tyrosine kinase 3 in postoperative gastric cancer. Mol. Clin. Oncol. 2014, 2, 756–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Geddert, H.; Zur Hausen, A.; Gabbert, H.E.; Sarbia, M. EBV-infection in cardiac and non-cardiac gastric adenocarcinomas is associated with promoter methylation of p16, p14 and APC, but not hMLH1. Anal. Cell. Pathol. 2010, 33, 143–149. [Google Scholar] [CrossRef]
- Nakano, H.; Saito, M.; Nakajima, S.; Saito, K.; Nakayama, Y.; Kase, K.; Yamada, L.; Kanke, Y.; Hanayama, H.; Onozawa, H.; et al. PD-L1 overexpression in EBV-positive gastric cancer is caused by unique genomic or epigenomic mechanisms. Sci. Rep. 2021, 11, 1982. [Google Scholar] [CrossRef] [PubMed]
- Murphy, G.; Pfeiffer, R.; Camargo, M.C.; Rabkin, C.S. Meta-analysis shows that prevalence of Epstein-Barr virus-positive gastric cancer differs based on sex and anatomic location. Gastroenterology 2009, 137, 824–833. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.H.; Zhang, S.Y.; Hou, Q.Q.; Qin, Y.; Chen, X.Z.; Zhou, Z.G.; Shu, Y.; Xu, H.; Hu, J.K. The Significance of the CLDN18-ARHGAP Fusion Gene in Gastric Cancer: A Systematic Review and Meta-Analysis. Front. Oncol. 2020, 10, 1214. [Google Scholar] [CrossRef]
- Rihawi, K.; Ricci, A.D.; Rizzo, A.; Brocchi, S.; Marasco, G.; Pastore, L.V.; Llimpe, F.L.R.; Golfieri, R.; Renzulli, M. Tumor-Associated Macrophages and Inflammatory Microenvironment in Gastric Cancer: Novel Translational Implications. Int. J. Mol. Sci. 2021, 22, 3805. [Google Scholar] [CrossRef]
- Räihä, M.R.; Puolakkainen, P.A. Tumor-associated macrophages (TAMs) as biomarkers for gastric cancer: A review. Chronic Dis. Transl. Med. 2018, 4, 156–163. [Google Scholar] [CrossRef]
- Wang, X.L.; Jiang, J.T.; Wu, C.P. Prognostic significance of tumor-associated macrophage infiltration in gastric cancer: A meta-analysis. Genet. Mol. Res. 2016, 15, gmr15049040. [Google Scholar] [CrossRef]
- Zhang, Q.W.; Liu, L.; Gong, C.Y.; Shi, H.S.; Zeng, Y.H.; Wang, X.Z.; Zhao, Y.W.; Wei, Y.Q. Prognostic significance of tumor-associated macrophages in solid tumor: A meta-analysis of the literature. PLoS ONE 2012, 7, e50946. [Google Scholar] [CrossRef]
- Wang, Z.; Yin, N.; Zhang, Z.; Zhang, Y.; Zhang, G.; Chen, W. Upregulation of T-cell Immunoglobulin and Mucin-Domain Containing-3 (Tim-3) in Monocytes/Macrophages Associates with Gastric Cancer Progression. Immunol. Investig. 2017, 46, 134–148. [Google Scholar] [CrossRef]
- Sunakawa, Y.; Stremitzer, S.; Cao, S.; Zhang, W.; Yang, D.; Wakatsuki, T.; Ning, Y.; Yamauchi, S.; Stintzing, S.; Sebio, A.; et al. Association of variants in genes encoding for macrophage-related functions with clinical outcome in patients with locoregional gastric cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2015, 26, 332–339. [Google Scholar] [CrossRef]
- Ju, X.; Zhang, H.; Zhou, Z.; Chen, M.; Wang, Q. Tumor-associated macrophages induce PD-L1 expression in gastric cancer cells through IL-6 and TNF-ɑ signaling. Exp. Cell Res. 2020, 396, 112315. [Google Scholar] [CrossRef]
- Shen, Z.; Kauttu, T.; Seppänen, H.; Vainionpää, S.; Ye, Y.; Wang, S.; Mustonen, H.; Puolakkainen, P. Both macrophages and hypoxia play critical role in regulating invasion of gastric cancer in vitro. Acta Oncol. 2013, 52, 852–860. [Google Scholar] [CrossRef]
- Rahma, O.E.; Hodi, F.S. The Intersection between Tumor Angiogenesis and Immune Suppression. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 5449–5457. [Google Scholar] [CrossRef]
- Harada, K.; Dong, X.; Estrella, J.S.; Correa, A.M.; Xu, Y.; Hofstetter, W.L.; Sudo, K.; Onodera, H.; Suzuki, K.; Suzuki, A.; et al. Tumor-associated macrophage infiltration is highly associated with PD-L1 expression in gastric adenocarcinoma. Gastric Cancer Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2018, 21, 31–40. [Google Scholar] [CrossRef]
- Cerwenka, A.; Lanier, L.L. Natural killers join the fight against cancer. Science 2018, 359, 1460–1461. [Google Scholar] [CrossRef]
- Martín-Antonio, B.; Suñe, G.; Perez-Amill, L.; Castella, M.; Urbano-Ispizua, A. Natural Killer Cells: Angels and Devils for Immunotherapy. Int. J. Mol. Sci. 2017, 18, 1868. [Google Scholar] [CrossRef]
- Xia, P.; Xu, X.Y. DKK3 attenuates the cytotoxic effect of natural killer cells on CD133(+) gastric cancer cells. Mol. Carcinog. 2017, 56, 1712–1721. [Google Scholar] [CrossRef]
- Saito, H.; Takaya, S.; Osaki, T.; Ikeguchi, M. Increased apoptosis and elevated Fas expression in circulating natural killer cells in gastric cancer patients. Gastric Cancer Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2013, 16, 473–479. [Google Scholar] [CrossRef]
- González, S.; López-Soto, A.; Suarez-Alvarez, B.; López-Vázquez, A.; López-Larrea, C. NKG2D ligands: Key targets of the immune response. Trends Immunol. 2008, 29, 397–403. [Google Scholar] [CrossRef]
- Chen, J.; Yang, J.; Jiang, J.; Zhuang, Y.; He, W. Function and subsets of dendritic cells and natural killer cells were decreased in gastric cancer. Int. J. Clin. Exp. Pathol. 2014, 7, 8304–8311. [Google Scholar] [PubMed]
- Li, T.; Zhang, Q.; Jiang, Y.; Yu, J.; Hu, Y.; Mou, T.; Chen, G.; Li, G. Gastric cancer cells inhibit natural killer cell proliferation and induce apoptosis via prostaglandin E2. Oncoimmunology 2016, 5, e1069936. [Google Scholar] [CrossRef] [PubMed]
