We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Akkermansia muciniphila improves gastric cancer treatment by modulating the immune microenvironment

    Jianming Fang

    Department of Hepatobiliary Pancreatic Surgery, GuangFu Oncology Hospital, Jinhua, 321000, China

    ,
    Huizhong Zhang

    Department of Hepatobiliary Pancreatic Surgery, GuangFu Oncology Hospital, Jinhua, 321000, China

    ,
    Xiaodong Zhang

    Department of Hepatobiliary Pancreatic Surgery, GuangFu Oncology Hospital, Jinhua, 321000, China

    ,
    Xiaolong Lu

    Department of Hepatobiliary Pancreatic Surgery, GuangFu Oncology Hospital, Jinhua, 321000, China

    ,
    Junjie Liu

    Department of Hepatobiliary Pancreatic Surgery, GuangFu Oncology Hospital, Jinhua, 321000, China

    ,
    Haiyang Li

    Department of Hepatobiliary Pancreatic Surgery, GuangFu Oncology Hospital, Jinhua, 321000, China

    &
    Jianxin Huang

    *Author for correspondence: Tel.: +86 159 0579 0573;

    E-mail Address: xinxin_hjx@163.com

    Department of Hepatobiliary Pancreatic Surgery, GuangFu Oncology Hospital, Jinhua, 321000, China

    Published Online:https://doi.org/10.2217/fmb-2023-0210

    Background: Gut microbiota is pivotal in tumor occurrence and development, and there is a close relationship between Akkermansia muciniphila (AKK) and cancer immunotherapy. Methods: The effects of AKK and its outer membrane proteins on gastric cancer (GC) were evaluated in vitro and in vivo using cell counting kit-8 assay, flow cytometry, western blotting, ELISA, immunohistochemistry and immunofluorescence. Results: AKK outer membrane protein facilitated apoptosis of GC cells and exerted an immunostimulatory effect (by promoting M1 polarization of macrophages, enhancing expression of cytotoxic T-lymphocyte-related cytokines and suppressing that of Treg-related cytokines). Additionally, AKK and its formulation could inhibit tumor growth of GC and enhance the infiltration of immune cells in tumor tissues. Conclusion: AKK could improve GC treatment by modulating the immune microenvironment.

