Skip to main content
Top
Published in: Journal of Translational Medicine 1/2022

Open Access 01-12-2022 | Respiratory Microbiota | Review

Emerging role of human microbiome in cancer development and response to therapy: special focus on intestinal microflora

Authors: Hourieh Sadrekarimi, Zhanna R. Gardanova, Morteza Bakhshesh, Farnoosh Ebrahimzadeh, Amirhossein Fakhre Yaseri, Lakshmi Thangavelu, Zahra Hasanpoor, Firoozeh Abolhasani Zadeh, Mohammad Saeed Kahrizi

Published in: Journal of Translational Medicine | Issue 1/2022

Login to get access

Abstract

In recent years, there has been a greater emphasis on the impact of microbial populations inhabiting the gastrointestinal tract on human health and disease. According to the involvement of microbiota in modulating physiological processes (such as immune system development, vitamins synthesis, pathogen displacement, and nutrient uptake), any alteration in its composition and diversity (i.e., dysbiosis) has been linked to a variety of pathologies, including cancer. In this bidirectional relationship, colonization with various bacterial species is correlated with a reduced or elevated risk of certain cancers. Notably, the gut microflora could potentially play a direct or indirect role in tumor initiation and progression by inducing chronic inflammation and producing toxins and metabolites. Therefore, identifying the bacterial species involved and their mechanism of action could be beneficial in preventing the onset of tumors or controlling their advancement. Likewise, the microbial community affects anti-cancer approaches’ therapeutic potential and adverse effects (such as immunotherapy and chemotherapy). Hence, their efficiency should be evaluated in the context of the microbiome, underlining the importance of personalized medicine. In this review, we summarized the evidence revealing the microbiota's involvement in cancer and its mechanism. We also delineated how microbiota could predict colon carcinoma development or response to current treatments to improve clinical outcomes.
Literature
2.
go back to reference Yang Q, Liang Q, Balakrishnan B, Belobrajdic DP, Feng QJ, Zhang W. Role of dietary nutrients in the modulation of gut microbiota: a narrative review. Nutrients. 2020;12(2):381.PubMedCentralCrossRef Yang Q, Liang Q, Balakrishnan B, Belobrajdic DP, Feng QJ, Zhang W. Role of dietary nutrients in the modulation of gut microbiota: a narrative review. Nutrients. 2020;12(2):381.PubMedCentralCrossRef
3.
go back to reference Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, Knight R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA. 2010;107(26):11971–5.PubMedPubMedCentralCrossRef Dominguez-Bello MG, Costello EK, Contreras M, Magris M, Hidalgo G, Fierer N, Knight R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA. 2010;107(26):11971–5.PubMedPubMedCentralCrossRef
4.
go back to reference Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–7.PubMedPubMedCentralCrossRef Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–7.PubMedPubMedCentralCrossRef
5.
go back to reference Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT, et al. Human genetics shape the gut microbiome. Cell. 2014;159(4):789–99.PubMedPubMedCentralCrossRef Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, Beaumont M, Van Treuren W, Knight R, Bell JT, et al. Human genetics shape the gut microbiome. Cell. 2014;159(4):789–99.PubMedPubMedCentralCrossRef
6.
go back to reference Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7(3):189–200.PubMedPubMedCentralCrossRef Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7(3):189–200.PubMedPubMedCentralCrossRef
8.
go back to reference Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar RD. Role of the normal gut microbiota. World J Gastroenterol. 2015;21(29):8787–803.PubMedPubMedCentralCrossRef Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar RD. Role of the normal gut microbiota. World J Gastroenterol. 2015;21(29):8787–803.PubMedPubMedCentralCrossRef
9.
go back to reference Peterson J, Garges S, Giovanni M, McInnes P, Wang L, Schloss JA, Bonazzi V, McEwen JE, Wetterstrand KA, Deal C, et al. The NIH human microbiome project. Genome Res. 2009;19(12):2317–23.PubMedPubMedCentralCrossRef Peterson J, Garges S, Giovanni M, McInnes P, Wang L, Schloss JA, Bonazzi V, McEwen JE, Wetterstrand KA, Deal C, et al. The NIH human microbiome project. Genome Res. 2009;19(12):2317–23.PubMedPubMedCentralCrossRef
10.
go back to reference Dy P, Wang W, Bhattaram P, Wang Q, Wang L, Ballock RT, Lefebvre V. Sox9 directs hypertrophic maturation and blocks osteoblast differentiation of growth plate chondrocytes. Dev Cell. 2012;22(3):597–609.PubMedPubMedCentralCrossRef Dy P, Wang W, Bhattaram P, Wang Q, Wang L, Ballock RT, Lefebvre V. Sox9 directs hypertrophic maturation and blocks osteoblast differentiation of growth plate chondrocytes. Dev Cell. 2012;22(3):597–609.PubMedPubMedCentralCrossRef
11.
go back to reference Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65.PubMedPubMedCentralCrossRef Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65.PubMedPubMedCentralCrossRef
13.
go back to reference Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17(4):219–32.PubMedCrossRef Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17(4):219–32.PubMedCrossRef
14.
go back to reference Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018;11(1):1–10.PubMedCrossRef Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018;11(1):1–10.PubMedCrossRef
15.
go back to reference Bostanciklioğlu M. The role of gut microbiota in pathogenesis of Alzheimer’s disease. J Appl Microbiol. 2019;127(4):954–67.PubMedCrossRef Bostanciklioğlu M. The role of gut microbiota in pathogenesis of Alzheimer’s disease. J Appl Microbiol. 2019;127(4):954–67.PubMedCrossRef
16.
go back to reference Luca M, Di Mauro M, Di Mauro M, Luca A. Gut microbiota in alzheimer’s disease, depression, and type 2 diabetes mellitus: the role of oxidative stress. Oxid Med Cell Longev. 2019;2019:4730539.PubMedPubMedCentral Luca M, Di Mauro M, Di Mauro M, Luca A. Gut microbiota in alzheimer’s disease, depression, and type 2 diabetes mellitus: the role of oxidative stress. Oxid Med Cell Longev. 2019;2019:4730539.PubMedPubMedCentral
17.
go back to reference Lin C, Cai X, Zhang J, Wang W, Sheng Q, Hua H, Zhou X. Role of gut microbiota in the development and treatment of colorectal cancer. Digestion. 2019;100(1):72–8.PubMedCrossRef Lin C, Cai X, Zhang J, Wang W, Sheng Q, Hua H, Zhou X. Role of gut microbiota in the development and treatment of colorectal cancer. Digestion. 2019;100(1):72–8.PubMedCrossRef
18.
go back to reference Pitocco D, Di Leo M, Tartaglione L, De Leva F, Petruzziello C, Saviano A, Pontecorvi A, Ojetti V. The role of gut microbiota in mediating obesity and diabetes mellitus. Eur Rev Med Pharmacol Sci. 2020;24(3):1548–62.PubMed Pitocco D, Di Leo M, Tartaglione L, De Leva F, Petruzziello C, Saviano A, Pontecorvi A, Ojetti V. The role of gut microbiota in mediating obesity and diabetes mellitus. Eur Rev Med Pharmacol Sci. 2020;24(3):1548–62.PubMed
20.
go back to reference Jiao Y, Wu L, Huntington ND, Zhang X. Crosstalk between gut microbiota and innate immunity and its implication in autoimmune diseases. Front Immunol. 2020;11:282.PubMedPubMedCentralCrossRef Jiao Y, Wu L, Huntington ND, Zhang X. Crosstalk between gut microbiota and innate immunity and its implication in autoimmune diseases. Front Immunol. 2020;11:282.PubMedPubMedCentralCrossRef
21.