- Ishigami, S.; Natsugoe, S.; Tokuda, K.; Nakajo, A.; Xiangming, C.; Iwashige, H.; Aridome, K.; Hokita, S.; Aikou, T. Clinical impact of intratumoral natural killer cell and dendritic cell infiltration in gastric cancer. Cancer Lett. 2000, 159, 103–108. [Google Scholar] [CrossRef]
- Kashimura, S.; Saze, Z.; Terashima, M.; Soeta, N.; Ohtani, S.; Osuka, F.; Kogure, M.; Gotoh, M. CD83(+) dendritic cells and Foxp3(+) regulatory T cells in primary lesions and regional lymph nodes are inversely correlated with prognosis of gastric cancer. Gastric Cancer Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2012, 15, 144–153. [Google Scholar] [CrossRef]
- Kono, K.; Takahashi, A.; Sugai, H.; Fujii, H.; Choudhury, A.R.; Kiessling, R.; Matsumoto, Y. Dendritic cells pulsed with HER-2/neu-derived peptides can induce specific T-cell responses in patients with gastric cancer. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2002, 8, 3394–3400. [Google Scholar]
- Tewari, M.; Sahai, S.; Mishra, R.R.; Shukla, S.K.; Shukla, H.S. Dendritic cell therapy in advanced gastric cancer: A promising new hope? Surg. Oncol. 2012, 21, 164–171. [Google Scholar]
- Kang, B.W.; Kim, J.G.; Lee, I.H.; Bae, H.I.; Seo, A.N. Clinical significance of tumor-infiltrating lymphocytes for gastric cancer in the era of immunology. World J. Gastrointest. Oncol. 2017, 9, 293–299. [Google Scholar] [CrossRef]
- Kang, B.W.; Seo, A.N.; Yoon, S.; Bae, H.I.; Jeon, S.W.; Kwon, O.K.; Chung, H.Y.; Yu, W.; Kang, H.; Kim, J.G. Prognostic value of tumor-infiltrating lymphocytes in Epstein-Barr virus-associated gastric cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2016, 27, 494–501. [Google Scholar] [CrossRef] [PubMed]
- Grogg, K.L.; Lohse, C.M.; Pankratz, V.S.; Halling, K.C.; Smyrk, T.C. Lymphocyte-rich gastric cancer: Associations with Epstein-Barr virus, microsatellite instability, histology, and survival. Mod. Pathol. 2003, 16, 641–651. [Google Scholar] [CrossRef]
- Lee, H.E.; Chae, S.W.; Lee, Y.J.; Kim, M.A.; Lee, H.S.; Lee, B.L.; Kim, W.H. Prognostic implications of type and density of tumour-infiltrating lymphocytes in gastric cancer. Br. J. Cancer 2008, 99, 1704–1711. [Google Scholar] [CrossRef]
- Kawazoe, A.; Kuwata, T.; Kuboki, Y.; Shitara, K.; Nagatsuma, A.K.; Aizawa, M.; Yoshino, T.; Doi, T.; Ohtsu, A.; Ochiai, A. Clinicopathological features of programmed death ligand 1 expression with tumor-infiltrating lymphocyte, mismatch repair, and Epstein-Barr virus status in a large cohort of gastric cancer patients. Gastric Cancer Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2017, 20, 407–415. [Google Scholar] [CrossRef] [PubMed]
- Liu, T.; Peng, L.; Yu, P.; Zhao, Y.; Shi, Y.; Mao, X.; Chen, W.; Cheng, P.; Wang, T.; Chen, N.; et al. Increased circulating Th22 and Th17 cells are associated with tumor progression and patient survival in human gastric cancer. J. Clin. Immunol. 2012, 32, 1332–1339. [Google Scholar] [CrossRef] [PubMed]
- Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Böger, C.; Behrens, H.M.; Mathiak, M.; Krüger, S.; Kalthoff, H.; Röcken, C. PD-L1 is an independent prognostic predictor in gastric cancer of Western patients. Oncotarget 2016, 7, 24269–24283. [Google Scholar] [CrossRef]
- Peek, R.M.; Blaser, M.J., Jr. Helicobacter pylori and gastrointestinal tract adenocarcinomas. Nat. Rev. Cancer 2002, 2, 28–37. [Google Scholar] [CrossRef]
- Wroblewski, L.E.; Peek, R.M.; Wilson, K.T., Jr. Helicobacter pylori and gastric cancer: Factors that modulate disease risk. Clin. Microbiol. Rev. 2010, 23, 713–739. [Google Scholar] [CrossRef]
- Sokic-Milutinovic, A.; Alempijevic, T.; Milosavljevic, T. Role of Helicobacter pylori infection in gastric carcinogenesis: Current knowledge and future directions. World J. Gastroenterol. 2015, 21, 11654–11672. [Google Scholar] [CrossRef]
- Talley, N.J.; Zinsmeister, A.R.; Weaver, A.; DiMagno, E.P.; Carpenter, H.A.; Perez-Perez, G.I.; Blaser, M.J. Gastric adenocarcinoma and Helicobacter pylori infection. J. Natl. Cancer Inst. 1991, 83, 1734–1739. [Google Scholar] [CrossRef]
- Uemura, N.; Okamoto, S.; Yamamoto, S.; Matsumura, N.; Yamaguchi, S.; Yamakido, M.; Taniyama, K.; Sasaki, N.; Schlemper, R.J. Helicobacter pylori infection and the development of gastric cancer. N. Engl. J. Med. 2001, 345, 784–789. [Google Scholar] [CrossRef]
- Morgan, E.; Arnold, M.; Camargo, M.C.; Gini, A.; Kunzmann, A.T.; Matsuda, T.; Meheus, F.; Verhoeven, R.H.A.; Vignat, J.; Laversanne, M.; et al. The current and future incidence and mortality of gastric cancer in 185 countries, 2020–2040: A population-based modelling study. EClinicalMedicine 2022, 47, 101404. [Google Scholar] [CrossRef]
- Fuccio, L.; Eusebi, L.H.; Bazzoli, F. Gastric cancer, Helicobacter pylori infection and other risk factors. World J. Gastrointest. Oncol. 2010, 2, 342–347. [Google Scholar] [CrossRef]
- Karkhah, A.; Ebrahimpour, S.; Rostamtabar, M.; Koppolu, V.; Darvish, S.; Vasigala, V.K.R.; Validi, M.; Nouri, H.R. Helicobacter pylori evasion strategies of the host innate and adaptive immune responses to survive and develop gastrointestinal diseases. Microbiol. Res. 2019, 218, 49–57. [Google Scholar] [CrossRef]