    Plain language summary

    Akkermansia muciniphila (AKK) is a type of bacteria found in the human gut that is good for the immune system. We wanted to investigate the effect of AKK on cancer. We extracted a protein from AKK called Amuc. AKK and Amuc inhibited the growth of stomach cancer by encouraging the action of immune cells. AKK may therefore be able to treat stomach cancer.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Thrift AP, El-Serag HB. Burden of gastric cancer. Clin. Gastroenterol Hepatol. 18(3), 534–542 (2020).
    • 2. Sung H, Ferlay J, Siegel RL et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71(3), 209–249 (2021).
    • 3. Li K, Zhang A, Li X, Zhang H, Zhao L. Advances in clinical immunotherapy for gastric cancer. Biochim. Biophys. Acta Rev. Cancer 1876(2), 188615 (2021).
    • 4. Smyth E, Knodler M, Giraut A et al. VESTIGE: adjuvant immunotherapy in patients with resected esophageal, gastroesophageal junction and gastric cancer following preoperative chemotherapy with high risk for recurrence (N+ and/or R1): an open label randomized controlled phase-2-study. Front. Oncol. 9, 1320 (2019).
    • 5. Bang YJ, Van Cutsem E, Fuchs CS et al. KEYNOTE-585: phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Future Oncol. 15(9), 943–952 (2019).
    • 6. Janjigian YY, Shitara K, Moehler M 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 398(10294), 27–40 (2021).
    • 7. Sun JM, Shen L, Shah MA et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): a randomised, placebo-controlled, phase 3 study. Lancet 398(10302), 759–771 (2021).
    • 8. Wang F, Wei XL, Wang FH et al. Safety, efficacy and tumor mutational burden as a biomarker of overall survival benefit in chemo-refractory gastric cancer treated with toripalimab, a PD-1 antibody in phase Ib/II clinical trial NCT02915432. Ann. Oncol. 30(9), 1479–1486 (2019).
    • 9. Jin X, Liu Z, Yang D, Yin K, Chang X. Recent progress and future perspectives of immunotherapy in advanced gastric cancer. Front. Immunol. 13, 948647 (2022).
    • 10. Meng C, Bai C, Brown TD, Hood LE, Tian Q. Human gut microbiota and gastrointestinal cancer. Genomics Proteomics Bioinformatics 16(1), 33–49 (2018).
    • 11. Correa P, Piazuelo MB. Helicobacter pylori infection and gastric adenocarcinoma. US Gastroenterol. Hepatol. Rev. 7(1), 59–64 (2011).
    • 12. Routy B, Le Chatelier E, Derosa L et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359(6371), 91–97 (2018). • In vivo experiments showed that the PD-1-based immunotherapy Akkermansia muciniphila (AKK) strain improved antitumor activity.
    • 13. Zitvogel L, Ma Y, Raoult D, Kroemer G, Gajewski TF. The microbiome in cancer immunotherapy: diagnostic tools and therapeutic strategies. Science 359(6382), 1366–1370 (2018).
    • 14. Sivan A, Corrales L, Hubert N et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350(6264), 1084–1089 (2015).
    • 15. Das S, Suarez G, Beswick EJ, Sierra JC, Graham DY, Reyes VE. Expression of B7-H1 on gastric epithelial cells: its potential role in regulating T cells during Helicobacter pylori infection. J. Immunol. 176(5), 3000–3009 (2006).
    • 16. Reunanen J, Kainulainen V, Huuskonen L et al. Akkermansia muciniphila adheres to enterocytes and strengthens the integrity of the epithelial cell layer. Appl. Environ. Microbiol. 81(11), 3655–3662 (2015).
    • 17. Dao MC, Everard A, Aron-Wisnewsky J et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut 65(3), 426–436 (2016).
    • 18. Shin NR, Lee JC, Lee HY et al. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 63(5), 727–735 (2014).
    • 19. Barcena C, Valdes-Mas R, Mayoral P et al. Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat. Med. 25(8), 1234–1242 (2019).
    • 20. Bian X, Wu W, Yang L et al. Administration of Akkermansia muciniphila ameliorates dextran sulfate sodium-induced ulcerative colitis in mice. Front. Microbiol. 10, 2259 (2019).
    • 21. Zhai R, Xue X, Zhang L, Yang X, Zhao L, Zhang C. Strain-specific anti-inflammatory properties of two Akkermansia muciniphila strains on chronic colitis in mice. Front. Cell. Infect. Microbiol. 9, 239 (2019).
    • 22. Ansaldo E, Slayden LC, Ching KL et al. Akkermansia muciniphila induces intestinal adaptive immune responses during homeostasis. Science 364(6446), 1179–1184 (2019). •• Showed that Akkermansia muciniphila is an intestinal bacterium associated with host metabolism and systemic effects of PD-1 checkpoint immunotherapy.
    • 23. Zhang M, Liu K, Zhang Q et al. Alpha fetoprotein promotes polarization of macrophages towards M2-like phenotype and inhibits macrophages to phagocytize hepatoma cells. Front. Immunol. 14, 1081572 (2023).
    • 24. Wang L, Tang L, Feng Y et al. A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8(+) T cells in mice. Gut 69(11), 1988–1997 (2020). • AKK or Amuc_1100 ameliorates colitis and reduces infiltrating macrophages and CD8 cytotoxic T lymphocytes in the colon.
