The Roles of Immune Cells in Gastric Cancer: Anti-Cancer or Pro-Cancer?
Abstract
:Simple Summary
Abstract
1. Introduction
2. Prevalence and Incidence of Gastric Cancer Worldwide
3. Pathogenesis of Gastric Cancer
4. Functions of Immune Cells in Gastric Cancer
4.1. T Cells
4.2. B Cells
4.3. Macrophages
4.4. Natural Killer Cells
4.5. Dendritic Cells
5. Immune Checkpoints in Gastric Cancer
6. Molecular Subtypes and Immune Cells in Gastric Cancer
7. Immune Cell Response in Intestinal vs. Diffuse Gastric Cancer
8. Challenges to Associate the Immune System with Gastric Cancer
9. Conclusions
Author Contributions
Funding
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]
- Yusefi, A.R.; Bagheri Lankarani, K.; Bastani, P.; Radinmanesh, M.; Kavosi, Z. Risk factors for gastric cancer: A systematic review. Asian Pac. J. Cancer Prev. 2018, 19, 591–603. [Google Scholar] [PubMed]
- Necula, L.; Matei, L.; Dragu, D.; Neagu, A.I.; Mambet, C.; Nedeianu, S.; Bleotu, C.; Diaconu, C.C.; Chivu-Economescu, M. Recent advances in gastric cancer early diagnosis. World J. Gastroenterol. 2019, 25, 2029–2044. [Google Scholar] [CrossRef] [PubMed]
- Orditura, M.; Galizia, G.; Sforza, V.; Gambardella, V.; Fabozzi, A.; Laterza, M.M.; Andreozzi, F.; Ventriglia, J.; Savastano, B.; Mabilia, A.; et al. Treatment of gastric cancer. World J. Gastroenterol. 2014, 20, 1635–1649. [Google Scholar] [CrossRef] [PubMed]
- Swann, J.B.; Smyth, M.J. Immune surveillance of tumors. J. Clin. Investig. 2007, 117, 1137–1146. [Google Scholar] [CrossRef] [PubMed]
- Airoldi, I.; Di Carlo, E.; Cocco, C.; Sorrentino, C.; Fais, F.; Cilli, M.; D’Antuono, T.; Colombo, M.P.; Pistoia, V. Lack of Il12rb2 signaling predisposes to spontaneous autoimmunity and malignancy. Blood 2005, 106, 3846–3853. [Google Scholar] [CrossRef]
- Clementi, R.; Locatelli, F.; Dupré, L.; Garaventa, A.; Emmi, L.; Bregni, M.; Cefalo, G.; Moretta, A.; Danesino, C.; Comis, M.; et al. A proportion of patients with lymphoma may harbor mutations of the perforin gene. Blood 2005, 105, 4424–4428. [Google Scholar] [CrossRef]
- Mittal, D.; Gubin, M.M.; Schreiber, R.D.; Smyth, M.J. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr. Opin. Immunol. 2014, 27, 16–25. [Google Scholar] [CrossRef]
- Dighe, A.S.; Richards, E.; Old, L.J.; Schreiber, R.D. Enhanced in vivo growth and resistance to rejection of tumor cells expressing dominant negative IFN gamma receptors. Immunity 1994, 1, 447–456. [Google Scholar] [CrossRef]
- Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107–1111. [Google Scholar] [CrossRef] [PubMed]
- Müller-Hermelink, N.; Braumüller, H.; Pichler, B.; Wieder, T.; Mailhammer, R.; Schaak, K.; Ghoreschi, K.; Yazdi, A.; Haubner, R.; Sander, C.A.; et al. TNFR1 signaling and IFN-gamma signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell 2008, 13, 507–518. [Google Scholar] [CrossRef]
- Teng, M.W.; Galon, J.; Fridman, W.H.; Smyth, M.J. From mice to humans: Developments in cancer immunoediting. J. Clin. Investig. 2015, 125, 3338–3346. [Google Scholar] [CrossRef] [PubMed]
- Matsueda, S.; Graham, D.Y. Immunotherapy in gastric cancer. World J. Gastroenterol. 2014, 20, 1657–1666. [Google Scholar] [CrossRef] [PubMed]