- Posselt, G.; Backert, S.; Wessler, S. The functional interplay of Helicobacter pylori factors with gastric epithelial cells induces a multi-step process in pathogenesis. Cell Commun. Signal. 2013, 11, 77. [Google Scholar] [CrossRef] [Green Version]
- Alfarouk, K.O.; Bashir, A.H.H.; Aljarbou, A.N.; Ramadan, A.M.; Muddathir, A.K.; AlHoufie, S.T.S.; Hifny, A.; Elhassan, G.O.; Ibrahim, M.E.; Alqahtani, S.S.; et al. The Possible Role of Helicobacter pylori in Gastric Cancer and Its Management. Front. Oncol. 2019, 9, 75. [Google Scholar] [CrossRef]
- Amieva, M.R.; El-Omar, E.M. Host-bacterial interactions in Helicobacter pylori infection. Gastroenterology 2008, 134, 306–323. [Google Scholar] [CrossRef]
- Dunne, C.; Dolan, B.; Clyne, M. Factors that mediate colonization of the human stomach by Helicobacter pylori. World J. Gastroenterol. 2014, 20, 5610–5624. [Google Scholar] [CrossRef]
- Chichirau, B.E.; Diechler, S.; Posselt, G.; Wessler, S. Tyrosine Kinases in Helicobacter pylori Infections and Gastric Cancer. Toxins 2019, 11, 591. [Google Scholar] [CrossRef]
- Ansari, S.; Yamaoka, Y. Helicobacter pylori Virulence Factors Exploiting Gastric Colonization and its Pathogenicity. Toxins 2019, 11, 677. [Google Scholar] [CrossRef]
- Gu, H. Role of Flagella in the Pathogenesis of Helicobacter pylori. Curr. Microbiol. 2017, 74, 863–869. [Google Scholar] [CrossRef]
- Robinson, K.; Argent, R.H.; Atherton, J.C. The inflammatory and immune response to Helicobacter pylori infection. Best Pract. Res. Clin. Gastroenterol. 2007, 21, 237–259. [Google Scholar] [CrossRef]
- Matos, J.I.; de Sousa, H.A.; Marcos-Pinto, R.; Dinis-Ribeiro, M. Helicobacter pylori CagA and VacA genotypes and gastric phenotype: A meta-analysis. Eur. J. Gastroenterol. Hepatol. 2013, 25, 1431–1441. [Google Scholar] [CrossRef]
- Lu, H.; Hsu, P.I.; Graham, D.Y.; Yamaoka, Y. Duodenal ulcer promoting gene of Helicobacter pylori. Gastroenterology 2005, 128, 833–848. [Google Scholar] [CrossRef]
- Rad, R.; Gerhard, M.; Lang, R.; Schöniger, M.; Rösch, T.; Schepp, W.; Becker, I.; Wagner, H.; Prinz, C. The Helicobacter pylori blood group antigen-binding adhesin facilitates bacterial colonization and augments a nonspecific immune response. J. Immunol. 2002, 168, 3033–3041. [Google Scholar] [CrossRef] [Green Version]
- Müller, A.; Oertli, M.; Arnold, I.C.H. H. pylori exploits and manipulates innate and adaptive immune cell signaling pathways to establish persistent infection. Cell Commun. Signal. 2011, 9, 25. [Google Scholar] [CrossRef]
- Lima de Souza Gonçalves, V.; Cordeiro Santos, M.L.; Silva Luz, M.; Santos Marques, H.; de Brito, B.B.; França da Silva, F.A.; Souza, C.L.; Oliveira, M.V.; de Melo, F.F. From Helicobacter pylori infection to gastric cancer: Current evidence on the immune response. World J. Clin. Oncol. 2022, 13, 186–199. [Google Scholar] [CrossRef]
- Mentis, A.A.; Boziki, M.; Grigoriadis, N.; Papavassiliou, A.G. Helicobacter pylori infection and gastric cancer biology: Tempering a double-edged sword. Cell. Mol. Life Sci. 2019, 76, 2477–2486. [Google Scholar] [CrossRef] [PubMed]
- Wilson, K.T.; Crabtree, J.E. Immunology of Helicobacter pylori: Insights into the failure of the immune response and perspectives on vaccine studies. Gastroenterology 2007, 133, 288–308. [Google Scholar] [CrossRef]
- Abadi, A.T.B. Strategies used by Helicobacter pylori to establish persistent infection. World J. Gastroenterol. 2017, 23, 2870–2882. [Google Scholar] [CrossRef]
- Baldari, C.T.; Lanzavecchia, A.; Telford, J.L. Immune subversion by Helicobacter pylori. Trends Immunol. 2005, 26, 199–207. [Google Scholar] [CrossRef]
- Oster, P.; Vaillant, L.; McMillan, B.; Velin, D. The Efficacy of Cancer Immunotherapies Is Compromised by Helicobacter pylori Infection. Front. Immunol. 2022, 13, 899161. [Google Scholar] [CrossRef]
- Oster, P.; Vaillant, L.; Riva, E.; McMillan, B.; Begka, C.; Truntzer, C.; Richard, C.; Leblond, M.M.; Messaoudene, M.; Machremi, E.; et al. Helicobacter pylori infection has a detrimental impact on the efficacy of cancer immunotherapies. Gut 2022, 71, 457–466. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Guo, F. Recent updates on cancer immunotherapy. Precis. Clin. Med. 2018, 1, 65–74. [Google Scholar] [CrossRef]
- Paluch, C.; Santos, A.M.; Anzilotti, C.; Cornall, R.J.; Davis, S.J. Immune Checkpoints as Therapeutic Targets in Autoimmunity. Front. Immunol. 2018, 9, 2306. [Google Scholar] [CrossRef]
- Zhao, L.; Liu, Y.; Zhang, S.; Wei, L.; Cheng, H.; Wang, J.; Wang, J. Impacts and mechanisms of metabolic reprogramming of tumor microenvironment for immunotherapy in gastric cancer. Cell Death Dis. 2022, 13, 378. [Google Scholar] [CrossRef]
- Luo, B.; Wang, M.; Hou, N.; Hu, X.; Jia, G.; Qin, X.; Zuo, X.; Liu, Y.; Luo, K.; Song, W.; et al. ATP-Dependent Lon Protease Contributes to Helicobacter pylori-Induced Gastric Carcinogenesis. Neoplasia 2016, 18, 242–252. [Google Scholar] [CrossRef]
- Lewis, N.D.; Asim, M.; Barry, D.P.; Singh, K.; de Sablet, T.; Boucher, J.L.; Gobert, A.P.; Chaturvedi, R.; Wilson, K.T. Arginase II restricts host defense to Helicobacter pylori by attenuating inducible nitric oxide synthase translation in macrophages. J. Immunol. 2010, 184, 2572–2582. [Google Scholar] [CrossRef]
