The dynamic change of immune and non-immune cells during
Abstract
Helicobacter pylori infection is identified as a primary risk factor for gastric cancer (GC). Chronic inflammation is usually induced by H. pylori infection and accompanied by inherent immune disorders. According to Correa’s model, gastritis could progress to premalignant lesions, such as intestinal metaplasia and dysplasia, and ultimately GC. The interaction of H. pylori with the gastric mucosa leads to the recruitment of immune cells, including dendritic (DC) cells, natural killer (NK) cells, and T and B lymphocytes, and triggers inflammatory response with cytokine production, which results in the pathogenesis of stomach. The balance between inflammation and immunity is important to gastric cancer development. However, the dynamic change of immune response during the transition from normal to metaplasia to dysplasia and GC is largely undefined. In this review, we summarized the involvement of key immune cells during GC progression, aiming to help identify inflection points and associated biomarkers for early GC detection, diagnosis, and therapies.
Keywords
- Helicobacter pylori
- gastric cancer
- immune response
- diagnosis
- therapy
1. Introduction
The lifetime risk of gastric cancer in patients with
2. H. pylori infection associated with T cells
2.1 T helper (Th) cell-mediated gastric mucosal inflammation
CD4+ T cells play a critical role in the immune response elicited during
Th17, a newly identified CD4+ T-cell subtype expressing IL-17A, IL-17F, IL-21, and the chemokine CCL20, as well as the transcription factor RORγt, is involved in the gastric mucosal inflammatory response to
Th22 cells and IL-22 play a proinflammatory role in infectious disease [10]. After
Although
2.2 Regulatory T-cell mediated gastric mucosal tolerance
Regulatory T cells (Tregs) are characterized by high levels of CD25, low levels of CD127, and inhibitory cytokines (e.g., IL-10 and the transcription factor Foxp3). They are essential in regulating cellular immune response and maintaining autoimmune tolerance. Tregs inhibit antigen-induced immune reactions, and disordered Treg activation induced by microbial antigens may be a mechanism by which
Tregs, CD4+CD25+Foxp3+ T cells, are essential in regulating the cellular immune response and maintaining autoimmune tolerance. Tregs negatively regulate the immune response mainly in two ways: one is to inhibit the activation of the target cells directly, and the other is to secrete cytokines, such as TGF-β and IL-10, to inhibit the immune response. In addition, Tregs can hinder the immune response induced by antigens and antibodies, and the abnormal activation of Tregs induced by microbial antigens may be one of the mechanisms by which
Although
2.3 Cytotoxic T-lymphocyte-mediated gastric mucosal inflammation
3. H. pylori infection associated with B cells
The primary function of B cells is to produce antibodies to mediate the humoral immune response. When
4. H. pylori infection associated with NK cells
NK cells are a class of nonspecific immune lymphocytes different from T and B cells and directly kill target cells. NK cells regulate the host’s immune function by releasing inflammatory factors, such as IFN-γ and TNF-α. Some cytokines, such as IL-12 and TNF-α, can regulate NK cell function and induce NK cells to produce IFN-γ. NK cells are the primary effector cells exerting an intrinsic immune response in the gastric mucosa against
5. H. pylori infection associated with dendritic cells
In DCs infected with
6. H. pylori infection associated with macrophages
Macrophages are critical immune cells in the defense of gastric mucosal tissues against
7. H. pylori infection associated with immune molecules
We will elaborate on the membrane molecular, cytokine, and signal pathway related to
Gastric cancer | ||
Cell types | CD4+T cell, CD8+T cell, NK cell, B cell, Macrophage, DC | CD4+T cell, CD8+T cell, B cell, Tumor-Associated Macrophages |
Molecules | TLR2, TLR, TLR10, B7H3, SOCS3, CCR2, CXCL2, MMP10, ITGB1 | PD-L1, FGFR4, MMPs, CTLA-4, CD44 |
IL23, IL33, BAFF, T cell derived cytokines | IL6, IL-10, IL-17A | |
STAT3, NF-κB, NLRP3, MyD88 | STAT1, STAT3, NF-κB |
A search team found a new regulatory mechanism employed by
8. Dynamic changes in gastric cancer
Chronic
Compared with the normal tissue, Treg cells in gastric cancer increased remarkably [49]. However, relevant transcriptional regulators still need more research to identify [50].