- Slamon, D.J.; Leyland-Jones, B.; Shak, S.; Fuchs, H.; Paton, V.; Bajamonde, A.; Fleming, T.; Eiermann, W.; Wolter, J.; Pegram, M.; et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N. Engl. J. Med. 2001, 344, 783–792. [Google Scholar] [CrossRef] [PubMed]
- Jiang, J.; Xu, N.; Wu, C.; Deng, H.; Lu, M.; Li, M.; Xu, B.; Wu, J.; Wang, R.; Xu, J.; et al. Treatment of advanced gastric cancer by chemotherapy combined with autologous cytokine-induced killer cells. Anticancer Res. 2006, 26, 2237–2242. [Google Scholar] [PubMed]
- Global Burden of Disease 2019 Cancer Collaboration. Cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life years for 29 cancer groups from 2010 to 2019: A systematic analysis for the global burden of disease study 2019. JAMA Oncol. 2022, 8, 420–444. [Google Scholar] [CrossRef]
- Ferro, A.; Peleteiro, B.; Malvezzi, M.; Bosetti, C.; Bertuccio, P.; Levi, F.; Negri, E.; La Vecchia, C.; Lunet, N. Worldwide trends in gastric cancer mortality (1980–2011), with predictions to 2015, and incidence by subtype. Eur. J. Cancer 2014, 50, 1330–1344. [Google Scholar] [CrossRef]
- Mabe, K.; Inoue, K.; Kamada, T.; Kato, K.; Kato, M.; Haruma, K. Endoscopic screening for gastric cancer in Japan: Current status and future perspectives. Dig. Endosc. 2022, 34, 412–419. [Google Scholar] [CrossRef]
- Sukri, A.; Lopes, B.S.; Hanafiah, A. The emergence of multidrug-resistant Helicobacter pylori in Southeast Asia: A systematic review on the trends and intervention strategies using antimicrobial peptides. Antibiotics 2021, 10, 1061. [Google Scholar] [CrossRef]
- He, Y.; Wang, Y.; Luan, F.; Yu, Z.; Feng, H.; Chen, B.; Chen, W. Chinese and global burdens of gastric cancer from 1990 to 2019. Cancer Med. 2021, 10, 3461–3473. [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] [PubMed]
- Ning, F.L.; Lyu, J.; Pei, J.P.; Gu, W.J.; Zhang, N.N.; Cao, S.Y.; Zeng, Y.J.; Abe, M.; Nishiyama, K.; Zhang, C.D. The burden and trend of gastric cancer and possible risk factors in five Asian countries from 1990 to 2019. Sci. Rep. 2022, 12, 5980. [Google Scholar] [CrossRef] [PubMed]
- Colquhoun, A.; Arnold, M.; Ferlay, J.; Goodman, K.J.; Forman, D.; Soerjomataram, I. Global patterns of cardia and non-cardia gastric cancer incidence in 2012. Gut 2015, 64, 1881–1888. [Google Scholar] [CrossRef] [PubMed]
- Arnold, M.; Ferlay, J.; van Berge Henegouwen, M.I.; Soerjomataram, I. Global burden of oesophageal and gastric cancer by histology and subsite in 2018. Gut 2020, 69, 1564–1571. [Google Scholar] [CrossRef]
- Correa, P. Human gastric carcinogenesis: A multistep and multifactorial process--First American Cancer Society Award Lecture on Cancer Epidemiology and Prevention. Cancer Res. 1992, 52, 6735–6740. [Google Scholar]
- Correa, P.; Piazuelo, M.B. The gastric precancerous cascade. J. Dig. Dis. 2012, 13, 2–9. [Google Scholar] [CrossRef]
- Bakhoum, S.F.; Compton, D.A. Chromosomal instability and cancer: A complex relationship with therapeutic potential. J. Clin. Investig. 2012, 122, 1138–1143. [Google Scholar] [CrossRef]
- Ottini, L.; Falchetti, M.; Lupi, R.; Rizzolo, P.; Agnese, V.; Colucci, G.; Bazan, V.; Russo, A. Patterns of genomic instability in gastric cancer: Clinical implications and perspectives. Ann. Oncol. 2006, 17, 97–102. [Google Scholar] [CrossRef]
- Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature 2014, 51, 202–209. [Google Scholar]