go back to reference De Luca F, Shoenfeld Y. The microbiome in autoimmune diseases. Clin Exp Immunol. 2019;195(1):74–85.PubMedCrossRef De Luca F, Shoenfeld Y. The microbiome in autoimmune diseases. Clin Exp Immunol. 2019;195(1):74–85.PubMedCrossRef
22.
go back to reference Hassanpour SH, Dehghani M. Review of cancer from perspective of molecular. J Cancer Res Pract. 2017;4(4):127–9.CrossRef Hassanpour SH, Dehghani M. Review of cancer from perspective of molecular. J Cancer Res Pract. 2017;4(4):127–9.CrossRef
23.
go back to reference García-Castillo V, Sanhueza E, McNerney E, Onate SA, García A. Microbiota dysbiosis: a new piece in the understanding of the carcinogenesis puzzle. J Med Microbiol. 2016;65(12):1347–62.PubMedCrossRef García-Castillo V, Sanhueza E, McNerney E, Onate SA, García A. Microbiota dysbiosis: a new piece in the understanding of the carcinogenesis puzzle. J Med Microbiol. 2016;65(12):1347–62.PubMedCrossRef
24.
go back to reference Zhang X, Li C, Cao W, Zhang Z. Alterations of gastric microbiota in gastric cancer and precancerous stages. Front Cell Infect Microbiol. 2021;11: 559148.PubMedPubMedCentralCrossRef Zhang X, Li C, Cao W, Zhang Z. Alterations of gastric microbiota in gastric cancer and precancerous stages. Front Cell Infect Microbiol. 2021;11: 559148.PubMedPubMedCentralCrossRef
25.
go back to reference Abreu MT, Peek RM Jr. Gastrointestinal malignancy and the microbiome. Gastroenterology. 2014;146(6):1534-46.e3.PubMedCrossRef Abreu MT, Peek RM Jr. Gastrointestinal malignancy and the microbiome. Gastroenterology. 2014;146(6):1534-46.e3.PubMedCrossRef
26.
go back to reference Sun D, Chen Y, Fang JY. Influence of the microbiota on epigenetics in colorectal cancer. Natl Sci Rev. 2019;6(6):1138–48.PubMedCrossRef Sun D, Chen Y, Fang JY. Influence of the microbiota on epigenetics in colorectal cancer. Natl Sci Rev. 2019;6(6):1138–48.PubMedCrossRef
27.
go back to reference Rezasoltani S, Asadzadeh-Aghdaei H, Nazemalhosseini-Mojarad E, Dabiri H, Ghanbari R, Zali MR. Gut microbiota, epigenetic modification and colorectal cancer. Iran J Microbiol. 2017;9(2):55–63.PubMedPubMedCentral Rezasoltani S, Asadzadeh-Aghdaei H, Nazemalhosseini-Mojarad E, Dabiri H, Ghanbari R, Zali MR. Gut microbiota, epigenetic modification and colorectal cancer. Iran J Microbiol. 2017;9(2):55–63.PubMedPubMedCentral
29.
go back to reference Shen J, Xiao Z, Wu WK, Wang MH, To KF, Chen Y, Yang W, Li MS, Shin VY, Tong JH, et al. Epigenetic silencing of miR-490-3p reactivates the chromatin remodeler SMARCD1 to promote Helicobacter pylori-induced gastric carcinogenesis. Cancer Res. 2015;75(4):754–65.PubMedCrossRef Shen J, Xiao Z, Wu WK, Wang MH, To KF, Chen Y, Yang W, Li MS, Shin VY, Tong JH, et al. Epigenetic silencing of miR-490-3p reactivates the chromatin remodeler SMARCD1 to promote Helicobacter pylori-induced gastric carcinogenesis. Cancer Res. 2015;75(4):754–65.PubMedCrossRef
30.
go back to reference Matson V, Chervin CS, Gajewski TF. Cancer and the microbiome-influence of the commensal microbiota on cancer, immune responses, and immunotherapy. Gastroenterology. 2021;160(2):600–13.PubMedCrossRef Matson V, Chervin CS, Gajewski TF. Cancer and the microbiome-influence of the commensal microbiota on cancer, immune responses, and immunotherapy. Gastroenterology. 2021;160(2):600–13.PubMedCrossRef
32.
go back to reference Huët MAL, Lee CZ, Rahman S. A review on association of fungi with the development and progression of carcinogenesis in the human body. Curr Res Microb Sci. 2022;3: 100090.PubMed Huët MAL, Lee CZ, Rahman S. A review on association of fungi with the development and progression of carcinogenesis in the human body. Curr Res Microb Sci. 2022;3: 100090.PubMed
33.
go back to reference Luan C, Xie L, Yang X, Miao H, Lv N, Zhang R, Xiao X, Hu Y, Liu Y, Wu N, et al. Dysbiosis of fungal microbiota in the intestinal mucosa of patients with colorectal adenomas. Sci Rep. 2015;5:7980.PubMedPubMedCentralCrossRef Luan C, Xie L, Yang X, Miao H, Lv N, Zhang R, Xiao X, Hu Y, Liu Y, Wu N, et al. Dysbiosis of fungal microbiota in the intestinal mucosa of patients with colorectal adenomas. Sci Rep. 2015;5:7980.PubMedPubMedCentralCrossRef
34.
go back to reference Coker OO, Nakatsu G, Dai RZ, Wu WKK, Wong SH, Ng SC, Chan FKL, Sung JJY, Yu J. Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 2019;68(4):654–62.PubMedCrossRef Coker OO, Nakatsu G, Dai RZ, Wu WKK, Wong SH, Ng SC, Chan FKL, Sung JJY, Yu J. Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 2019;68(4):654–62.PubMedCrossRef
35.
go back to reference Gao R, Kong C, Li H, Huang L, Qu X, Qin N, Qin H. Dysbiosis signature of mycobiota in colon polyp and colorectal cancer. Eur J Clin Microbiol Infect Dis. 2017;36(12):2457–68.PubMedCrossRef Gao R, Kong C, Li H, Huang L, Qu X, Qin N, Qin H. Dysbiosis signature of mycobiota in colon polyp and colorectal cancer. Eur J Clin Microbiol Infect Dis. 2017;36(12):2457–68.PubMedCrossRef
36.
go back to reference Vannucci L, Stepankova R, Kozakova H, Fiserova A, Rossmann P, Tlaskalova-Hogenova H. Colorectal carcinogenesis in germ-free and conventionally reared rats: different intestinal environments affect the systemic immunity. Int J Oncol. 2008;32(3):609–17.PubMed Vannucci L, Stepankova R, Kozakova H, Fiserova A, Rossmann P, Tlaskalova-Hogenova H. Colorectal carcinogenesis in germ-free and conventionally reared rats: different intestinal environments affect the systemic immunity. Int J Oncol. 2008;32(3):609–17.PubMed
37.
go back to reference Mishra R, Rajsiglová L, Lukáč P, Tenti P, Šima P, Čaja F, Vannucci L. Spontaneous and induced tumors in germ-free animals: a general review. Medicina (Kaunas). 2021;57(3):260.PubMedPubMedCentralCrossRef Mishra R, Rajsiglová L, Lukáč P, Tenti P, Šima P, Čaja F, Vannucci L. Spontaneous and induced tumors in germ-free animals: a general review. Medicina (Kaunas). 2021;57(3):260.PubMedPubMedCentralCrossRef
38.
go back to reference Mizutani T, Mitsuoka T. Effect of intestinal bacteria on incidence of liver tumors in gnotobiotic C3H/He male mice. J Natl Cancer Inst. 1979;63(6):1365–70.PubMed Mizutani T, Mitsuoka T. Effect of intestinal bacteria on incidence of liver tumors in gnotobiotic C3H/He male mice. J Natl Cancer Inst. 1979;63(6):1365–70.PubMed
39.