Kynureninase (KYNU), a biomarker of tumor-associated macrophages, is related to the immunosuppressive microenvironment in gastric cancer [51], suggesting the involvement of macrophages in GC. Moreover, converting M2-type TAMs back into M1-type (reprogramming of TAMs) is effective in antitumor therapy in tumor microenvironment (TME) [52]. Cyclase-associated protein 2 (CAP2) promotes gastric cancer metastasis by mediating the interaction between tumor cells and tumor-associated macrophages. M2 macrophage-derived TGFβ1 can influence the activation of M2 macrophage by a TGF β1/JUN/CAP2 positive-feedback loop [53].
Fibroblast growth factor receptors (FGFRs) are a gene family of transmembrane tyrosine kinase receptors that activate critical downstream signaling pathways [61]. FGFR4 overexpression is relevant to the biology of gastric cancer. Also, FGFR4 protects gastric cancer cells from apoptosis during
9. Conclusion
Recently, there has been prominent progress in accessing the pathogenesis, diagnosis, and treatment of
Acknowledgments
This work was supported by the National Natural Science Foundation of China (Grant Number 82270592).
Authorship
W.F. conceived the proposal. M.Z. prepared the manuscript. W.F., J.Z., and S.D. revised the manuscript.
Disclosures
The authors declare no conflicts of interest.
Abbreviations
activating factor | |
cytotoxin-associated gene A | |
cag pathogenicity island | |
cyclase-associated protein 2 | |
chemokine receptor 6 | |
cytotoxic T lymphocyte | |
dendritic cell | |
extracellular signal-regulated kinases | |
fibroblast growth factor receptors | |
gastric cancer | |
γ-glutamyl transpeptidase | |
human monocyte-derived dendritic cells | |
interferon | |
interleukins | |
interleukin-1 receptor (IL-1R)-associated kinase | |
kynureninase | |
mucosa-associated lymphoid tissue | |
myeloid-derived suppressor cells | |
matrix metalloproteinases | |
myeloid differentiation primary response 88 | |
NLR family pyrin domain containing 3 | |
nucleotide-binding oligomerization domain-containing protein 1 | |
natural killer | |
pathogen-associated molecular patterns | |
peripheral blood mononuclear cells | |
tumor-associated macrophages | |
transforming growth factor β | |
T helper | |
toll-like receptor | |
tumor microenvironment | |
regulatory T cells | |
tissue-resident memory (TRM) phenotype; | |
urease subunit B | |
vacuolar cytotoxin A. |
References
- 1.
Liou J-M, Malfertheiner P, Lee Y-C, Sheu B-S, Sugano K, Cheng H-C, et al. Screening and eradication of Helicobacter pylori for gastric cancer prevention: The Taipei global consensus. Gut. 2020;69 :2093-2112. DOI: 10.1136/gutjnl-2020-322368 - 2.
de Martel C, Georges D, Bray F, Ferlay J, Clifford GM. Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. The Lancet Global Health. 2020; 8 :e180-e190. DOI: 10.1016/S2214-109X(19)30488-7 - 3.
Mattapallil JJ, Dandekar S, Canfield DR, Solnick JV. A predominant Th1 type of immune response is induced early during acute Helicobacter pylori infection in rhesus macaques. Gastroenterology. 2000;118 :307-315. DOI: 10.1016/S0016-5085(00)70213-7 - 4.