- Eun, C.S.; Kim, B.K.; Han, D.S.; Kim, S.Y.; Kim, K.M.; Choi, B.Y.; Song, K.S.; Kim, Y.S.; Kim, J.F. Differences in gastric mucosal microbiota profiling in patients with chronic gastritis, intestinal metaplasia, and gastric cancer using pyrosequencing methods. Helicobacter 2014, 19, 407–416. [Google Scholar] [CrossRef]
- Ling, Z.; Shao, L.; Liu, X.; Cheng, Y.; Yan, C.; Mei, Y.; Ji, F.; Liu, X. Regulatory T Cells and Plasmacytoid Dendritic Cells Within the Tumor Microenvironment in Gastric Cancer Are Correlated with Gastric Microbiota Dysbiosis: A Preliminary Study. Front. Immunol. 2019, 10, 533. [Google Scholar] [CrossRef]
- Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillere, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef]
- Vetizou, M.; Pitt, J.M.; Daillere, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
- Silva, R.; Gullo, I.; Carneiro, F. The PD-1:PD-L1 immune inhibitory checkpoint in Helicobacter pylori infection and gastric cancer: A comprehensive review and future perspectives. Porto Biomed. J. 2016, 1, 4–11. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Zheng, H.; Wang, M.; Ding, S. Influence of Helicobacter pylori infection on PD-1/PD-L1 blockade therapy needs more attention. Helicobacter 2022, 27, e12878. [Google Scholar] [CrossRef]
- Lengauer, C.; Kinzler, K.W.; Vogelstein, B. Genetic instabilities in human cancers. Nature 1998, 396, 643–649. [Google Scholar] [CrossRef]
- Belien, J.A.M.; Buffart, T.E.; Gill, A.J.; Broeckaert, M.A.M.; Quirke, P.; Meijer, G.A.; Grabsch, H.I. Gross genomic damage measured by DNA image cytometry independently predicts gastric cancer patient survival. Br. J. Cancer 2009, 101, 1011–1018. [Google Scholar] [CrossRef]
- Ratti, M.; Lampis, A.; Hahne, J.C.; Passalacqua, R.; Valeri, N. Microsatellite instability in gastric cancer: Molecular bases, clinical perspectives, and new treatment approaches. Cell. Mol. Life Sci. 2018, 75, 4151–4162. [Google Scholar] [CrossRef]
- Puliga, E.; Corso, S.; Pietrantonio, F.; Giordano, S. Microsatellite instability in Gastric Cancer: Between lights and shadows. Cancer Treat. Rev. 2021, 95, 102175. [Google Scholar] [CrossRef]
- Julié, C.; Trésallet, C.; Brouquet, A.; Vallot, C.; Zimmermann, U.; Mitry, E.; Radvanyi, F.; Rouleau, E.; Lidereau, R.; Coulet, F.; et al. Identification in daily practice of patients with Lynch syndrome (hereditary nonpolyposis colorectal cancer): Revised Bethesda guidelines-based approach versus molecular screening. Am. J. Gastroenterol. 2008, 103, 2825–2835, quiz 36. [Google Scholar] [CrossRef] [PubMed]
- Hudler, P. Genetic aspects of gastric cancer instability. Sci. World J. 2012, 2012, 761909. [Google Scholar] [CrossRef] [PubMed]
- Yuza, K.; Nagahashi, M.; Watanabe, S.; Takabe, K.; Wakai, T. Hypermutation and microsatellite instability in gastrointestinal cancers. Oncotarget 2017, 8, 112103–112115. [Google Scholar] [CrossRef]
- Leite, M.; Corso, G.; Sousa, S.; Milanezi, F.; Afonso, L.P.; Henrique, R.; Soares, J.M.; Castedo, S.; Carneiro, F.; Roviello, F.; et al. MSI phenotype and MMR alterations in familial and sporadic gastric cancer. Int. J. Cancer 2011, 128, 1606–1613. [Google Scholar] [CrossRef] [PubMed]
- Gu, M.; Kim, D.; Bae, Y.; Choi, J.; Kim, S.; Song, S. Analysis of microsatellite instability, protein expression and methylation status of hMLH1 and hMSH2 genes in gastric carcinomas. Hepato-Gastroenterology 2009, 56, 899–904. [Google Scholar] [PubMed]
- Bevilacqua, R.A.; Simpson, A.J. Methylation of the hMLH1 promoter but no hMLH1 mutations in sporadic gastric carcinomas with high-level microsatellite instability. Int. J. Cancer 2000, 87, 200–203. [Google Scholar] [CrossRef]
- Wu, M.-S.; Sheu, J.-C.; Shun, C.-T.; Lee, W.-J.; Wang, J.-T.; Wang, T.-H.; Cheng, A.-L.; Lin, J.-T. Infrequent hMSH2 mutations in sporadic gastric adenocarcinoma with microsatellite instability. Cancer Lett. 1997, 112, 161–166. [Google Scholar]
- Sugimoto, R.; Sugai, T.; Habano, W.; Endoh, M.; Eizuka, M.; Yamamoto, E.; Uesugi, N.; Ishida, K.; Kawasaki, T.; Matsumoto, T.; et al. Clinicopathological and molecular alterations in early gastric cancers with the microsatellite instability-high phenotype. Int. J. Cancer 2016, 138, 1689–1697. [Google Scholar] [CrossRef]
- Li, B.; Liu, H.Y.; Guo, S.H.; Sun, P.; Gong, F.M.; Jia, B.Q. Microsatellite instability of gastric cancer and precancerous lesions. Int. J. Clin. Exp. Med. 2015, 8, 21138–21144. [Google Scholar] [CrossRef] [PubMed]
- Ling, Z.-Q.; Tanaka, A.; Li, P.; Nakayama, T.; Fujiyama, Y.; Hattori, T.; Sugihara, H. Microsatellite instability with promoter methylation and silencing of hMLH1 can regionally occur during progression of gastric carcinoma. Cancer Lett. 2010, 297, 244–251. [Google Scholar] [PubMed]
- Corso, G.; Velho, S.; Paredes, J.; Pedrazzani, C.; Martins, D.; Milanezi, F.; Pascale, V.; Vindigni, C.; Pinheiro, H.; Leite, M.; et al. Oncogenic mutations in gastric cancer with microsatellite instability. Eur. J. Cancer 2011, 47, 443–451. [Google Scholar] [CrossRef] [PubMed]
- Ligtenberg, M.J.; Kuiper, R.P.; Chan, T.L.; Goossens, M.; Hebeda, K.M.; Voorendt, M.; Lee, T.Y.; Bodmer, D.; Hoenselaar, E.; Hendriks-Cornelissen, S.J.; et al. Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nat. Genet. 2009, 41, 112–117. [Google Scholar] [CrossRef]