go back to reference Mizutani T, Yamamoto T, Ozaki A, Oowada T, Mitsuoka T. Spontaneous polyposis in the small intestine of germ-free and conventionalized BALB/c mice. Cancer Lett. 1984;25(1):19–23.PubMedCrossRef Mizutani T, Yamamoto T, Ozaki A, Oowada T, Mitsuoka T. Spontaneous polyposis in the small intestine of germ-free and conventionalized BALB/c mice. Cancer Lett. 1984;25(1):19–23.PubMedCrossRef
40.
go back to reference Pilgrim HI, Labrecque AD. The incidence of mammary tumors in germ-free C3H mice. Cancer Res. 1967;27(3):584–6.PubMed Pilgrim HI, Labrecque AD. The incidence of mammary tumors in germ-free C3H mice. Cancer Res. 1967;27(3):584–6.PubMed
41.
go back to reference Reddy BS, Narisawa T, Wright P, Vukusich D, Weisburger JH, Wynder EL. Colon carcinogenesis with azoxymethane and dimethylhydrazine in germ-free rats. Cancer Res. 1975;35(2):287–90.PubMed Reddy BS, Narisawa T, Wright P, Vukusich D, Weisburger JH, Wynder EL. Colon carcinogenesis with azoxymethane and dimethylhydrazine in germ-free rats. Cancer Res. 1975;35(2):287–90.PubMed
42.
go back to reference Pollard M, Salomon JC. Oncogenic effect of methyl-cholanthrene in new-born germfree mice. Proc Soc Exp Biol Med. 1963;112:256–9.PubMedCrossRef Pollard M, Salomon JC. Oncogenic effect of methyl-cholanthrene in new-born germfree mice. Proc Soc Exp Biol Med. 1963;112:256–9.PubMedCrossRef
43.
go back to reference Burstein NA, McIntire KR, Allison AC. Pulmonary tumors in germfree mice: induction with urethan. J Natl Cancer Inst. 1970;44(1):211–4.PubMed Burstein NA, McIntire KR, Allison AC. Pulmonary tumors in germfree mice: induction with urethan. J Natl Cancer Inst. 1970;44(1):211–4.PubMed
44.
go back to reference Reddy BS, Narisawa T, Weisburger JH. Colon carcinogenesis in germ-free rats with intrarectal 1,2-dimethylhydrazine and subcutaneous azoxymethane. Cancer Res. 1976;36(8):2874–6.PubMed Reddy BS, Narisawa T, Weisburger JH. Colon carcinogenesis in germ-free rats with intrarectal 1,2-dimethylhydrazine and subcutaneous azoxymethane. Cancer Res. 1976;36(8):2874–6.PubMed
45.
go back to reference Zhan Y, Chen PJ, Sadler WD, Wang F, Poe S, Núñez G, Eaton KA, Chen GY. Gut microbiota protects against gastrointestinal tumorigenesis caused by epithelial injury. Cancer Res. 2013;73(24):7199–210.PubMedCrossRef Zhan Y, Chen PJ, Sadler WD, Wang F, Poe S, Núñez G, Eaton KA, Chen GY. Gut microbiota protects against gastrointestinal tumorigenesis caused by epithelial injury. Cancer Res. 2013;73(24):7199–210.PubMedCrossRef
46.
48.
go back to reference Mo S, Ru H, Huang M, Cheng L, Mo X, Yan L. Oral-intestinal microbiota in colorectal cancer: inflammation and immunosuppression. J Inflamm Res. 2022;15:747–59.PubMedPubMedCentralCrossRef Mo S, Ru H, Huang M, Cheng L, Mo X, Yan L. Oral-intestinal microbiota in colorectal cancer: inflammation and immunosuppression. J Inflamm Res. 2022;15:747–59.PubMedPubMedCentralCrossRef
50.
go back to reference Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, Ojesina AI, Jung J, Bass AJ, Tabernero J, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22(2):292–8.PubMedPubMedCentralCrossRef Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, Ojesina AI, Jung J, Bass AJ, Tabernero J, et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012;22(2):292–8.PubMedPubMedCentralCrossRef
51.
go back to reference Drewes JL, White JR, Dejea CM, Fathi P, Iyadorai T, Vadivelu J, Roslani AC, Wick EC, Mongodin EF, Loke MF, et al. High-resolution bacterial 16S rRNA gene profile meta-analysis and biofilm status reveal common colorectal cancer consortia. NPJ Biofilms Microbiomes. 2017;3:34.PubMedPubMedCentralCrossRef Drewes JL, White JR, Dejea CM, Fathi P, Iyadorai T, Vadivelu J, Roslani AC, Wick EC, Mongodin EF, Loke MF, et al. High-resolution bacterial 16S rRNA gene profile meta-analysis and biofilm status reveal common colorectal cancer consortia. NPJ Biofilms Microbiomes. 2017;3:34.PubMedPubMedCentralCrossRef
52.
go back to reference Komiya Y, Shimomura Y, Higurashi T, Sugi Y, Arimoto J, Umezawa S, Uchiyama S, Matsumoto M, Nakajima A. Patients with colorectal cancer have identical strains of Fusobacterium nucleatum in their colorectal cancer and oral cavity. Gut. 2019;68(7):1335–7.PubMedCrossRef Komiya Y, Shimomura Y, Higurashi T, Sugi Y, Arimoto J, Umezawa S, Uchiyama S, Matsumoto M, Nakajima A. Patients with colorectal cancer have identical strains of Fusobacterium nucleatum in their colorectal cancer and oral cavity. Gut. 2019;68(7):1335–7.PubMedCrossRef
53.
go back to reference Flemer B, Warren RD, Barrett MP, Cisek K, Das A, Jeffery IB, Hurley E, O’Riordain M, Shanahan F, O’Toole PW. The oral microbiota in colorectal cancer is distinctive and predictive. Gut. 2018;67(8):1454–63.PubMedCrossRef Flemer B, Warren RD, Barrett MP, Cisek K, Das A, Jeffery IB, Hurley E, O’Riordain M, Shanahan F, O’Toole PW. The oral microbiota in colorectal cancer is distinctive and predictive. Gut. 2018;67(8):1454–63.PubMedCrossRef
55.
57.
go back to reference Wong SH, Zhao L, Zhang X, Nakatsu G, Han J, Xu W, Xiao X, Kwong TNY, Tsoi H, Wu WKK, et al. Gavage of fecal samples from patients with colorectal cancer promotes intestinal carcinogenesis in germ-free and conventional mice. Gastroenterology. 2017;153(6):1621-33.e6.PubMedCrossRef Wong SH, Zhao L, Zhang X, Nakatsu G, Han J, Xu W, Xiao X, Kwong TNY, Tsoi H, Wu WKK, et al. Gavage of fecal samples from patients with colorectal cancer promotes intestinal carcinogenesis in germ-free and conventional mice. Gastroenterology. 2017;153(6):1621-33.e6.PubMedCrossRef
58.
go back to reference Parhi L, Alon-Maimon T, Sol A, Nejman D, Shhadeh A, Fainsod-Levi T, Yajuk O, Isaacson B, Abed J, Maalouf N, et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat Commun. 2020;11(1):3259.PubMedPubMedCentralCrossRef Parhi L, Alon-Maimon T, Sol A, Nejman D, Shhadeh A, Fainsod-Levi T, Yajuk O, Isaacson B, Abed J, Maalouf N, et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat Commun. 2020;11(1):3259.PubMedPubMedCentralCrossRef
59.