Bimczok D, Clements RH, Waites KB, Novak L, Eckhoff DE, Mannon PJ, et al. Human primary gastric dendritic cells induce a Th1 response to H. pylori . Mucosal Immunology. 2010;3 :260-269. DOI: 10.1038/mi.2010.10 - 5.
Della, Bella C, Soluri MF, Puccio S, Benagiano M, Grassi A, Bitetti J, et al. The Helicobacter pylori CagY protein drives gastric Th1 and Th17 inflammation and B cell proliferation in gastric MALT lymphoma. IJMS. 2021;22 :9459. DOI: 10.3390/ijms22179459 - 6.
Paydarnia N, Mansoori B, Esmaeili D, Kazemi T, Aghapour M, Hajiasgharzadeh K, et al. Helicobacter pylori recombinant CagA regulates Th1/Th2 balance in a BALB/c murine model. Advanced Pharmaceutical Bulletin. 2020;10 :264-270. DOI: 10.34172/apb.2020.031 - 7.
DeLyria ES, Redline RW, Blanchard TG. Vaccination of mice against H. Pylori induces a strong Th-17 response and immunity that is neutrophil dependent. Gastroenterology. 2009;136 :247-256. DOI: 10.1053/j.gastro.2008.09.017 - 8.
Serrano C, Wright SW, Bimczok D, Shaffer CL, Cover TL, Venegas A, et al. Downregulated Th17 responses are associated with reduced gastritis in Helicobacter pylori –infected children. Mucosal Immunology. 2013;6 :950-959. DOI: 10.1038/mi.2012.133 - 9.
Dong C. Cytokine regulation and function in T cells. Annual Review of Immunology. 2021; 39 :51-76. DOI: 10.1146/annurev-immunol-061020-053702 - 10.
Zhuang Y, Cheng P, Liu X, Peng L, Li B, Wang T, et al. A pro-inflammatory role for Th22 cells in Helicobacter pylori -associated gastritis. Gut. 2015;64 :1368-1378. DOI: 10.1136/gutjnl-2014-307020 - 11.
Aujla SJ, Chan YR, Zheng M, Fei M, Askew DJ, Pociask DA, et al. IL-22 mediates mucosal host defense against gram-negative bacterial pneumonia. Nature Medicine. 2008; 14 :275-281. DOI: 10.1038/nm1710 - 12.
Sonnenberg GF, Nair MG, Kirn TJ, Zaph C, Fouser LA, Artis D. Pathological versus protective functions of IL-22 in airway inflammation are regulated by IL-17A. Journal of Experimental Medicine. 2010; 207 :1293-1305. DOI: 10.1084/jem.20092054 - 13.
Oster P, Vaillant L, McMillan B, Velin D. The efficacy of cancer immunotherapies is compromised by Helicobacter pylori infection. Frontiers in Immunology. 2022;13 :899161. DOI: 10.3389/fimmu.2022.899161 - 14.
Michetti P, Svennerholm A. Helicobacter pylori – inflammation, immunity and vaccines. Helicobacter. 2003;8 :31-35. DOI: 10.1046/j.1523-5378.2003.00164.x - 15.
Cho KY, Cho MS, Seo JW. FOXP3+ regulatory T cells in children with Helicobacter pylori infection. Pediatric and Developmental Pathology. 2012;15 :118-126. DOI: 10.2350/11-06-1046-OA.1 - 16.
Bagheri N, Azadegan-Dehkordi F, Rahimian G, Rafieian-Kopaei M, Shirzad H. Role of regulatory T-cells in different clinical expressions of Helicobacter pylori infection. Archives of Medical Research. 2016;47 :245-254. DOI: 10.1016/j.arcmed.2016.07.013 - 17.
Leake I. Helicobacter pylori induces changes in regulatory T cells. Nature Reviews. Gastroenterology & Hepatology. 2014;11 :141-141. DOI: 10.1038/nrgastro.2014.13 - 18.