- Kovacs, M.E.; Papp, J.; Szentirmay, Z.; Otto, S.; Olah, E. Deletions removing the last exon of TACSTD1 constitute a distinct class of mutations predisposing to Lynch syndrome. Hum. Mutat. 2009, 30, 197–203. [Google Scholar] [CrossRef]
- Singh, S.S.; Yap, W.N.; Arfuso, F.; Kar, S.; Wang, C.; Cai, W.; Dharmarajan, A.M.; Sethi, G.; Kumar, A.P. Targeting the PI3K/Akt signaling pathway in gastric carcinoma: A reality for personalized medicine? World J. Gastroenterol. 2015, 21, 12261–12273. [Google Scholar] [CrossRef]
- The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 513, 202–209. [Google Scholar] [CrossRef]
- Wang, K.; Kan, J.; Yuen, S.T.; Shi, S.T.; Chu, K.M.; Law, S.; Chan, T.L.; Kan, Z.; Chan, A.S.; Tsui, W.Y.; et al. Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat. Genet. 2011, 43, 1219–1223. [Google Scholar] [PubMed]
- Wang, K.; Yuen, S.T.; Xu, J.; Lee, S.P.; Yan, H.H.; Shi, S.T.; Siu, H.C.; Deng, S.; Chu, K.M.; Law, S.; et al. Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat. Genet. 2014, 46, 573–582. [Google Scholar] [CrossRef] [PubMed]
- Kim, M.S.; Oh, J.E.; Kim, Y.R.; Park, S.W.; Kang, M.R.; Kim, S.S.; Ahn, C.H.; Yoo, N.J.; Lee, S.H. Somatic mutations and losses of expression of microRNA regulation-related genes AGO2 and TNRC6A in gastric and colorectal cancers. J. Pathol. 2010, 221, 139–146. [Google Scholar] [CrossRef]
- Takayama, T.; Sakabe, T.; Fujii, M.; Yamada, E.; Uno, M.; Ono, Y. In vitro production of human antibodies specifically reactive with human gastric cancer cells of established lines and autologous tissues. J. Surg. Oncol. 1987, 36, 215–224. [Google Scholar] [CrossRef]
- Bocian, J.; Januszkiewicz-Lewandowska, D. Epstein-Barr virus infection—Life cycle, methods of diagnosis, associated diseases. Postepy Hig. I Med. Dosw. 2011, 65, 286–298. [Google Scholar] [CrossRef]
- Rodriquenz, M.G.; Roviello, G.; D′Angelo, A.; Lavacchi, D.; Roviello, F.; Polom, K. MSI and EBV Positive Gastric Cancer’s Subgroups and Their Link with Novel Immunotherapy. J. Clin. Med. 2020, 9, 1427. [Google Scholar] [CrossRef]
- Sun, K.; Jia, K.; Lv, H.; Wang, S.Q.; Wu, Y.; Lei, H.; Chen, X. EBV-Positive Gastric Cancer: Current Knowledge and Future Perspectives. Front. Oncol. 2020, 10, 583463. [Google Scholar] [CrossRef]
- Lieberman, P.M. Virology. Epstein-Barr virus turns 50. Science 2014, 343, 1323–1325. [Google Scholar] [CrossRef]
- Hutt-Fletcher, L.M. The Long and Complicated Relationship between Epstein-Barr Virus and Epithelial Cells. J. Virol. 2016, 91, e01677-16. [Google Scholar] [CrossRef] [PubMed]
- Nanbo, A.; Kachi, K.; Yoshiyama, H.; Ohba, Y. Epstein-Barr virus exploits host endocytic machinery for cell-to-cell viral transmission rather than a virological synapse. J. Gen. Virol. 2016, 97, 2989–3006. [Google Scholar] [CrossRef] [PubMed]
- Lieberman, P.M. Keeping it quiet: Chromatin control of gammaherpesvirus latency. Nat. Rev. Microbiol. 2013, 11, 863–875. [Google Scholar] [CrossRef]
- Rowlands, D.C.; Ito, M.; Mangham, D.C.; Reynolds, G.; Herbst, H.; Hallissey, M.T.; Fielding, J.W.; Newbold, K.M.; Jones, E.L.; Young, L.S.; et al. Epstein-Barr virus and carcinomas: Rare association of the virus with gastric adenocarcinomas. Br. J. Cancer 1993, 68, 1014–1019. [Google Scholar] [CrossRef] [Green Version]
- Shinozaki, A.; Sakatani, T.; Ushiku, T.; Hino, R.; Isogai, M.; Ishikawa, S.; Uozaki, H.; Takada, K.; Fukayama, M. Downregulation of microRNA-200 in EBV-associated gastric carcinoma. Cancer Res. 2010, 70, 4719–4727. [Google Scholar] [CrossRef]
- Zheng, X.; Wang, J.; Wei, L.; Peng, Q.; Gao, Y.; Fu, Y.; Lu, Y.; Qin, Z.; Zhang, X.; Lu, J.; et al. Epstein-Barr Virus MicroRNA miR-BART5-3p Inhibits p53 Expression. J. Virol. 2018, 92, e01022-18. [Google Scholar] [CrossRef]
- Song, Y.; Li, Q.; Liao, S.; Zhong, K.; Jin, Y.; Zeng, T. Epstein-Barr virus-encoded miR-BART11 promotes tumor-associated macrophage-induced epithelial-mesenchymal transition via targeting FOXP1 in gastric cancer. Virology 2020, 548, 6–16. [Google Scholar] [CrossRef]
- Dong, M.; Gong, L.P.; Chen, J.N.; Zhang, X.F.; Zhang, Y.W.; Hui, D.Y.; Zhao, X.X.; Wu, X.Y.; Shao, C.K. EBV-miR-BART10-3p and EBV-miR-BART22 promote metastasis of EBV-associated gastric carcinoma by activating the canonical Wnt signaling pathway. Cell. Oncol. 2020, 43, 901–913. [Google Scholar] [CrossRef]
- Matsusaka, K.; Kaneda, A.; Nagae, G.; Ushiku, T.; Kikuchi, Y.; Hino, R.; Uozaki, H.; Seto, Y.; Takada, K.; Aburatani, H.; et al. Classification of Epstein-Barr virus-positive gastric cancers by definition of DNA methylation epigenotypes. Cancer Res. 2011, 71, 7187–7197. [Google Scholar] [CrossRef]
- Liang, Q.; Yao, X.; Tang, S.; Zhang, J.; Yau, T.O.; Li, X.; Tang, C.M.; Kang, W.; Lung, R.W.; Li, J.W.; et al. Integrative identification of Epstein-Barr virus-associated mutations and epigenetic alterations in gastric cancer. Gastroenterology 2014, 147, 1350–1362.e4. [Google Scholar] [CrossRef]
- Yu, J.; Liang, Q.; Wang, J.; Wang, K.; Gao, J.; Zhang, J.; Zeng, Y.; Chiu, P.W.Y.; Ng, E.K.W.; Sung, J.J.Y. REC8 functions as a tumor suppressor and is epigenetically downregulated in gastric cancer, especially in EBV-positive subtype. Oncogene 2017, 36, 182–193. [Google Scholar] [CrossRef] [PubMed]