go back to reference Abed J, Emgård JE, Zamir G, Faroja M, Almogy G, Grenov A, Sol A, Naor R, Pikarsky E, Atlan KA, et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed gal-GalNAc. Cell Host Microbe. 2016;20(2):215–25.PubMedPubMedCentralCrossRef Abed J, Emgård JE, Zamir G, Faroja M, Almogy G, Grenov A, Sol A, Naor R, Pikarsky E, Atlan KA, et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed gal-GalNAc. Cell Host Microbe. 2016;20(2):215–25.PubMedPubMedCentralCrossRef
61.
go back to reference Gagliani N, Hu B, Huber S, Elinav E, Flavell RA. The fire within: microbes inflame tumors. Cell. 2014;157(4):776–83.PubMedCrossRef Gagliani N, Hu B, Huber S, Elinav E, Flavell RA. The fire within: microbes inflame tumors. Cell. 2014;157(4):776–83.PubMedCrossRef
62.
go back to reference Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14(2):195–206.PubMedPubMedCentralCrossRef Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. 2013;14(2):195–206.PubMedPubMedCentralCrossRef
63.
go back to reference Guo P, Tian Z, Kong X, Yang L, Shan X, Dong B, Ding X, Jing X, Jiang C, Jiang N, et al. FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J Exp Clin Cancer Res. 2020;39(1):202.PubMedPubMedCentralCrossRef Guo P, Tian Z, Kong X, Yang L, Shan X, Dong B, Ding X, Jing X, Jiang C, Jiang N, et al. FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J Exp Clin Cancer Res. 2020;39(1):202.PubMedPubMedCentralCrossRef
65.
go back to reference Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009;30(7):1073–81.PubMedCrossRef Colotta F, Allavena P, Sica A, Garlanda C, Mantovani A. Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis. 2009;30(7):1073–81.PubMedCrossRef
66.
68.
go back to reference Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013;13(11):759–71.PubMedCrossRef Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat Rev Cancer. 2013;13(11):759–71.PubMedCrossRef
69.
go back to reference Wei Z, Cao S, Liu S, Yao Z, Sun T, Li Y, Li J, Zhang D, Zhou Y. Could gut microbiota serve as prognostic biomarker associated with colorectal cancer patients’ survival? A pilot study on relevant mechanism. Oncotarget. 2016;7(29):46158–72.PubMedPubMedCentralCrossRef Wei Z, Cao S, Liu S, Yao Z, Sun T, Li Y, Li J, Zhang D, Zhou Y. Could gut microbiota serve as prognostic biomarker associated with colorectal cancer patients’ survival? A pilot study on relevant mechanism. Oncotarget. 2016;7(29):46158–72.PubMedPubMedCentralCrossRef
70.
go back to reference Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118(2):229–41.PubMedCrossRef Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell. 2004;118(2):229–41.PubMedCrossRef
71.
go back to reference Fukata M, Chen A, Vamadevan AS, Cohen J, Breglio K, Krishnareddy S, Hsu D, Xu R, Harpaz N, Dannenberg AJ, et al. Toll-like receptor-4 promotes the development of colitis-associated colorectal tumors. Gastroenterology. 2007;133(6):1869–81.PubMedCrossRef Fukata M, Chen A, Vamadevan AS, Cohen J, Breglio K, Krishnareddy S, Hsu D, Xu R, Harpaz N, Dannenberg AJ, et al. Toll-like receptor-4 promotes the development of colitis-associated colorectal tumors. Gastroenterology. 2007;133(6):1869–81.PubMedCrossRef
72.
go back to reference Rakoff-Nahoum S, Medzhitov R. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science. 2007;317(5834):124–7.PubMedCrossRef Rakoff-Nahoum S, Medzhitov R. Regulation of spontaneous intestinal tumorigenesis through the adaptor protein MyD88. Science. 2007;317(5834):124–7.PubMedCrossRef
73.
go back to reference Cianci R, Franza L, Schinzari G, Rossi E, Ianiro G, Tortora G, Gasbarrini A, Gambassi G, Cammarota G. The interplay between immunity and microbiota at intestinal immunological niche: the case of cancer. Int J Mol Sci. 2019;20(3):501.PubMedCentralCrossRef Cianci R, Franza L, Schinzari G, Rossi E, Ianiro G, Tortora G, Gasbarrini A, Gambassi G, Cammarota G. The interplay between immunity and microbiota at intestinal immunological niche: the case of cancer. Int J Mol Sci. 2019;20(3):501.PubMedCentralCrossRef
74.
go back to reference Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004;118(3):285–96.PubMedCrossRef Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, Kagnoff MF, Karin M. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell. 2004;118(3):285–96.PubMedCrossRef
75.
go back to reference Karin M, Cao Y, Greten FR, Li ZW. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2(4):301–10.PubMedCrossRef Karin M, Cao Y, Greten FR, Li ZW. NF-kappaB in cancer: from innocent bystander to major culprit. Nat Rev Cancer. 2002;2(4):301–10.PubMedCrossRef
76.
go back to reference Qu B, Liu BR, Du YJ, Chen J, Cheng YQ, Xu W, Wang XH. Wnt/β-catenin signaling pathway may regulate the expression of angiogenic growth factors in hepatocellular carcinoma. Oncol Lett. 2014;7(4):1175–8.PubMedPubMedCentralCrossRef Qu B, Liu BR, Du YJ, Chen J, Cheng YQ, Xu W, Wang XH. Wnt/β-catenin signaling pathway may regulate the expression of angiogenic growth factors in hepatocellular carcinoma. Oncol Lett. 2014;7(4):1175–8.PubMedPubMedCentralCrossRef
77.
go back to reference Allen IC, TeKippe EM, Woodford RM, Uronis JM, Holl EK, Rogers AB, Herfarth HH, Jobin C, Ting JP. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med. 2010;207(5):1045–56.PubMedPubMedCentralCrossRef Allen IC, TeKippe EM, Woodford RM, Uronis JM, Holl EK, Rogers AB, Herfarth HH, Jobin C, Ting JP. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med. 2010;207(5):1045–56.PubMedPubMedCentralCrossRef
78.
go back to reference Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145(5):745–57.PubMedPubMedCentralCrossRef Elinav E, Strowig T, Kau AL, Henao-Mejia J, Thaiss CA, Booth CJ, Peaper DR, Bertin J, Eisenbarth SC, Gordon JI, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145(5):745–57.PubMedPubMedCentralCrossRef
79.
go back to reference Hu B, Elinav E, Huber S, Strowig T, Hao L, Hafemann A, Jin C, Wunderlich C, Wunderlich T, Eisenbarth SC, et al. Microbiota-induced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer. Proc Natl Acad Sci USA. 2013;110(24):9862–7.PubMedPubMedCentralCrossRef Hu B, Elinav E, Huber S, Strowig T, Hao L, Hafemann A, Jin C, Wunderlich C, Wunderlich T, Eisenbarth SC, et al. Microbiota-induced activation of epithelial IL-6 signaling links inflammasome-driven inflammation with transmissible cancer. Proc Natl Acad Sci USA. 2013;110(24):9862–7.PubMedPubMedCentralCrossRef
80.
go back to reference Hu B, Friedman G, Elinav E, Flavell RA. Transmissible inflammation-induced colorectal cancer in inflammasome-deficient mice. Oncoimmunology. 2019;8(10): e981995.PubMedCrossRef Hu B, Friedman G, Elinav E, Flavell RA. Transmissible inflammation-induced colorectal cancer in inflammasome-deficient mice. Oncoimmunology. 2019;8(10): e981995.PubMedCrossRef
81.
go back to reference Couturier-Maillard A, Secher T, Rehman A, Normand S, De Arcangelis A, Haesler R, Huot L, Grandjean T, Bressenot A, Delanoye-Crespin A, et al. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest. 2013;123(2):700–11.PubMedPubMedCentral Couturier-Maillard A, Secher T, Rehman A, Normand S, De Arcangelis A, Haesler R, Huot L, Grandjean T, Bressenot A, Delanoye-Crespin A, et al. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Invest. 2013;123(2):700–11.PubMedPubMedCentral
82.