Arienti C, Pignatta S, Tesei A. Epidermal growth factor receptor family and its role in gastric cancer. Frontiers in Oncology. 2019; 9 :1308. DOI: 10.3389/fonc.2019.01308 - 19.
Bamford KB, Fan X, Crowe SE, Leary JF, Gourley WK, Luthra GK, et al. Lymphocytes in the human gastric mucosa during Helicobacter pylori have a T helper cell 1 phenotype. Gastroenterology. 1998;114 :482-492. DOI: 10.1016/S0016-5085(98)70531-1 - 20.
Koch MRA, Gong R, Friedrich V, Engelsberger V, Kretschmer L, Wanisch A, et al. CagA-specific gastric CD8+ tissue-resident T cells control Helicobacter pylori during the early infection phase. Gastroenterology. 2023;164 :550-566. DOI: 10.1053/j.gastro.2022.12.016 - 21.
Zhang Z, Chen X, Li B, Xia T, Wu X, Wu C. Helicobacter pylori induces urease subunit B-specific CD8+ T cell responses in infected individuals via cytosolic pathway of cross-presentation. Helicobacter. 2023;28 :e13005. DOI: 10.1111/hel.13005 - 22.
Kronsteiner B, Bassaganya-Riera J, Philipson N, Hontecillas R. Novel insights on the role of CD8+ T cells and cytotoxic responses during Helicobacter pylori infection. Gut Microbes. 2014;5 :357-362. DOI: 10.4161/gmic.28899 - 23.
Kronsteiner B, Bassaganya-Riera J, Philipson C, Viladomiu M, Carbo A, Pedragosa M, et al. Helicobacter pylori infection in a pig model is dominated by Th1 and cytotoxic CD8+ T cell responses. Infection and Immunity. 2013;81 :3803-3813. DOI: 10.1128/IAI.00660-13 - 24.
Wu J, Zhu X, Guo X, Yang Z, Cai Q , Gu D, et al. Helicobacter urease suppresses cytotoxic CD8+ T-cell responses through activating Myh9-dependent induction of PD-L1. International Immunology. 2021; 33 :491-504. DOI: 10.1093/intimm/dxab044 - 25.
Azem J, Svennerholm A-M, Lundin BS. B cells pulsed with Helicobacter pylori antigen efficiently activate memory CD8+ T cells fromH. Pylori -infected individuals. Clinical Immunology. 2006;118 :284-291. DOI: 10.1016/j.clim.2005.09.011 - 26.
Wei L, Wang J, Liu Y. Prior to Foxp3 + regulatory T-cell induction, interleukin-10-producing B cells expand after Helicobacter pylori infection. Pathogens Disease. 2014;72 :45-54. DOI: 10.1111/2049-632X.12182 - 27.
Reyes VE, Peniche AG. Helicobacter Pylori Deregulates T and B Cell Signaling to Trigger Immune Evasion. In Molecular Mechanisms of Inflammation: Induction, Resolution and Escape byHelicobacter pylori . In: Backert S, editor. Current Topics in Microbiology and Immunology. Vol. 421. Cham: Springer International Publishing; 2019. pp. 229-265, ISBN 978-3-030-15137-9 - 28.
Hernández C, Toledo-Stuardo K, García-González P, Garrido-Tapia M, Kramm K, Rodríguez-Siza JA, et al. Heat-killed helicobacter pylori upregulates NKG2D ligands expression on gastric adenocarcinoma cells via toll-like receptor 4. Helicobacter. 2021;26 :e12812. DOI: 10.1111/hel.12812 - 29.
Kuo S-H. Expression of CD86 and increased infiltration of NK cells are associated with Helicobacter pylori -dependent state of early stage high-grade gastric MALT lymphoma. WJG. 2005;11 :4357. DOI: 10.3748/wjg.v11.i28.4357 - 30.