- Weiss, M.M.; Kuipers, E.J.; Postma, C.; Snijders, A.M.; Pinkel, D.; Meuwissen, S.G.; Albertson, D.; Meijer, G.A. Genomic alterations in primary gastric adenocarcinomas correlate with clinicopathological characteristics and survival. Cell Oncol. 2004, 26, 307–317. [Google Scholar] [CrossRef]
- Weiss, M.M.; Kuipers, E.J.; Postma, C.; Snijders, A.M.; Siccama, I.; Pinkel, D.; Westerga, J.; Meuwissen, S.G.; Albertson, D.G.; Meijer, G.A. Genomic profiling of gastric cancer predicts lymph node status and survival. Oncogene 2003, 22, 1872–1879. [Google Scholar] [CrossRef] [PubMed]
- Wirtz, H.C.; Müller, W.; Noguchi, T.; Scheven, M.; Rüschoff, J.; Hommel, G.; Gabbert, H.E. Prognostic value and clinicopathological profile of microsatellite instability in gastric cancer. Clin. Cancer Res. 1998, 4, 1749–1754. [Google Scholar] [PubMed]
- An, J.Y.; Kim, H.; Cheong, J.H.; Hyung, W.J.; Kim, H.; Noh, S.H. Microsatellite instability in sporadic gastric cancer: Its prognostic role and guidance for 5-FU based chemotherapy after R0 resection. Int. J. Cancer 2012, 131, 505–511. [Google Scholar] [CrossRef] [PubMed]
- Zhu, L.; Li, Z.; Wang, Y.; Zhang, C.; Liu, Y.; Qu, X. Microsatellite instability and survival in gastric cancer: A systematic review and meta-analysis. Mol. Clin. Oncol. 2015, 3, 699–705. [Google Scholar] [CrossRef] [PubMed]
- Kang, C.; Song, J.J.; Lee, J.; Kim, M.Y. Epigenetics: An emerging player in gastric cancer. World J. Gastroenterol. 2014, 20, 6433–6447. [Google Scholar] [CrossRef]
- Illingworth, R.S.; Bird, A.P. CpG islands—‘A rough guide’. FEBS Lett. 2009, 583, 1713–1720. [Google Scholar] [CrossRef]
- Esteller, M. CpG island hypermethylation and tumor suppressor genes: A booming present, a brighter future. Oncogene 2002, 21, 5427–5440. [Google Scholar] [CrossRef]
- Zong, L.; Seto, Y. CpG island methylator phenotype, Helicobacter pylori, Epstein-Barr virus, and microsatellite instability and prognosis in gastric cancer: A systematic review and meta-analysis. PLoS ONE 2014, 9, e86097. [Google Scholar] [CrossRef]
- Loh, M.; Liem, N.; Vaithilingam, A.; Lim, P.L.; Sapari, N.S.; Elahi, E.; Mok, Z.Y.; Cheng, C.L.; Yan, B.; Pang, B.; et al. DNA methylation subgroups and the CpG island methylator phenotype in gastric cancer: A comprehensive profiling approach. BMC Gastroenterol. 2014, 14, 55. [Google Scholar] [CrossRef]
- Maekita, T.; Nakazawa, K.; Mihara, M.; Nakajima, T.; Yanaoka, K.; Iguchi, M.; Arii, K.; Kaneda, A.; Tsukamoto, T.; Tatematsu, M.; et al. High levels of aberrant DNA methylation in Helicobacter pylori-infected gastric mucosae and its possible association with gastric cancer risk. Clin. Cancer Res. 2006, 12, 989–995. [Google Scholar] [CrossRef]
- Ropero, S.; Esteller, M. The role of histone deacetylases (HDACs) in human cancer. Mol. Oncol. 2007, 1, 19–25. [Google Scholar] [CrossRef] [PubMed]
- Wisnieski, F.; Calcagno, D.Q.; Leal, M.F.; Chen, E.S.; Gigek, C.O.; Santos, L.C.; Pontes, T.B.; Rasmussen, L.T.; Payão, S.L.; Assumpção, P.P.; et al. Differential expression of histone deacetylase and acetyltransferase genes in gastric cancer and their modulation by trichostatin A. Tumour Biol. 2014, 35, 6373–6381. [Google Scholar] [CrossRef] [PubMed]