go back to reference Petnicki-Ocwieja T, Hrncir T, Liu YJ, Biswas A, Hudcovic T, Tlaskalova-Hogenova H, Kobayashi KS. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci USA. 2009;106(37):15813–8.PubMedPubMedCentralCrossRef Petnicki-Ocwieja T, Hrncir T, Liu YJ, Biswas A, Hudcovic T, Tlaskalova-Hogenova H, Kobayashi KS. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci USA. 2009;106(37):15813–8.PubMedPubMedCentralCrossRef
83.
go back to reference Rehman A, Sina C, Gavrilova O, Häsler R, Ott S, Baines JF, Schreiber S, Rosenstiel P. Nod2 is essential for temporal development of intestinal microbial communities. Gut. 2011;60(10):1354–62.PubMedCrossRef Rehman A, Sina C, Gavrilova O, Häsler R, Ott S, Baines JF, Schreiber S, Rosenstiel P. Nod2 is essential for temporal development of intestinal microbial communities. Gut. 2011;60(10):1354–62.PubMedCrossRef
84.
go back to reference Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, Taniguchi K, Yu GY, Osterreicher CH, Hung KE, et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature. 2012;491(7423):254–8.PubMedPubMedCentralCrossRef Grivennikov SI, Wang K, Mucida D, Stewart CA, Schnabl B, Jauch D, Taniguchi K, Yu GY, Osterreicher CH, Hung KE, et al. Adenoma-linked barrier defects and microbial products drive IL-23/IL-17-mediated tumour growth. Nature. 2012;491(7423):254–8.PubMedPubMedCentralCrossRef
85.
go back to reference Tosolini M, Kirilovsky A, Mlecnik B, Fredriksen T, Mauger S, Bindea G, Berger A, Bruneval P, Fridman WH, Pagès F, et al. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. Cancer Res. 2011;71(4):1263–71.PubMedCrossRef Tosolini M, Kirilovsky A, Mlecnik B, Fredriksen T, Mauger S, Bindea G, Berger A, Bruneval P, Fridman WH, Pagès F, et al. Clinical impact of different classes of infiltrating T cytotoxic and helper cells (Th1, th2, treg, th17) in patients with colorectal cancer. Cancer Res. 2011;71(4):1263–71.PubMedCrossRef
86.
go back to reference Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, Yagita H, Ishii N, Evans R, Honda K, et al. ATP drives lamina propria T(H)17 cell differentiation. Nature. 2008;455(7214):808–12.PubMedCrossRef Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, Yagita H, Ishii N, Evans R, Honda K, et al. ATP drives lamina propria T(H)17 cell differentiation. Nature. 2008;455(7214):808–12.PubMedCrossRef
87.
go back to reference Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–98.PubMedPubMedCentralCrossRef Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–98.PubMedPubMedCentralCrossRef
88.
go back to reference Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, Huso DL, Brancati FL, Wick E, McAllister F, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med. 2009;15(9):1016–22.PubMedPubMedCentralCrossRef Wu S, Rhee KJ, Albesiano E, Rabizadeh S, Wu X, Yen HR, Huso DL, Brancati FL, Wick E, McAllister F, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med. 2009;15(9):1016–22.PubMedPubMedCentralCrossRef
89.
go back to reference Housseau F, Wu S, Wick EC, Fan H, Wu X, Llosa NJ, Smith KN, Tam A, Ganguly S, Wanyiri JW, et al. Redundant innate and adaptive sources of IL17 production drive colon tumorigenesis. Cancer Res. 2016;76(8):2115–24.PubMedPubMedCentralCrossRef Housseau F, Wu S, Wick EC, Fan H, Wu X, Llosa NJ, Smith KN, Tam A, Ganguly S, Wanyiri JW, et al. Redundant innate and adaptive sources of IL17 production drive colon tumorigenesis. Cancer Res. 2016;76(8):2115–24.PubMedPubMedCentralCrossRef
90.
go back to reference Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971–6.PubMedPubMedCentralCrossRef Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971–6.PubMedPubMedCentralCrossRef
91.
go back to reference Viaud S, Flament C, Zoubir M, Pautier P, LeCesne A, Ribrag V, Soria JC, Marty V, Vielh P, Robert C, et al. Cyclophosphamide induces differentiation of Th17 cells in cancer patients. Cancer Res. 2011;71(3):661–5.PubMedCrossRef Viaud S, Flament C, Zoubir M, Pautier P, LeCesne A, Ribrag V, Soria JC, Marty V, Vielh P, Robert C, et al. Cyclophosphamide induces differentiation of Th17 cells in cancer patients. Cancer Res. 2011;71(3):661–5.PubMedCrossRef
94.
go back to reference Marques HS, de Brito BB, da Silva FAF, Santos MLC, de Souza JCB, Correia TML, Lopes LW, Neres NSM, Dórea R, Dantas ACS, et al. Relationship between Th17 immune response and cancer. World J Clin Oncol. 2021;12(10):845–67.PubMedPubMedCentralCrossRef Marques HS, de Brito BB, da Silva FAF, Santos MLC, de Souza JCB, Correia TML, Lopes LW, Neres NSM, Dórea R, Dantas ACS, et al. Relationship between Th17 immune response and cancer. World J Clin Oncol. 2021;12(10):845–67.PubMedPubMedCentralCrossRef
96.
go back to reference Erdman SE, Rao VP, Poutahidis T, Ihrig MM, Ge Z, Feng Y, Tomczak M, Rogers AB, Horwitz BH, Fox JG. CD4(+)CD25(+) regulatory lymphocytes require interleukin 10 to interrupt colon carcinogenesis in mice. Cancer Res. 2003;63(18):6042–50.PubMed Erdman SE, Rao VP, Poutahidis T, Ihrig MM, Ge Z, Feng Y, Tomczak M, Rogers AB, Horwitz BH, Fox JG. CD4(+)CD25(+) regulatory lymphocytes require interleukin 10 to interrupt colon carcinogenesis in mice. Cancer Res. 2003;63(18):6042–50.PubMed
97.
go back to reference Erdman SE, Poutahidis T, Tomczak M, Rogers AB, Cormier K, Plank B, Horwitz BH, Fox JG. CD4+ CD25+ regulatory T lymphocytes inhibit microbially induced colon cancer in Rag2-deficient mice. Am J Pathol. 2003;162(2):691–702.PubMedPubMedCentralCrossRef Erdman SE, Poutahidis T, Tomczak M, Rogers AB, Cormier K, Plank B, Horwitz BH, Fox JG. CD4+ CD25+ regulatory T lymphocytes inhibit microbially induced colon cancer in Rag2-deficient mice. Am J Pathol. 2003;162(2):691–702.PubMedPubMedCentralCrossRef
98.
go back to reference Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453(7195):620–5.PubMedCrossRef Mazmanian SK, Round JL, Kasper DL. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature. 2008;453(7195):620–5.PubMedCrossRef
99.
go back to reference Dejea C, Wick E, Sears CL. Bacterial oncogenesis in the colon. Future Microbiol. 2013;8(4):445–60.PubMedCrossRef Dejea C, Wick E, Sears CL. Bacterial oncogenesis in the colon. Future Microbiol. 2013;8(4):445–60.PubMedCrossRef
100.
go back to reference Lax AJ. Opinion: bacterial toxins and cancer—a case to answer? Nat Rev Microbiol. 2005;3(4):343–9.PubMedCrossRef Lax AJ. Opinion: bacterial toxins and cancer—a case to answer? Nat Rev Microbiol. 2005;3(4):343–9.PubMedCrossRef
101.