Kim D, Park J, Franchi L, Backert S, Núñez G. The cag pathogenicity island and interaction between TLR 2/NOD 2 and NLRP 3 regulate IL-1β production in Helicobacter pylori infected dendritic cells. European Journal of Immunology. 2013;43 :2650-2658. DOI: 10.1002/eji.201243281 - 31.
Oertli M, Sundquist M, Hitzler I, Engler DB, Arnold IC, Reuter S, et al. DC-derived IL-18 drives Treg differentiation, murine helicobacter pylori –specific immune tolerance, and asthma protection. The Journal of Clinical Investigation. 2012;122 :1082-1096. DOI: 10.1172/JCI61029 - 32.
Kao JY, Zhang M, Miller MJ, Mills JC, Wang B, Liu M, et al. Helicobacter pylori immune escape is mediated by dendritic cell–induced Treg skewing and Th17 suppression in mice. Gastroenterology. 2010;138 :1046-1054. DOI: 10.1053/j.gastro.2009.11.043 - 33.
Slomiany BL, Slomiany A. Modulation of gastric mucosal inflammatory responses to helicobacter pylori via ghrelin-induced protein kinase Cδ tyrosine phosphorylation. Inflammopharmacology. 2014;22 :251-262. DOI: 10.1007/s10787-014-0206-z - 34.
Munari F, Fassan M, Capitani N, Codolo G, Vila-Caballer M, Pizzi M, et al. Cytokine BAFF released by helicobacter pylori – Infected macrophages triggers the Th17 response in human chronic gastritis. The Journal of Immunology. 2014;193 :5584-5594. DOI: 10.4049/jimmunol.1302865 - 35.
Kareem RA, Kadhim Baqer L. Immunological role of toll like receptor markers (TLR2 and TLR4) In patients with helicobacter pylori infection At Basrah, Iraq. Archives of Razi Institute. 2022;78 (2):601-609. DOI: 10.22092/ari.2022.359715.2458 - 36.
Neuper T, Frauenlob T, Sarajlic M, Posselt G, Wessler S, Horejs-Hoeck J. TLR2, TLR4 and TLR10 shape the cytokine and chemokine release of H. pylori -infected human DCs. IJMS. 2020;21 :3897. DOI: 10.3390/ijms21113897 - 37.
Rizzuti D, Ang M, Sokollik C, Wu T, Abdullah M, Greenfield L, et al. Helicobacter pylori inhibits dendritic cell maturation via Interleukin-10-mediated activation of the signal transducer and activator of transcription 3 pathway. Journal of Innate Immunity. 2015;7 :199-211. DOI: 10.1159/000368232 - 38.
Kaebisch R, Mejías-Luque R, Prinz C, Gerhard M. Helicobacter pylori cytotoxin-associated Gene a impairs human dendritic cell maturation and function through IL-10–mediated activation of STAT3. The Journal of Immunology. 2014;192 :316-323. DOI: 10.4049/jimmunol.1302476 - 39.
Pickert G, Neufert C, Leppkes M, Zheng Y, Wittkopf N, Warntjen M, et al. STAT3 links IL-22 signaling in intestinal epithelial cells to mucosal wound healing. Journal of Experimental Medicine. 2009; 206 :1465-1472. DOI: 10.1084/jem.20082683 - 40.
Lina TT, Gonzalez J, Pinchuk IV, Beswick EJ, Reyes VE. Helicobacter pylori elicits B7-H3 expression on gastric epithelial cells: Implications in local T cell regulation and subset development during infection. COR. 2019;2 :1-12. DOI: 10.31487/j.COR.2019.05.05 - 41.
Sarajlic M, Neuper T, Vetter J, Schaller S, Klicznik MM, Gratz IK, et al. H. pylori modulates DC functions via T4SS/TNFα/P38-dependent SOCS3 expression. Cell Communication and Signaling: CCS. 2020;18 :160. DOI: 10.1186/s12964-020-00655-1 - 42.