- Hino, R.; Uozaki, H.; Murakami, N.; Ushiku, T.; Shinozaki, A.; Ishikawa, S.; Morikawa, T.; Nakaya, T.; Sakatani, T.; Takada, K.; et al. Activation of DNA methyltransferase 1 by EBV latent membrane protein 2A leads to promoter hypermethylation of PTEN gene in gastric carcinoma. Cancer Res. 2009, 69, 2766–2774. [Google Scholar] [CrossRef]
- Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Zhou, M.; Ouyang, Y.; Du, L.; Xu, L.; Li, H. Identification of potential biomarkers and their clinical significance in gastric cancer using bioinformatics analysis methods. PeerJ 2020, 8, e9174. [Google Scholar] [CrossRef] [PubMed]
- Strosberg, J.; Mizuno, N.; Doi, T.; Grande, E.; Delord, J.P.; Shapira-Frommer, R.; Bergsland, E.; Shah, M.; Fakih, M.; Takahashi, S.; et al. Efficacy and Safety of Pembrolizumab in Previously Treated Advanced Neuroendocrine Tumors: Results from the Phase II KEYNOTE-158 Study. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2020, 26, 2124–2130. [Google Scholar] [CrossRef]
- Kono, K.; Nakajima, S.; Mimura, K. Current status of immune checkpoint inhibitors for gastric cancer. Gastric Cancer Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2020, 23, 565–578. [Google Scholar] [CrossRef]
- Sundar, R.; Smyth, E.C.; Peng, S.; Yeong, J.P.S.; Tan, P. Predictive Biomarkers of Immune Checkpoint Inhibition in Gastroesophageal Cancers. Front. Oncol. 2020, 10, 763. [Google Scholar] [CrossRef]
- Burgers, P.M.J.; Kunkel, T.A. Eukaryotic DNA Replication Fork. Annu. Rev. Biochem. 2017, 86, 417–438. [Google Scholar] [CrossRef]
- Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef]
- Wang, F.; Zhao, Q.; Wang, Y.N.; Jin, Y.; He, M.M.; Liu, Z.X.; Xu, R.H. Evaluation of POLE and POLD1 Mutations as Biomarkers for Immunotherapy Outcomes Across Multiple Cancer Types. JAMA Oncol. 2019, 5, 1504–1506. [Google Scholar] [CrossRef]
- Zhu, M.; Cui, H.; Zhang, L.; Zhao, K.; Jia, X.; Jin, H. Assessment of POLE and POLD1 mutations as prognosis and immunotherapy biomarkers for stomach adenocarcinoma. Transl. Cancer Res. 2022, 11, 193–205. [Google Scholar] [CrossRef] [PubMed]
- Abrahao-Machado, L.F.; Scapulatempo-Neto, C. HER2 testing in gastric cancer: An update. World J. Gastroenterol. 2016, 22, 4619–4625. [Google Scholar] [CrossRef] [PubMed]
- Baykara, M.; Benekli, M.; Ekinci, O.; Irkkan, S.C.; Karaca, H.; Demirci, U.; Akinci, M.B.; Unal, O.U.; Dane, F.; Turkoz, F.P.; et al. Clinical Significance of HER2 Overexpression in Gastric and Gastroesophageal Junction Cancers. J. Gastrointest. Surg. 2015, 19, 1565–1571. [Google Scholar] [CrossRef] [PubMed]
- Jørgensen, J.T.; Hersom, M. HER2 as a Prognostic Marker in Gastric Cancer—A Systematic Analysis of Data from the Literature. J. Cancer 2012, 3, 137–144. [Google Scholar] [CrossRef]
- Bang, Y.J.; Van Cutsem, E.; Feyereislova, A.; Chung, H.C.; Shen, L.; Sawaki, A.; Lordick, F.; Ohtsu, A.; Omuro, Y.; Satoh, T.; et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): A phase 3, open-label, randomised controlled trial. Lancet 2010, 376, 687–697. [Google Scholar] [CrossRef]
- Whiteside, T.L. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008, 27, 5904–5912. [Google Scholar] [CrossRef]
- Suh, K.J.; Sung, J.H.; Kim, J.W.; Han, S.-H.; Lee, H.S.; Min, A.; Kang, M.H.; Kim, J.E.; Kim, J.-W.; Kim, S.H.; et al. EGFR or HER2 inhibition modulates the tumor microenvironment by suppression of PD-L1 and cytokines release. Oncotarget 2017, 8, 63901–63910. [Google Scholar] [CrossRef]
- Lian, J.; Zhang, G.; Zhang, Y.; Liu, H.; Zhang, J.; Nan, P.; Tian, W. PD-L1 and HER2 expression in gastric adenocarcinoma and their prognostic significance. Dig. Liver Dis. 2022, in press. [Google Scholar] [CrossRef]
- Chakrabarti, J.; Koh, V.; Steele, N.; Hawkins, J.; Ito, Y.; Merchant, J.L.; Wang, J.; Helmrath, M.A.; Ahmad, S.A.; So, J.B.Y.; et al. Disruption of Her2-Induced PD-L1 Inhibits Tumor Cell Immune Evasion in Patient-Derived Gastric Cancer Organoids. Cancers 2021, 13, 6158. [Google Scholar] [CrossRef]
- Morihiro, T.; Kuroda, S.; Kanaya, N.; Kakiuchi, Y.; Kubota, T.; Aoyama, K.; Tanaka, T.; Kikuchi, S.; Nagasaka, T.; Nishizaki, M.; et al. PD-L1 expression combined with microsatellite instability/CD8+ tumor infiltrating lymphocytes as a useful prognostic biomarker in gastric cancer. Sci. Rep. 2019, 9, 4633. [Google Scholar] [CrossRef]
- Chang, H.; Jung, W.Y.; Kang, Y.; Lee, H.; Kim, A.; Kim, H.K.; Shin, B.K.; Kim, B.H. Programmed death-ligand 1 expression in gastric adenocarcinoma is a poor prognostic factor in a high CD8+ tumor infiltrating lymphocytes group. Oncotarget 2016, 7, 80426–80434. [Google Scholar] [CrossRef] [PubMed]
- Cho, J.; Lee, J.; Bang, H.; Kim, S.T.; Park, S.H.; An, J.Y.; Choi, M.G.; Lee, J.H.; Sohn, T.S.; Bae, J.M.; et al. Programmed cell death-ligand 1 expression predicts survival in patients with gastric carcinoma with microsatellite instability. Oncotarget 2017, 8, 13320–13328. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Xu, D.; Huang, C.; Guo, Y.; Wang, S.; Zhu, C.; Xu, J.; Zhang, Z.; Shen, Y.; Zhao, W.; et al. Regulatory T cells and M2 macrophages present diverse prognostic value in gastric cancer patients with different clinicopathologic characteristics and chemotherapy strategies. J. Transl. Med. 2019, 17, 192. [Google Scholar] [CrossRef]