- Lagger, G.; Doetzlhofer, A.; Schuettengruber, B.; Haidweger, E.; Simboeck, E.; Tischler, J.; Chiocca, S.; Suske, G.; Rotheneder, H.; Wintersberger, E.; et al. The tumor suppressor p53 and histone deacetylase 1 are antagonistic regulators of the cyclin-dependent kinase inhibitor p21/WAF1/CIP1 gene. Mol. Cell Biol. 2003, 23, 2669–2679. [Google Scholar] [CrossRef] [PubMed]
- Song, J.; Noh, J.H.; Lee, J.H.; Eun, J.W.; Ahn, Y.M.; Kim, S.Y.; Lee, S.H.; Park, W.S.; Yoo, N.J.; Lee, J.Y.; et al. Increased expression of histone deacetylase 2 is found in human gastric cancer. APMIS 2005, 113, 264–268. [Google Scholar] [CrossRef]
- Ding, S.Z.; Fischer, W.; Kaparakis-Liaskos, M.; Liechti, G.; Merrell, D.S.; Grant, P.A.; Ferrero, R.L.; Crowe, S.E.; Haas, R.; Hatakeyama, M.; et al. Helicobacter pylori-induced histone modification, associated gene expression in gastric epithelial cells, and its implication in pathogenesis. PLoS ONE 2010, 5, e9875. [Google Scholar] [CrossRef]
- Fehri, L.F.; Rechner, C.; Janssen, S.; Mak, T.N.; Holland, C.; Bartfeld, S.; Brüggemann, H.; Meyer, T.F. Helicobacter pylori-induced modification of the histone H3 phosphorylation status in gastric epithelial cells reflects its impact on cell cycle regulation. Epigenetics 2009, 4, 577–586. [Google Scholar] [CrossRef]
- Ying, H.Y.; Yu, B.W.; Yang, Z.; Yang, S.S.; Bo, L.H.; Shan, X.Y.; Wang, H.J.; Zhu, Y.J.; Wu, X.S. Interleukin-1B 31 C>T polymorphism combined with Helicobacter pylori-modified gastric cancer susceptibility: Evidence from 37 studies. J. Cell Mol. Med. 2016, 20, 526–536. [Google Scholar] [CrossRef]
- Pohjanen, V.M.; Koivurova, O.P.; Mäkinen, J.M.; Karhukorpi, J.M.; Joensuu, T.; Koistinen, P.O.; Valtonen, J.M.; Niemelä, S.E.; Karttunen, R.A.; Karttunen, T.J. Interleukin 6 gene polymorphism -174 is associated with the diffuse type gastric carcinoma. Genes Chromosomes Cancer 2013, 52, 976–982. [Google Scholar] [CrossRef]
- Lee, K.E.; Khoi, P.N.; Xia, Y.; Park, J.S.; Joo, Y.E.; Kim, K.K.; Choi, S.Y.; Jung, Y.D. Helicobacter pylori and interleukin-8 in gastric cancer. World J. Gastroenterol. 2013, 19, 8192–8202. [Google Scholar] [CrossRef]
- Long, Z.W.; Yu, H.M.; Wang, Y.N.; Liu, D.; Chen, Y.Z.; Zhao, Y.X.; Bai, L. Association of IL-17 polymorphisms with gastric cancer risk in Asian populations. World J. Gastroenterol. 2015, 21, 5707–5718. [Google Scholar] [CrossRef]
- Rokkas, T.; Sechopoulos, P.; Pistiolas, D.; Kothonas, F.; Margantinis, G.; Koukoulis, G. Population differences concerning TNF-α gene polymorphisms in gastric carcinogenesis based on meta-analysis. Ann. Gastroenterol. 2014, 27, 139–1348. [Google Scholar] [PubMed]
- Bailey, S.R.; Nelson, M.H.; Himes, R.A.; Li, Z.; Mehrotra, S.; Paulos, C.M. Th17 cells in cancer: The ultimate identity crisis. Front. Immunol. 2014, 5, 276. [Google Scholar] [CrossRef] [PubMed]
- Larussa, T.; Leone, I.; Suraci, E.; Imeneo, M.; Luzza, F. Helicobacter pylori and T Helper Cells: Mechanisms of Immune Escape and Tolerance. J. Immunol. Res. 2015, 2015, 981328. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, M.H. Th9 cells: Differentiation and disease. Immunol. Rev. 2013, 252, 104–115. [Google Scholar] [CrossRef]
- Eisenstein, E.M.; Williams, C.B. The T(reg)/Th17 cell balance: A new paradigm for autoimmunity. Pediatr Res. 2009, 65, 26R–31R. [Google Scholar] [CrossRef]