go back to reference Hatakeyama M. Structure and function of Helicobacter pylori CagA, the first-identified bacterial protein involved in human cancer. Proc Jpn Acad Ser B Phys Biol Sci. 2017;93(4):196–219.PubMedPubMedCentralCrossRef Hatakeyama M. Structure and function of Helicobacter pylori CagA, the first-identified bacterial protein involved in human cancer. Proc Jpn Acad Ser B Phys Biol Sci. 2017;93(4):196–219.PubMedPubMedCentralCrossRef
103.
go back to reference Saadat I, Higashi H, Obuse C, Umeda M, Murata-Kamiya N, Saito Y, Lu H, Ohnishi N, Azuma T, Suzuki A, et al. Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt epithelial cell polarity. Nature. 2007;447(7142):330–3.PubMedCrossRef Saadat I, Higashi H, Obuse C, Umeda M, Murata-Kamiya N, Saito Y, Lu H, Ohnishi N, Azuma T, Suzuki A, et al. Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt epithelial cell polarity. Nature. 2007;447(7142):330–3.PubMedCrossRef
104.
go back to reference Ansari S, Yamaoka Y. Helicobacter pylori virulence factor cytotoxin-associated gene A (CagA)-mediated gastric pathogenicity. Int J Mol Sci. 2020;21(19):7430.PubMedCentralCrossRef Ansari S, Yamaoka Y. Helicobacter pylori virulence factor cytotoxin-associated gene A (CagA)-mediated gastric pathogenicity. Int J Mol Sci. 2020;21(19):7430.PubMedCentralCrossRef
105.
go back to reference Hatakeyama M. Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis. Cell Host Microbe. 2014;15(3):306–16.PubMedCrossRef Hatakeyama M. Helicobacter pylori CagA and gastric cancer: a paradigm for hit-and-run carcinogenesis. Cell Host Microbe. 2014;15(3):306–16.PubMedCrossRef
106.
go back to reference Kim JM, Cho SJ, Oh YK, Jung HY, Kim YJ, Kim N. Nuclear factor-kappa B activation pathway in intestinal epithelial cells is a major regulator of chemokine gene expression and neutrophil migration induced by Bacteroides fragilis enterotoxin. Clin Exp Immunol. 2002;130(1):59–66.PubMedPubMedCentralCrossRef Kim JM, Cho SJ, Oh YK, Jung HY, Kim YJ, Kim N. Nuclear factor-kappa B activation pathway in intestinal epithelial cells is a major regulator of chemokine gene expression and neutrophil migration induced by Bacteroides fragilis enterotoxin. Clin Exp Immunol. 2002;130(1):59–66.PubMedPubMedCentralCrossRef
107.
go back to reference Sanfilippo L, Li CK, Seth R, Balwin TJ, Menozzi MG, Mahida YR. Bacteroides fragilis enterotoxin induces the expression of IL-8 and transforming growth factor-beta (TGF-beta) by human colonic epithelial cells. Clin Exp Immunol. 2000;119(3):456–63.PubMedPubMedCentralCrossRef Sanfilippo L, Li CK, Seth R, Balwin TJ, Menozzi MG, Mahida YR. Bacteroides fragilis enterotoxin induces the expression of IL-8 and transforming growth factor-beta (TGF-beta) by human colonic epithelial cells. Clin Exp Immunol. 2000;119(3):456–63.PubMedPubMedCentralCrossRef
108.
go back to reference Purcell RV, Pearson J, Aitchison A, Dixon L, Frizelle FA, Keenan JI. Colonization with enterotoxigenic Bacteroides fragilis is associated with early-stage colorectal neoplasia. PLoS ONE. 2017;12(2): e0171602.PubMedPubMedCentralCrossRef Purcell RV, Pearson J, Aitchison A, Dixon L, Frizelle FA, Keenan JI. Colonization with enterotoxigenic Bacteroides fragilis is associated with early-stage colorectal neoplasia. PLoS ONE. 2017;12(2): e0171602.PubMedPubMedCentralCrossRef
109.
go back to reference Goodwin AC, Destefano Shields CE, Wu S, Huso DL, Wu X, Murray-Stewart TR, Hacker-Prietz A, Rabizadeh S, Woster PM, Sears CL, et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc Natl Acad Sci USA. 2011;108(37):15354–9.PubMedPubMedCentralCrossRef Goodwin AC, Destefano Shields CE, Wu S, Huso DL, Wu X, Murray-Stewart TR, Hacker-Prietz A, Rabizadeh S, Woster PM, Sears CL, et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc Natl Acad Sci USA. 2011;108(37):15354–9.PubMedPubMedCentralCrossRef
110.
go back to reference Weikel CS, Grieco FD, Reuben J, Myers LL, Sack RB. Human colonic epithelial cells, HT29/C1, treated with crude Bacteroides fragilis enterotoxin dramatically alter their morphology. Infect Immun. 1992;60(2):321–7.PubMedPubMedCentralCrossRef Weikel CS, Grieco FD, Reuben J, Myers LL, Sack RB. Human colonic epithelial cells, HT29/C1, treated with crude Bacteroides fragilis enterotoxin dramatically alter their morphology. Infect Immun. 1992;60(2):321–7.PubMedPubMedCentralCrossRef
111.
go back to reference Wu S, Lim KC, Huang J, Saidi RF, Sears CL. Bacteroides fragilis enterotoxin cleaves the zonula adherens protein, E-cadherin. Proc Natl Acad Sci USA. 1998;95(25):14979–84.PubMedPubMedCentralCrossRef Wu S, Lim KC, Huang J, Saidi RF, Sears CL. Bacteroides fragilis enterotoxin cleaves the zonula adherens protein, E-cadherin. Proc Natl Acad Sci USA. 1998;95(25):14979–84.PubMedPubMedCentralCrossRef
112.
go back to reference Wu S, Morin PJ, Maouyo D, Sears CL. Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation. Gastroenterology. 2003;124(2):392–400.PubMedCrossRef Wu S, Morin PJ, Maouyo D, Sears CL. Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation. Gastroenterology. 2003;124(2):392–400.PubMedCrossRef
113.
go back to reference Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014;12(10):661–72.PubMedCrossRef Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014;12(10):661–72.PubMedCrossRef
114.
go back to reference Buda A, Qualtrough D, Jepson MA, Martines D, Paraskeva C, Pignatelli M. Butyrate downregulates alpha2beta1 integrin: a possible role in the induction of apoptosis in colorectal cancer cell lines. Gut. 2003;52(5):729–34.PubMedPubMedCentralCrossRef Buda A, Qualtrough D, Jepson MA, Martines D, Paraskeva C, Pignatelli M. Butyrate downregulates alpha2beta1 integrin: a possible role in the induction of apoptosis in colorectal cancer cell lines. Gut. 2003;52(5):729–34.PubMedPubMedCentralCrossRef
115.
go back to reference Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–73.PubMedCrossRef Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–73.PubMedCrossRef
116.
go back to reference Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–50.PubMedCrossRef Furusawa Y, Obata Y, Fukuda S, Endo TA, Nakato G, Takahashi D, Nakanishi Y, Uetake C, Kato K, Kato T, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504(7480):446–50.PubMedCrossRef
117.
go back to reference Ewaschuk JB, Walker JW, Diaz H, Madsen KL. Bioproduction of conjugated linoleic acid by probiotic bacteria occurs in vitro and in vivo in mice. J Nutr. 2006;136(6):1483–7.PubMedCrossRef Ewaschuk JB, Walker JW, Diaz H, Madsen KL. Bioproduction of conjugated linoleic acid by probiotic bacteria occurs in vitro and in vivo in mice. J Nutr. 2006;136(6):1483–7.PubMedCrossRef
118.