- Dong, M.; Wang, H.Y.; Zhao, X.X.; Chen, J.N.; Zhang, Y.W.; Huang, Y.; Xue, L.; Li, H.G.; Du, H.; Wu, X.Y.; et al. Expression and prognostic roles of PIK3CA, JAK2, PD-L1, and PD-L2 in Epstein-Barr virus-associated gastric carcinoma. Hum. Pathol. 2016, 53, 25–34. [Google Scholar] [CrossRef]
- Wei, X.L.; Liu, Q.W.; Liu, F.R.; Yuan, S.S.; Li, X.F.; Li, J.N.; Yang, A.L.; Ling, Y.H. The clinicopathological significance and predictive value for immunotherapy of programmed death ligand-1 expression in Epstein-Barr virus-associated gastric cancer. Oncoimmunology 2021, 10, 1938381. [Google Scholar] [CrossRef] [PubMed]
- Gu, L.; Chen, M.; Guo, D.; Zhu, H.; Zhang, W.; Pan, J.; Zhong, X.; Li, X.; Qian, H.; Wang, X. PD-L1 and gastric cancer prognosis: A systematic review and meta-analysis. PLoS ONE 2017, 12, e0182692. [Google Scholar] [CrossRef]
- Kang, Y.K.; Boku, N.; Satoh, T.; Ryu, M.H.; Chao, Y.; Kato, K.; Chung, H.C.; Chen, J.S.; Muro, K.; Kang, W.K.; 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, 2461–2471. [Google Scholar] [CrossRef]
- Chen, L.T.; Satoh, T.; Ryu, M.H.; Chao, Y.; Kato, K.; Chung, H.C.; Chen, J.S.; Muro, K.; Kang, W.K.; Yeh, K.H.; et al. A phase 3 study of nivolumab in previously treated advanced gastric or gastroesophageal junction cancer (ATTRACTION-2): 2-year update data. Gastric Cancer Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2020, 23, 510–519. [Google Scholar] [CrossRef]
- Shitara, K.; Özgüroğlu, M.; Bang, Y.J.; Di Bartolomeo, M.; Mandalà, M.; Ryu, M.H.; Fornaro, L.; Olesiński, T.; Caglevic, C.; Chung, H.C.; 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, 123–133. [Google Scholar] [CrossRef]
- Fuchs, C.S.; Özgüroğlu, M.; Bang, Y.J.; Di Bartolomeo, M.; Mandala, M.; Ryu, M.H.; Fornaro, L.; Olesinski, T.; Caglevic, C.; Chung, H.C.; et al. Pembrolizumab versus paclitaxel for previously treated PD-L1-positive advanced gastric or gastroesophageal junction cancer: 2-year update of the randomized phase 3 KEYNOTE-061 trial. Gastric Cancer Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2022, 25, 197–206. [Google Scholar] [CrossRef]
- Bang, Y.J.; Ruiz, E.Y.; Van Cutsem, E.; Lee, K.W.; Wyrwicz, L.; Schenker, M.; Alsina, M.; Ryu, M.H.; Chung, H.C.; Evesque, L.; 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. Off. J. Eur. Soc. Med. Oncol. 2018, 29, 2052–2060. [Google Scholar] [CrossRef] [PubMed]
- Shitara, K.; Van Cutsem, E.; Bang, Y.J.; Fuchs, C.; Wyrwicz, L.; Lee, K.W.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; et al. Efficacy and Safety of Pembrolizumab or Pembrolizumab Plus Chemotherapy vs Chemotherapy Alone for Patients with First-line, Advanced Gastric Cancer: The KEYNOTE-062 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1571–1580. [Google Scholar] [CrossRef] [PubMed]
- Wainberg, Z.A.; Shitara, K.; Van Cutsem, E.; Wyrwicz, L.; Lee, K.W.; Kudaba, I.; Garrido, M.; Chung, H.C.C.; Lee, J.; Castro-Salguero, H.R.; et al. Pembrolizumab with or without chemotherapy versus chemotherapy alone for patients with PD-L1–positive advanced gastric or gastroesophageal junction adenocarcinoma: Update from the phase 3 KEYNOTE-062 trial. J. Clin. Oncol. 2022, 40 (Suppl. 4), iv152–iv153. [Google Scholar] [CrossRef]
- Moehler, M.; Dvorkin, M.; Boku, N.; Özgüroğlu, M.; Ryu, M.H.; Muntean, A.S.; Lonardi, S.; Nechaeva, M.; Bragagnoli, A.C.; Coşkun, H.S.; et al. Phase III Trial of Avelumab Maintenance After First-Line Induction Chemotherapy Versus Continuation of Chemotherapy in Patients with Gastric Cancers: Results From JAVELIN Gastric 100. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2021, 39, 966–977. [Google Scholar] [CrossRef] [PubMed]
- Janjigian, Y.Y.; Shitara, K.; Moehler, M.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Campos Bragagnoli, A.; et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): A randomised, open-label, phase 3 trial. Lancet 2021, 398, 27–40. [Google Scholar] [CrossRef]
- Shitara, K.; Janjigian, Y.Y.; Moehler, M.H.; Garrido, M.; Gallardo, C.; Shen, L.; Yamaguchi, K.; Wyrwicz, L.; Skoczylas, T.; Campos Bragagnoli, A.S.; et al. Nivolumab (NIVO) plus chemotherapy (chemo) versus chemo as first-line (1L) treatment for advanced gastric cancer/gastroesophageal junction cancer/esophageal adenocarcinoma (GC/GEJC/EAC): Expanded efficacy, safety, and subgroup analyses from CheckMate 649. J. Clin. Oncol. 2022, 40 (Suppl. S4), 240. [Google Scholar] [CrossRef]
- Shitara, K.; Ajani, J.A.; Moehler, M.; Garrido, M.; Gallardo, C.; Shen, L.; Yamaguchi, K.; Wyrwicz, L.; Skoczylas, T.; Bragagnoli, A.C.; et al. Nivolumab plus chemotherapy or ipilimumab in gastro-oesophageal cancer. Nature 2022, 603, 942–948. [Google Scholar] [CrossRef]
- Kang, Y.K.; Chen, L.T.; Ryu, M.H.; Oh, D.Y.; Oh, S.C.; Chung, H.C.; Lee, K.W.; Omori, T.; Shitara, K.; Sakuramoto, S.; et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): A randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2022, 23, 234–247. [Google Scholar]
- Xu, J.; Jiang, H.; Pan, Y.; Gu, K.; Cang, S.; Han, L.; Shu, Y.; Li, J.; Zhao, J.; Pan, H.; et al. LBA53 Sintilimab plus chemotherapy (chemo) versus chemo as first-line treatment for advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma (ORIENT-16): First results of a randomized, double-blind, phase III study. Ann. Oncol. 2021, 32, S1331. [Google Scholar] [CrossRef]