Mostaghimi T, Bahadoran E, Bakht M, Taheri S, Sadeghi H, Babaei A. Role of lncRNAs in helicobacter pylori and Epstein-Barr virus associated gastric cancers. Life Sciences. 2024;336 :122316. DOI: 10.1016/j.lfs.2023.122316 - 43.
Lv Y, Teng Y, Mao F, Peng L, Zhang J, Cheng P, et al. Helicobacter pylori -induced IL-33 modulates mast cell responses, benefits bacterial growth, and contributes to gastritis. Cell Death & Disease. 2018;9 :457. DOI: 10.1038/s41419-018-0493-1 - 44.
Tran LS, Tran D, De Paoli A, D’Costa K, Creed SJ, Ng GZ, et al. NOD1 is required for helicobacter pylori induction of IL-33 responses in gastric epithelial cells. Cellular Microbiology. 2018;20 :e12826. DOI: 10.1111/cmi.12826 - 45.
Arnold IC, Zhang X, Urban S, Artola-Borán M, Manz MG, Ottemann KM, et al. NLRP3 controls the development of gastrointestinal CD11b + dendritic cells in the steady state and during chronic bacterial infection. Cell Reports. 2017; 21 :3860-3872. DOI: 10.1016/j.celrep.2017.12.015 - 46.
Zhang X, Arnold IC, Müller A. Mechanisms of persistence, innate immune activation and immunomodulation by the gastric pathogen helicobacter pylori . Current Opinion in Microbiology. 2020;54 :1-10. DOI: 10.1016/j.mib.2020.01.003 - 47.
Lv Y, Cheng P, Zhang J, Mao F, Teng Y, Liu Y, et al. Helicobacter pylori – induced matrix metallopeptidase-10 promotes gastric bacterial colonization and gastritis. Science Advances. 2019;5 :eaau6547. DOI: 10.1126/sciadv.aau6547 - 48.
Liu M, Hu Z, Wang C, Zhang Y. The TLR/MyD88 signalling cascade in inflammation and gastric cancer: The immune regulatory network of helicobacter pylori . Journal of Molecular Medicine. 2023;101 :767-781. DOI: 10.1007/s00109-023-02332-5 - 49.
Sathe A, Grimes SM, Lau BT, Chen J, Suarez C, Huang RJ, et al. Single-cell genomic characterization reveals the cellular reprogramming of the gastric tumor microenvironment. Clinical Cancer Research. 2020; 26 :2640-2653. DOI: 10.1158/1078-0432.CCR-19-3231 - 50.
Tang F, Du X, Liu M, Zheng P, Liu Y. Anti-CTLA-4 antibodies in cancer immunotherapy: Selective depletion of Intratumoral regulatory T cells or checkpoint blockade? Cell & Bioscience. 2018; 8 :30. DOI: 10.1186/s13578-018-0229-z - 51.
Shen K, Chen B, Yang L, Gao W. KYNU as a biomarker of tumor-associated macrophages and correlates with immunosuppressive microenvironment and poor prognosis in gastric cancer. International Journal of Genomics. 2023; 2023 :4662480. DOI: 10.1155/2023/4662480 - 52.
Shime H, Matsumoto M, Oshiumi H, Tanaka S, Nakane A, Iwakura Y, et al. Toll-like receptor 3 signaling converts tumor-supporting myeloid cells to tumoricidal effectors. Proceedings of the National Academy of Sciences of the United States of America. 2012; 109 :2066-2071. DOI: 10.1073/pnas.1113099109 - 53.
Zhang G, Gao Z, Guo X, Ma R, Wang X, Zhou P, et al. CAP2 promotes gastric cancer metastasis by mediating the interaction between tumor cells and tumor-associated macrophages. The Journal of Clinical Investigation. 2023; 133 :e166224. DOI: 10.1172/JCI166224 - 54.