go back to reference Maggiora M, Bologna M, Cerù MP, Possati L, Angelucci A, Cimini A, Miglietta A, Bozzo F, Margiotta C, Muzio G, et al. An overview of the effect of linoleic and conjugated-linoleic acids on the growth of several human tumor cell lines. Int J Cancer. 2004;112(6):909–19.PubMedCrossRef Maggiora M, Bologna M, Cerù MP, Possati L, Angelucci A, Cimini A, Miglietta A, Bozzo F, Margiotta C, Muzio G, et al. An overview of the effect of linoleic and conjugated-linoleic acids on the growth of several human tumor cell lines. Int J Cancer. 2004;112(6):909–19.PubMedCrossRef
119.
go back to reference Nakashima C, Shingo K, Fujiwara-Tani R, Luo Y, Kawahara I, Goto K, Sasaki T, Fujii K, Ohmori H, Kuniyasu H. Expression of long-chain fatty acid receptor GPR40 is associated with cancer progression in colorectal cancer: a retrospective study. Oncol Lett. 2018;15(6):8641–6.PubMedPubMedCentral Nakashima C, Shingo K, Fujiwara-Tani R, Luo Y, Kawahara I, Goto K, Sasaki T, Fujii K, Ohmori H, Kuniyasu H. Expression of long-chain fatty acid receptor GPR40 is associated with cancer progression in colorectal cancer: a retrospective study. Oncol Lett. 2018;15(6):8641–6.PubMedPubMedCentral
120.
go back to reference Tsvetikova SA, Koshel EI. Microbiota and cancer: host cellular mechanisms activated by gut microbial metabolites. Int J Med Microbiol. 2020;310(4): 151425.PubMedCrossRef Tsvetikova SA, Koshel EI. Microbiota and cancer: host cellular mechanisms activated by gut microbial metabolites. Int J Med Microbiol. 2020;310(4): 151425.PubMedCrossRef
121.
go back to reference Bernstein H, Bernstein C, Payne CM, Dvorak K. Bile acids as endogenous etiologic agents in gastrointestinal cancer. World J Gastroenterol. 2009;15(27):3329–40.PubMedPubMedCentralCrossRef Bernstein H, Bernstein C, Payne CM, Dvorak K. Bile acids as endogenous etiologic agents in gastrointestinal cancer. World J Gastroenterol. 2009;15(27):3329–40.PubMedPubMedCentralCrossRef
122.
go back to reference Pai R, Tarnawski AS, Tran T. Deoxycholic acid activates beta-catenin signaling pathway and increases colon cell cancer growth and invasiveness. Mol Biol Cell. 2004;15(5):2156–63.PubMedPubMedCentralCrossRef Pai R, Tarnawski AS, Tran T. Deoxycholic acid activates beta-catenin signaling pathway and increases colon cell cancer growth and invasiveness. Mol Biol Cell. 2004;15(5):2156–63.PubMedPubMedCentralCrossRef
123.
go back to reference Jain T, Sharma P, Are AC, Vickers SM, Dudeja V. New insights into the cancer-microbiome-immune axis: decrypting a decade of discoveries. Front Immunol. 2021;12: 622064.PubMedPubMedCentralCrossRef Jain T, Sharma P, Are AC, Vickers SM, Dudeja V. New insights into the cancer-microbiome-immune axis: decrypting a decade of discoveries. Front Immunol. 2021;12: 622064.PubMedPubMedCentralCrossRef
124.
go back to reference Fluckiger A, Daillère R, Sassi M, Sixt BS, Liu P, Loos F, Richard C, Rabu C, Alou MT, Goubet AG, et al. Cross-reactivity between tumor MHC class I-restricted antigens and an enterococcal bacteriophage. Science. 2020;369(6506):936–42.PubMedCrossRef Fluckiger A, Daillère R, Sassi M, Sixt BS, Liu P, Loos F, Richard C, Rabu C, Alou MT, Goubet AG, et al. Cross-reactivity between tumor MHC class I-restricted antigens and an enterococcal bacteriophage. Science. 2020;369(6506):936–42.PubMedCrossRef
125.
go back to reference Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–84.PubMedPubMedCentralCrossRef Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–84.PubMedPubMedCentralCrossRef
126.
go back to reference Shao Y, Zeng X. Molecular mechanisms of gut microbiota-associated colorectal carcinogenesis. Infect Microbes Dis. 2020;2(3):96–106. Shao Y, Zeng X. Molecular mechanisms of gut microbiota-associated colorectal carcinogenesis. Infect Microbes Dis. 2020;2(3):96–106.
127.
go back to reference Buc E, Dubois D, Sauvanet P, Raisch J, Delmas J, Darfeuille-Michaud A, Pezet D, Bonnet R. High prevalence of mucosa-associated E. coli producing cyclomodulin and genotoxin in colon cancer. PLoS ONE. 2013;8(2): e56964.PubMedPubMedCentralCrossRef Buc E, Dubois D, Sauvanet P, Raisch J, Delmas J, Darfeuille-Michaud A, Pezet D, Bonnet R. High prevalence of mucosa-associated E. coli producing cyclomodulin and genotoxin in colon cancer. PLoS ONE. 2013;8(2): e56964.PubMedPubMedCentralCrossRef
128.
go back to reference Dalmasso G, Cougnoux A, Delmas J, Darfeuille-Michaud A, Bonnet R. The bacterial genotoxin colibactin promotes colon tumor growth by modifying the tumor microenvironment. Gut Microbes. 2014;5(5):675–80.PubMedPubMedCentralCrossRef Dalmasso G, Cougnoux A, Delmas J, Darfeuille-Michaud A, Bonnet R. The bacterial genotoxin colibactin promotes colon tumor growth by modifying the tumor microenvironment. Gut Microbes. 2014;5(5):675–80.PubMedPubMedCentralCrossRef
129.
go back to reference Cougnoux A, Dalmasso G, Martinez R, Buc E, Delmas J, Gibold L, Sauvanet P, Darcha C, Déchelotte P, Bonnet M, et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut. 2014;63(12):1932–42.PubMedCrossRef Cougnoux A, Dalmasso G, Martinez R, Buc E, Delmas J, Gibold L, Sauvanet P, Darcha C, Déchelotte P, Bonnet M, et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut. 2014;63(12):1932–42.PubMedCrossRef
130.
go back to reference Seely KD, Morgan AD, Hagenstein LD, Florey GM, Small JM. Bacterial involvement in progression and metastasis of colorectal neoplasia. Cancers (Basel). 2022;14(4):1019.PubMedPubMedCentralCrossRef Seely KD, Morgan AD, Hagenstein LD, Florey GM, Small JM. Bacterial involvement in progression and metastasis of colorectal neoplasia. Cancers (Basel). 2022;14(4):1019.PubMedPubMedCentralCrossRef
131.
go back to reference Whisner CM, Athena AC. The role of the microbiome in cancer initiation and progression: how microbes and cancer cells utilize excess energy and promote one another’s growth. Curr Nutr Rep. 2019;8(1):42–51.PubMedPubMedCentralCrossRef Whisner CM, Athena AC. The role of the microbiome in cancer initiation and progression: how microbes and cancer cells utilize excess energy and promote one another’s growth. Curr Nutr Rep. 2019;8(1):42–51.PubMedPubMedCentralCrossRef
132.
go back to reference Kich DM, Vincenzi A, Majolo F, Volken de Souza CF, Goettert MI. Probiotic: effectiveness nutrition in cancer treatment and prevention. Nutr Hosp. 2016;33(6):1430–7.PubMedCrossRef Kich DM, Vincenzi A, Majolo F, Volken de Souza CF, Goettert MI. Probiotic: effectiveness nutrition in cancer treatment and prevention. Nutr Hosp. 2016;33(6):1430–7.PubMedCrossRef
133.