- Eyerich, S.; Eyerich, K.; Pennino, D.; Carbone, T.; Nasorri, F.; Pallotta, S.; Cianfarani, F.; Odorisio, T.; Traidl-Hoffmann, C.; Behrendt, H.; et al. Th22 cells represent a distinct human T cell subset involved in epidermal immunity and remodeling. J. Clin. Investig. 2009, 119, 3573–3585. [Google Scholar] [CrossRef]
- Thompson, E.D.; Zahurak, M.; Murphy, A.; Cornish, T.; Cuka, N.; Abdelfatah, E.; Yang, S.; Duncan, M.; Ahuja, N.; Taube, J.M.; et al. Patterns of PD-L1 expression and CD8 T cell infiltration in gastric adenocarcinomas and associated immune stroma. Gut 2017, 66, 794–801. [Google Scholar] [CrossRef]
- Francisco, L.M.; Sage, P.T.; Sharpe, A.H. The PD-1 pathway in tolerance and autoimmunity. Immunol. Rev. 2010, 236, 219–242. [Google Scholar] [CrossRef]
- Kuzushima, K.; Nakamura, S.; Nakamura, T.; Yamamura, Y.; Yokoyama, N.; Fujita, M.; Kiyono, T.; Tsurumi, T. Increased frequency of antigen-specific CD8(+) cytotoxic T lymphocytes infiltrating an Epstein-Barr virus-associated gastric carcinoma. J. Clin. Investig. 1999, 104, 163–171. [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]
- Tan, M.P.; Pedersen, J.; Zhan, Y.; Lew, A.M.; Pearse, M.J.; Wijburg, O.L.; Strugnell, R.A. CD8+ T cells are associated with severe gastritis in Helicobacter pylori-infected mice in the absence of CD4+ T cells. Infect. Immun. 2008, 76, 1289–1297. [Google Scholar] [CrossRef] [PubMed]
- Smythies, L.E.; Waites, K.B.; Lindsey, J.R.; Harris, P.R.; Ghiara, P.; Smith, P.D. Helicobacter pylori-induced mucosal inflammation is Th1 mediated and exacerbated in IL-4, but not IFN-gamma, gene-deficient mice. J. Immunol. 2000, 165, 1022–1029. [Google Scholar] [CrossRef] [PubMed]
- Akhiani, A.A.; Pappo, J.; Kabok, Z.; Schön, K.; Gao, W.; Franzén, L.E.; Lycke, N. Protection against Helicobacter pylori infection following immunization is IL-12-dependent and mediated by Th1 cells. J. Immunol. 2002, 169, 6977–6984. [Google Scholar] [CrossRef] [PubMed]
- Yang, P.; Qiu, G.; Wang, S.; Su, Z.; Chen, J.; Wang, S.; Kong, F.; Lu, L.; Ezaki, T.; Xu, H. The mutations of Th1 cell-specific T-box transcription factor may be associated with a predominant Th2 phenotype in gastric cancers. Int. J. Immunogenet. 2010, 37, 111–1115. [Google Scholar] [CrossRef]
- Corthay, A. How do regulatory T cells work? Scand. J. Immunol. 2009, 70, 326–336. [Google Scholar] [CrossRef]
- Ichihara, F.; Kono, K.; Takahashi, A.; Kawaida, H.; Sugai, H.; Fujii, H. Increased populations of regulatory T cells in peripheral blood and tumor-infiltrating lymphocytes in patients with gastric and esophageal cancers. Clin. Cancer Res. 2003, 9, 4404–4408. [Google Scholar]
- Kim, K.J.; Lee, K.S.; Cho, H.J.; Kim, Y.H.; Yang, H.K.; Kim, W.H.; Kang, G.H. Prognostic implications of tumor-infiltrating FoxP3+ regulatory T cells and CD8+ cytotoxic T cells in microsatellite-unstable gastric cancers. Hum. Pathol. 2014, 45, 285–293. [Google Scholar] [CrossRef]
- Nagase, H.; Takeoka, T.; Urakawa, S.; Morimoto-Okazawa, A.; Kawashima, A.; Iwahori, K.; Takiguchi, S.; Nishikawa, H.; Sato, E.; Sakaguchi, S.; et al. ICOS+ Foxp3+ TILs in gastric cancer are prognostic markers and effector regulatory T cells associated with Helicobacter pylori. Int. J. Cancer 2017, 140, 686–695. [Google Scholar] [CrossRef]