    • 25. Luo ZW, Xia K, Liu YW et al. Extracellular vesicles from Akkermansia muciniphila elicit antitumor immunity against prostate cancer via modulation of CD8(+) T cells and macrophages. Int. J. Nanomedicine 16, 2949–2963 (2021).
    • 26. Zhou Y, Xia L, Liu Q et al. Induction of pro-inflammatory response via activated macrophage-mediated NF-κB and STAT3 pathways in gastric cancer cells. Cell. Physiol. Biochem. 47(4), 1399–1410 (2018).
    • 27. Jiang Y, Zhang M, Guo T, Yang C, Zhang C, Hao J. MicroRNA-21-5p promotes proliferation of gastric cancer cells through targeting SMAD7. Onco Targets Ther. 11, 4901–4911 (2018).
    • 28. Li Y, Zhang D, Yu K et al. CMPD1 inhibited human gastric cancer cell proliferation by inducing apoptosis and G2/M cell cycle arrest. Biol. Res. 51(1), 11 (2018).
    • 29. Yamaguchi T, Fushida S, Yamamoto Y et al. Tumor-associated macrophages of the M2 phenotype contribute to progression in gastric cancer with peritoneal dissemination. Gastric Cancer 19(4), 1052–1065 (2016).
    • 30. Zhao Q, Cao L, Guan L et al. Immunotherapy for gastric cancer: dilemmas and prospect. Brief. Funct. Genomics 18(2), 107–112 (2019).
    • 31. Hogner A, Moehler M. Immunotherapy in gastric cancer. Curr. Oncol. 29(3), 1559–1574 (2022).
    • 32. Xu R, Wu J, Zhang X et al. Modified Bu-zhong-yi-qi decoction synergies with 5 fluorouracile to inhibits gastric cancer progress via PD-1/PD-L1-dependent T cell immunization. Pharmacol. Res. 152, 104623 (2020).
    • 33. Zhang T, Li Q, Cheng L, Buch H, Zhang F. Akkermansia muciniphila is a promising probiotic. Microb. Biotechnol. 12(6), 1109–1125 (2019).
    • 34. Cani PD, Depommier C, Derrien M, Everard A, de Vos WM. Akkermansia muciniphila: paradigm for next-generation beneficial microorganisms. Nat. Rev. Gastroenterol. Hepatol. 19(10), 625–637 (2022).
    • 35. Gopalakrishnan V, Spencer CN, Nezi L et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359(6371), 97–103 (2018).
    • 36. Li T, Lin X, Shen B et al. Akkermansia muciniphila suppressing nonalcoholic steatohepatitis associated tumorigenesis through CXCR6(+) natural killer T cells. Front. Immunol. 13, 1047570 (2022).
    • 37. Jin Y, Dong H, Xia L et al. The diversity of gut microbiome is associated with favorable responses to anti-programmed death 1 immunotherapy in chinese patients with NSCLC. J. Thorac. Oncol. 14(8), 1378–1389 (2019). • AKK abundance significantly correlates with immune checkpoint inhibitors in non-small-cell lung cancer patients with better immune responses.
    • 38. Shi L, Sheng J, Chen G et al. Combining IL-2-based immunotherapy with commensal probiotics produces enhanced antitumor immune response and tumor clearance. J. Immunother. Cancer 8(2), e000973 (2020).
    • 39. Zhuo Q, Yu B, Zhou J et al. Lysates of Lactobacillus acidophilus combined with CTLA-4-blocking antibodies enhance antitumor immunity in a mouse colon cancer model. Sci. Rep. 9(1), 20128 (2019).
    • 40. Riehl TE, Alvarado D, Ee X et al. Lactobacillus rhamnosus GG protects the intestinal epithelium from radiation injury through release of lipoteichoic acid, macrophage activation and the migration of mesenchymal stem cells. Gut 68(6), 1003–1013 (2019).
    • 41. Ngambenjawong C, Gustafson HH, Pun SH. Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv. Drug Deliv. Rev. 114, 206–221 (2017).
    • 42. Sica A, Larghi P, Mancino A et al. Macrophage polarization in tumour progression. Semin. Cancer Biol. 18(5), 349–355 (2008).
    • 43. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat. Immunol. 11(10), 889–896 (2010).
    • 44. Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 877, 173090 (2020).
    • 45. Komohara Y, Fujiwara Y, Ohnishi K, Takeya M. Tumor-associated macrophages: potential therapeutic targets for anti-cancer therapy. Adv. Drug Deliv. Rev. 99(Pt B), 180–185 (2016).
    • 46. Fan L, Xu C, Ge Q et al. A. muciniphila suppresses colorectal tumorigenesis by inducing TLR2/NLRP3-mediated M1-like TAMs. Cancer Immunol. Res. 9(10), 1111–1124 (2021).
    • 47. Jiang Y, Xu Y, Zheng C et al. Acetyltransferase from Akkermansia muciniphila blunts colorectal tumourigenesis by reprogramming tumour microenvironment. Gut 72(7), 1308–1318 (2023).
    • 48. Plovier H, Everard A, Druart C et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat. Med. 23(1), 107–113 (2017).
    • 49. Depommier C, Everard A, Druart C et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat. Med. 25(7), 1096–1103 (2019).
    • 50. Zhu Z, Cai J, Hou W et al. Microbiome and spatially resolved metabolomics analysis reveal the anticancer role of gut Akkermansia muciniphila by crosstalk with intratumoral microbiota and reprogramming tumoral metabolism in mice. Gut Microbes 15(1), 2166700 (2023).