Yu B, Xiang L, Peppelenbosch MP, Fuhler GM. Overlapping cytokines in H. Pylori infection and gastric cancer: A tandem meta-analysis. Frontiers in Immunology. 2023;14 :1125658. DOI: 10.3389/fimmu.2023.1125658 - 55.
Kuhn KA, Manieri NA, Liu T-C, Stappenbeck TS. IL-6 stimulates intestinal epithelial proliferation and repair after injury. PLoS One. 2014; 9 :e114195. DOI: 10.1371/journal.pone.0114195 - 56.
Garay J, Piazuelo MB, Majumdar S, Li L, Trillo-Tinoco J, Del Valle L, et al. The homing receptor CD44 is involved in the progression of precancerous gastric lesions in patients infected with helicobacter pylori and in development of mucous metaplasia in mice. Cancer Letters. 2016;371 :90-98. DOI: 10.1016/j.canlet.2015.10.037 - 57.
Yu H, Lee H, Herrmann A, Buettner R, Jove R. Revisiting STAT3 Signalling in cancer: New and unexpected biological functions. Nature Reviews. Cancer. 2014; 14 :736-746. DOI: 10.1038/nrc3818 - 58.
Piao J, Lee HG, Kim S, Kim D, Han H, Ngo H, et al. Helicobacter pylori activates IL-6-STAT3 signaling in human gastric cancer cells: Potential roles for reactive oxygen species. Helicobacter. 2016;21 :405-416. DOI: 10.1111/hel.12298 - 59.
Kang JH, Park S, Rho J, Hong E-J, Cho Y-E, Won Y-S, et al. IL-17A promotes helicobacter pylori -induced gastric carcinogenesis via interactions with IL-17RC. Gastric Cancer. 2023;26 :82-94. DOI: 10.1007/s10120-022-01342-5 - 60.
Li X, Pan K, Vieth M, Gerhard M, Li W, Mejías-Luque R. JAK-STAT1 signaling pathway is an early response to helicobacter pylori infection and contributes to immune escape and gastric carcinogenesis. IJMS. 2022;23 :4147. DOI: 10.3390/ijms23084147 - 61.
Katoh M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nature Reviews. Clinical Oncology. 2019; 16 :105-122. DOI: 10.1038/s41571-018-0115-y - 62.
Zhang X, Soutto M, Chen Z, Bhat N, Zhu S, Eissmann MF, et al. Induction of fibroblast growth factor receptor 4 by helicobacter pylori via signal transducer and activator of transcription 3 with a feedforward activation loop involving SRC signaling in gastric cancer. Gastroenterology. 2022;163 :620-636.e9. DOI: 10.1053/j.gastro.2022.05.016 - 63.
Xu J, Yu Y, He X, Niu N, Li X, Zhang R, et al. Tumor-associated macrophages induce invasion and poor prognosis in human gastric cancer in a cyclooxygenase-2/MMP9-dependent manner. American Journal of Translational Research. 2019; 11 :6040-6054 - 64.
Zhang J, Fu L, Yasuda-Yoshihara N, Yonemura A, Wei F, Bu L, et al. IL-1β derived from mixed-polarized macrophages activates fibroblasts and synergistically forms a cancer-promoting microenvironment. Gastric Cancer. 2023; 26 :187-202. DOI: 10.1007/s10120-022-01352-3 - 65.
Piao H, Fu L, Wang Y, Liu Y, Wang Y, Meng X, et al. A positive feedback loop between gastric cancer cells and tumor-associated macrophage induces malignancy progression. Journal of Experimental & Clinical Cancer Research. 2022; 41 :174. DOI: 10.1186/s13046-022-02366-6 - 66.
Lam SY, Mommersteeg MC, Yu B, Broer L, Spaander MCW, Frost F, et al. Toll-like receptor 1 locus Re-examined in a genome-wide association study update on anti– helicobacter pylori IgG titers. Gastroenterology. 2022;162 :1705-1715. DOI: 10.1053/j.gastro.2022.01.011