- Li, Q.; Li, Q.; Chen, J.; Liu, Y.; Zhao, X.; Tan, B.; Ai, J.; Zhang, Z.; Song, J.; Shan, B. Prevalence of Th17 and Treg cells in gastric cancer patients and its correlation with clinical parameters. Oncol. Rep. 2013, 30, 1215–1222. [Google Scholar] [CrossRef]
- Shen, P.; Fillatreau, S. Antibody-independent functions of B cells: A focus on cytokines. Nat. Rev. Immunol. 2015, 15, 441–451. [Google Scholar] [CrossRef]
- Schwartz, M.; Zhang, Y.; Rosenblatt, J.D. B cell regulation of the anti-tumor response and role in carcinogenesis. J. Immunother Cancer 2016, 4, 40. [Google Scholar] [CrossRef] [PubMed]
- Sukri, A.; Hanafiah, A.; Kosai, N.R.; Mohamed Taher, M.; Mohamed Rose, I. Surface Antigen Profiling of Helicobacter pylori-Infected and -Uninfected Gastric Cancer Cells Using Antibody Microarray. Helicobacter 2016, 21, 417–427. [Google Scholar] [CrossRef] [PubMed]
- Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef] [PubMed]
- Aderem, A.; Underhill, D.M. Mechanisms of phagocytosis in macrophages. Annu. Rev. Immunol. 1999, 17, 593–623. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.H.; Lee, W.J.; Hua, K.T.; Kuo, M.L.; Lin, M.T. Macrophage infiltration induces gastric cancer invasiveness by activating the β-catenin pathway. PLoS ONE 2015, 10, e0134122. [Google Scholar] [CrossRef]
- Yamaguchi, T.; Fushida, S.; Yamamoto, Y.; Tsukada, T.; Kinoshita, J.; Oyama, K.; Miyashita, T.; Tajima, H.; Ninomiya, I.; Munesue, S.; et al. Tumor-associated macrophages of the M2 phenotype contribute to progression in gastric cancer with peritoneal dissemination. Gastric Cancer 2016, 19, 1052–1065. [Google Scholar] [CrossRef]
- Lund, S.A.; Giachelli, C.M.; Scatena, M. The role of osteopontin in inflammatory processes. J. Cell Commun. Signal 2009, 3, 311–322. [Google Scholar] [CrossRef]
- Lin, C.N.; Wang, C.J.; Chao, Y.J.; Lai, M.D.; Shan, Y.S. The significance of the co-existence of osteopontin and tumor-associated macrophages in gastric cancer progression. BMC Cancer 2015, 15, 128. [Google Scholar] [CrossRef]
- Cardoso, A.P.; Pinto, M.L.; Pinto, A.T.; Oliveira, M.I.; Pinto, M.T.; Gonçalves, R.; Relvas, J.B.; Figueiredo, C.; Seruca, R.; Mantovani, A.; et al. Macrophages stimulate gastric and colorectal cancer invasion through EGFR Y(1086), c-Src, Erk1/2 and Akt phosphorylation and smallGTPase activity. Oncogene 2014, 33, 2123–2133. [Google Scholar] [CrossRef]
- Tan, G.M.; Looi, C.Y.; Fernandez, K.C.; Vadivelu, J.; Loke, M.F.; Wong, W.F. Suppression of cell division-associated genes by Helicobacter pylori attenuates proliferation of RAW264.7 monocytic macrophage cells. Sci. Rep. 2015, 5, 11046. [Google Scholar] [CrossRef]
- Cerwenka, A.; Lanier, L.L. Natural killer cells, viruses and cancer. Nat. Rev. Immunol. 2001, 1, 41–49. [Google Scholar] [CrossRef] [PubMed]
- Nausch, N.; Cerwenka, A. NKG2D ligands in tumor immunity. Oncogene 2008, 27, 5944–5958. [Google Scholar] [CrossRef] [PubMed]
- Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef] [PubMed]
- Ishigami, S.; Natsugoe, S.; Tokuda, K.; Nakajo, A.; Che, X.; Iwashige, H.; Aridome, K.; Hokita, S.; Aikou, T. Prognostic value of intratumoral natural killer cells in gastric carcinoma. Cancer 2000, 88, 577–583. [Google Scholar] [CrossRef]