- Janjigian, Y.Y.; Kawazoe, A.; Yañez, P.; Li, N.; Lonardi, S.; Kolesnik, O.; Barajas, O.; Bai, Y.; Shen, L.; Tang, Y.; et al. The KEYNOTE-811 trial of dual PD-1 and HER2 blockade in HER2-positive gastric cancer. Nature 2021, 600, 727–730. [Google Scholar] [CrossRef]
- Kawazoe, A.; Shitara, K.; Kuboki, Y.; Bando, H.; Kojima, T.; Yoshino, T.; Ohtsu, A.; Ochiai, A.; Togashi, Y.; Nishikawa, H.; et al. Clinicopathological features of 22C3 PD-L1 expression with mismatch repair, Epstein-Barr virus status, and cancer genome alterations in metastatic gastric cancer. Gastric Cancer Off. J. Int. Gastric Cancer Assoc. Jpn. Gastric Cancer Assoc. 2019, 22, 69–76. [Google Scholar] [CrossRef] [PubMed]
- Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; et al. Efficacy of Pembrolizumab in Patients with Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020, 38, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Chao, J.; Fuchs, C.S.; Shitara, K.; Tabernero, J.; Muro, K.; Van Cutsem, E.; Bang, Y.J.; De Vita, F.; Landers, G.; Yen, C.J.; et al. Assessment of Pembrolizumab Therapy for the Treatment of Microsatellite Instability-High Gastric or Gastroesophageal Junction Cancer Among Patients in the KEYNOTE-059, KEYNOTE-061, and KEYNOTE-062 Clinical Trials. JAMA Oncol. 2021, 7, 895–902. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.T.; Cristescu, R.; Bass, A.J.; Kim, K.M.; Odegaard, J.I.; Kim, K.; Liu, X.Q.; Sher, X.; Jung, H.; Lee, M.; et al. Comprehensive molecular characterization of clinical responses to PD-1 inhibition in metastatic gastric cancer. Nat. Med. 2018, 24, 1449–1458. [Google Scholar] [CrossRef] [PubMed]
- Shitara, K.; Özgüroğlu, M.; Bang, Y.J.; Di Bartolomeo, M.; Mandalà, M.; Ryu, M.H.; Caglevic, C.; Chung, H.C.; Muro, K.; Van Cutsem, E.; et al. Molecular determinants of clinical outcomes with pembrolizumab versus paclitaxel in a randomized, open-label, phase III trial in patients with gastroesophageal adenocarcinoma. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2021, 32, 1127–1136. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.W.; Van Cutsem, E.; Bang, Y.J.; Fuchs, C.S.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; Castro, H.R.; Chao, J.; et al. Association of Tumor Mutational Burden with Efficacy of Pembrolizumab{plus minus}Chemotherapy as First-Line Therapy for Gastric Cancer in the Phase III KEYNOTE-062 Study. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2022, 28, 3489–3498. [Google Scholar] [CrossRef]
- Qi, C.; Gong, J.; Li, J.; Liu, D.; Qin, Y.; Ge, S.; Zhang, M.; Peng, Z.; Zhou, J.; Cao, Y.; et al. Claudin18.2-specific CAR T cells in gastrointestinal cancers: Phase 1 trial interim results. Nat. Med. 2022, 28, 1189–1198. [Google Scholar] [CrossRef]
Trial | Setting | Interventional Arm | Comparator | Outcomes | Comments |
---|---|---|---|---|---|
ATTRACTION-2 | Chemo-refractory | nivoluab | placebo | Positive for OS | - |
Keynote-061 | 2nd line | pembrolizumab | taxol | No difference for OS | Potential benefit in CPS ≥ 10 |
JAVELIN Gastric 300 | 3rd line | avelumab | Taxanes/irinotecan | No difference for OS | - |
Keynote-062 | 1st line | pembro or pembro+ chemo | Platinum chemo +placebo | Pembro non inferior to chemo in CPS ≥ 1 | Pembro numerically superior to chemo in CPS ≥ 10 |
JAVELIN Gastric 100 | 1st line maintenance | avelumab after chemo induction | Chemotherapy continuation | No difference for OS | Avelumab numerically superior in CPS (+) |
Checkmate-649 | 1st line (CPS ≥ 5) | nivo+ chemo or nivo-ipi | Platinum chemo | Nivo+chemo superior OS in CPS ≥ 5 | Nivo-ipi: no difference for OS |
ATTRACTION-4 | 1st line (Asian population) | Nivo + chemo | Platinum chemo + placebo | Nivo+ chemo superior for PFS regardless of PD-L1 expression | No difference in OS |
ORIENT-16 | 1st line in CPS ≥ 5 (Asian population) | Sintilimab + chemo | Platinum chemo + placebo | Sintilimab + chemo superior for OS in CPS ≥ 5 and all randomized patients | - |
Keynote-811 | 1st line HER2 positive | Pembro+trastuzumab + chemo | placebo+trastuzumab + chemo | Pembro arm superior for ORR | Very preliminary data |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Agnarelli, A.; Vella, V.; Samuels, M.; Papanastasopoulos, P.; Giamas, G. Incorporating Immunotherapy in the Management of Gastric Cancer: Molecular and Clinical Implications. Cancers 2022, 14, 4378. https://doi.org/10.3390/cancers14184378
Agnarelli A, Vella V, Samuels M, Papanastasopoulos P, Giamas G. Incorporating Immunotherapy in the Management of Gastric Cancer: Molecular and Clinical Implications. Cancers. 2022; 14(18):4378. https://doi.org/10.3390/cancers14184378
Chicago/Turabian StyleAgnarelli, Alessandro, Viviana Vella, Mark Samuels, Panagiotis Papanastasopoulos, and Georgios Giamas. 2022. "Incorporating Immunotherapy in the Management of Gastric Cancer: Molecular and Clinical Implications" Cancers 14, no. 18: 4378. https://doi.org/10.3390/cancers14184378
APA StyleAgnarelli, A., Vella, V., Samuels, M., Papanastasopoulos, P., & Giamas, G. (2022). Incorporating Immunotherapy in the Management of Gastric Cancer: Molecular and Clinical Implications. Cancers, 14(18), 4378. https://doi.org/10.3390/cancers14184378