- Takeuchi, H.; Maehara, Y.; Tokunaga, E.; Koga, T.; Kakeji, Y.; Sugimachi, K. Prognostic significance of natural killer cell activity in patients with gastric carcinoma: A multivariate analysis. Am. J. Gastroenterol. 2001, 96, 574–578. [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 2013, 16, 473–479. [Google Scholar] [CrossRef]
- 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 2015, 5, e1069936. [Google Scholar] [CrossRef]
- Banchereau, J.; Steinman, R.M. Dendritic cells and the control of immunity. Nature 1998, 392, 245–252. [Google Scholar] [CrossRef]
- Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 15, 3–20. [Google Scholar] [CrossRef]
- Krempski, J.; Karyampudi, L.; Behrens, M.D.; Erskine, C.L.; Hartmann, L.; Dong, H.; Goode, E.L.; Kalli, K.R.; Knutson, K.L. Tumor-infiltrating programmed death receptor-1+ dendritic cells mediate immune suppression in ovarian cancer. J. Immunol. 2011, 186, 6905–6913. [Google Scholar] [CrossRef]
- Hargadon, K.M. Tumor-altered dendritic cell function: Implications for anti-tumor immunity. Front. Immunol. 2013, 4, 192. [Google Scholar] [CrossRef] [PubMed]
- Okita, Y.; Tanaka, H.; Ohira, M.; Muguruma, K.; Kubo, N.; Watanabe, M.; Fukushima, W.; Hirakawa, K. Role of tumor-infiltrating CD11b+ antigen-presenting cells in the progression of gastric cancer. J. Surg. Res. 2014, 186, 192–200. [Google Scholar] [CrossRef] [PubMed]
- He, X.; Xu, C. Immune checkpoint signaling and cancer immunotherapy. Cell Res. 2020, 30, 660–669. [Google Scholar] [CrossRef] [PubMed]
- McBride, M.A.; Patil, T.K.; Bohannon, J.K.; Hernandez, A.; Sherwood, E.R.; Patil, N.K. Immune checkpoints: Novel therapeutic targets to attenuate sepsis-induced immunosuppression. Front. Immunol. 2021, 11, 624272. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.B.; Li, P.; Liu, X.L.; Zheng, Q.L.; Ma, Y.B.; Zhao, Y.J.; Xie, J.W.; Lin, J.X.; Lu, J.; Chen, Q.Y.; et al. An immune checkpoint score system for prognostic evaluation and adjuvant chemotherapy selection in gastric cancer. Nat. Commun. 2020, 11, 6352. [Google Scholar] [CrossRef]
- Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar]
- Joshi, S.S.; Badgwell, B.D. Current treatment and recent progress in gastric cancer. CA Cancer J. Clin. 2021, 71, 264–279. [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]
- Muro, K.; Chung, H.C.; Shankaran, V.; Geva, R.; Catenacci, D.; Gupta, S.; Eder, J.P.; Golan, T.; Le, D.T.; Burtness, B.; 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, 717–726. [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. Ocol. 2018, 29, 2052–2060. [Google Scholar] [CrossRef]
- 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]
- Sohn, B.H.; Hwang, J.E.; Jang, H.J.; Lee, H.S.; Oh, S.C.; Shim, J.J.; Lee, K.W.; Kim, E.H.; Yim, S.Y.; Lee, S.H.; et al. Clinical significance of four molecular subtypes of gastric cancer identified by the cancer genome atlas project. Clin. Cancer Res. 2017, 23, 4441–4449. [Google Scholar] [CrossRef] [PubMed]
- Mansuri, N.; Birkman, E.M.; Heuser, V.D.; Lintunen, M.; Ålgars, A.; Sundström, J.; Ristamäki, R.; Lehtinen, L.; Carpén, O. Association of tumor-infiltrating T lymphocytes with intestinal-type gastric cancer molecular subtypes and outcome. Virchows Archiv. 2021, 478, 707–717. [Google Scholar] [CrossRef] [PubMed]
- Kim, T.S.; da Silva, E.; Coit, D.G.; Tang, L.H. Intratumoral immune response to gastric cancer varies by molecular and histologic subtype. Am. J. Surg. Pathol. 2019, 43, 851–860. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Wang, X. Identification of gastric cancer subtypes based on pathway clustering. NPJ Precis Oncol. 2021, 5, 46. [Google Scholar] [CrossRef]
- Wu, D.; Feng, M.; Shen, H.; Shen, X.; Hu, J.; Liu, J.; Yang, Y.; Li, Y.; Yang, M.; Wang, W.; et al. Prediction of two molecular subtypes of gastric cancer based on immune signature. Front. Genet. 2022, 12, 793494. [Google Scholar] [CrossRef]
- Chen, Y.; Sun, Z.; Chen, W.; Liu, C.; Chai, R.; Ding, J.; Liu, W.; Feng, X.; Zhou, J.; Shen, X.; et al. The immune subtypes and landscape of gastric cancer and to predict based on the whole-slide images using deep learning. Front. Immunol. 2021, 12, 685992. [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]
- McLean, M.H.; El-Omar, E.M. Genetics of gastric cancer. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 664–674. [Google Scholar] [CrossRef]
- Chen, Y.C.; Fang, W.L.; Wang, R.F.; Liu, C.A.; Yang, M.H.; Lo, S.S.; Wu, C.W.; Li, A.F.; Shyr, Y.M.; Huang, K.H. Clinicopathological variation of Lauren classification in gastric cancer. Pathol. Oncol. Res. 2016, 22, 197–202. [Google Scholar] [CrossRef]
- Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef]
- Kim, J.; Park, C.; Kim, K.H.; Kim, E.H.; Kim, H.; Woo, J.K.; Seong, J.K.; Nam, K.T.; Lee, Y.C.; Cho, S.Y. Single-cell analysis of gastric pre-cancerous and cancer lesions reveals cell lineage diversity and intratumoral heterogeneity. NPJ Precis Oncol. 2022, 6, 9. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Wu, X.; Zhang, C.; Wang, J.; Fei, G.; Di, X.; Lu, X.; Feng, L.; Cheng, S.; Yang, A. Dissecting expression profiles of gastric precancerous lesions and early gastric cancer to explore crucial molecules in intestinal-type gastric cancer tumorigenesis. J. Pathol. 2020, 251, 135–146. [Google Scholar] [CrossRef] [PubMed]
- Pernot, S.; Terme, M.; Radosevic-Robin, N.; Castan, F.; Badoual, C.; Marcheteau, E.; Penault-Llorca, F.; Bouche, O.; Bennouna, J.; Francois, E.; et al. Infiltrating and peripheral immune cell analysis in advanced gastric cancer according to the Lauren classification and its prognostic significance. Gastric Cancer 2020, 23, 73–81. [Google Scholar] [CrossRef]
- Sukri, A.; Salleh, M.Z.; Masimirembwa, C.; Teh, L.K. A systematic review on the cost effectiveness of pharmacogenomics in developing countries: Implementation challenges. Pharm. J. 2022, 22, 147–159. [Google Scholar] [CrossRef]
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
Sukri, A.; Hanafiah, A.; Kosai, N.R. The Roles of Immune Cells in Gastric Cancer: Anti-Cancer or Pro-Cancer? Cancers 2022, 14, 3922. https://doi.org/10.3390/cancers14163922
Sukri A, Hanafiah A, Kosai NR. The Roles of Immune Cells in Gastric Cancer: Anti-Cancer or Pro-Cancer? Cancers. 2022; 14(16):3922. https://doi.org/10.3390/cancers14163922
Chicago/Turabian StyleSukri, Asif, Alfizah Hanafiah, and Nik Ritza Kosai. 2022. "The Roles of Immune Cells in Gastric Cancer: Anti-Cancer or Pro-Cancer?" Cancers 14, no. 16: 3922. https://doi.org/10.3390/cancers14163922
APA StyleSukri, A., Hanafiah, A., & Kosai, N. R. (2022). The Roles of Immune Cells in Gastric Cancer: Anti-Cancer or Pro-Cancer? Cancers, 14(16), 3922. https://doi.org/10.3390/cancers14163922