go back to reference Górska A, Przystupski D, Niemczura MJ, Kulbacka J. Probiotic bacteria: a promising tool in cancer prevention and therapy. Curr Microbiol. 2019;76(8):939–49.PubMedPubMedCentralCrossRef Górska A, Przystupski D, Niemczura MJ, Kulbacka J. Probiotic bacteria: a promising tool in cancer prevention and therapy. Curr Microbiol. 2019;76(8):939–49.PubMedPubMedCentralCrossRef
134.
go back to reference Dos Reis SA, da Conceição LL, Siqueira NP, Rosa DD, da Silva LL, Peluzio MD. Review of the mechanisms of probiotic actions in the prevention of colorectal cancer. Nutr Res. 2017;37:1–19.PubMedCrossRef Dos Reis SA, da Conceição LL, Siqueira NP, Rosa DD, da Silva LL, Peluzio MD. Review of the mechanisms of probiotic actions in the prevention of colorectal cancer. Nutr Res. 2017;37:1–19.PubMedCrossRef
135.
go back to reference Eslami M, Yousefi B, Kokhaei P, Hemati M, Nejad ZR, Arabkari V, Namdar A. Importance of probiotics in the prevention and treatment of colorectal cancer. J Cell Physiol. 2019;234(10):17127–43.PubMedCrossRef Eslami M, Yousefi B, Kokhaei P, Hemati M, Nejad ZR, Arabkari V, Namdar A. Importance of probiotics in the prevention and treatment of colorectal cancer. J Cell Physiol. 2019;234(10):17127–43.PubMedCrossRef
137.
go back to reference Mima K, Sukawa Y, Nishihara R, Qian ZR, Yamauchi M, Inamura K, Kim SA, Masuda A, Nowak JA, Nosho K, et al. Fusobacterium nucleatum and T cells in colorectal carcinoma. JAMA Oncol. 2015;1(5):653–61.PubMedPubMedCentralCrossRef Mima K, Sukawa Y, Nishihara R, Qian ZR, Yamauchi M, Inamura K, Kim SA, Masuda A, Nowak JA, Nosho K, et al. Fusobacterium nucleatum and T cells in colorectal carcinoma. JAMA Oncol. 2015;1(5):653–61.PubMedPubMedCentralCrossRef
138.
go back to reference Gur C, Ibrahim Y, Isaacson B, Yamin R, Abed J, Gamliel M, Enk J, Bar-On Y, Stanietsky-Kaynan N, Coppenhagen-Glazer S, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42(2):344–55.PubMedPubMedCentralCrossRef Gur C, Ibrahim Y, Isaacson B, Yamin R, Abed J, Gamliel M, Enk J, Bar-On Y, Stanietsky-Kaynan N, Coppenhagen-Glazer S, et al. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42(2):344–55.PubMedPubMedCentralCrossRef
141.
go back to reference Zhang W, Wen K, Azevedo MS, Gonzalez A, Saif LJ, Li G, Yousef AE, Yuan L. Lactic acid bacterial colonization and human rotavirus infection influence distribution and frequencies of monocytes/macrophages and dendritic cells in neonatal gnotobiotic pigs. Vet Immunol Immunopathol. 2008;121(3–4):222–31.PubMedCrossRef Zhang W, Wen K, Azevedo MS, Gonzalez A, Saif LJ, Li G, Yousef AE, Yuan L. Lactic acid bacterial colonization and human rotavirus infection influence distribution and frequencies of monocytes/macrophages and dendritic cells in neonatal gnotobiotic pigs. Vet Immunol Immunopathol. 2008;121(3–4):222–31.PubMedCrossRef
142.
go back to reference Mørland B, Midtvedt T. Phagocytosis, peritoneal influx, and enzyme activities in peritoneal macrophages from germfree, conventional, and ex-germfree mice. Infect Immun. 1984;44(3):750–2.PubMedPubMedCentralCrossRef Mørland B, Midtvedt T. Phagocytosis, peritoneal influx, and enzyme activities in peritoneal macrophages from germfree, conventional, and ex-germfree mice. Infect Immun. 1984;44(3):750–2.PubMedPubMedCentralCrossRef
143.
go back to reference Ohkubo T, Tsuda M, Tamura M, Yamamura M. Impaired superoxide production in peripheral blood neutrophils of germ-free rats. Scand J Immunol. 1990;32(6):727–9.PubMedCrossRef Ohkubo T, Tsuda M, Tamura M, Yamamura M. Impaired superoxide production in peripheral blood neutrophils of germ-free rats. Scand J Immunol. 1990;32(6):727–9.PubMedCrossRef
144.
go back to reference Ohkubo T, Tsuda M, Suzuki S, El Borai N, Yamamura M. Peripheral blood neutrophils of germ-free rats modified by in vivo granulocyte-colony-stimulating factor and exposure to natural environment. Scand J Immunol. 1999;49(1):73–7.PubMedCrossRef Ohkubo T, Tsuda M, Suzuki S, El Borai N, Yamamura M. Peripheral blood neutrophils of germ-free rats modified by in vivo granulocyte-colony-stimulating factor and exposure to natural environment. Scand J Immunol. 1999;49(1):73–7.PubMedCrossRef
145.
go back to reference He X, Dong Y, Wu CW, Zhao Z, Ng SSM, Chan FKL, Sung JJY, Yu J. MicroRNA-218 inhibits cell cycle progression and promotes apoptosis in colon cancer by downregulating BMI1 polycomb ring finger oncogene. Mol Med (Cambridge, Mass). 2012;18(1):1491–8.CrossRef He X, Dong Y, Wu CW, Zhao Z, Ng SSM, Chan FKL, Sung JJY, Yu J. MicroRNA-218 inhibits cell cycle progression and promotes apoptosis in colon cancer by downregulating BMI1 polycomb ring finger oncogene. Mol Med (Cambridge, Mass). 2012;18(1):1491–8.CrossRef
146.
go back to reference Dzutsev A, Goldszmid RS, Viaud S, Zitvogel L, Trinchieri G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol. 2015;45(1):17–31.PubMedCrossRef Dzutsev A, Goldszmid RS, Viaud S, Zitvogel L, Trinchieri G. The role of the microbiota in inflammation, carcinogenesis, and cancer therapy. Eur J Immunol. 2015;45(1):17–31.PubMedCrossRef
147.
go back to reference Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci USA. 2011;108(13):5354–9.PubMedPubMedCentralCrossRef Ichinohe T, Pang IK, Kumamoto Y, Peaper DR, Ho JH, Murray TS, Iwasaki A. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci USA. 2011;108(13):5354–9.PubMedPubMedCentralCrossRef
148.
go back to reference Heymann CJF, Bard JM, Heymann MF, Heymann D, Bobin-Dubigeon C. The intratumoral microbiome: characterization methods and functional impact. Cancer Lett. 2021;522:63–79.PubMedCrossRef Heymann CJF, Bard JM, Heymann MF, Heymann D, Bobin-Dubigeon C. The intratumoral microbiome: characterization methods and functional impact. Cancer Lett. 2021;522:63–79.PubMedCrossRef
149.
go back to reference Paulos CM, Wrzesinski C, Kaiser A, Hinrichs CS, Chieppa M, Cassard L, Palmer DC, Boni A, Muranski P, Yu Z, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest. 2007;117(8):2197–204.PubMedPubMedCentralCrossRef Paulos CM, Wrzesinski C, Kaiser A, Hinrichs CS, Chieppa M, Cassard L, Palmer DC, Boni A, Muranski P, Yu Z, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest. 2007;117(8):2197–204.PubMedPubMedCentralCrossRef
150.
go back to reference Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356–65.PubMedCrossRef Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356–65.PubMedCrossRef
151.
go back to reference Daillère R, Vétizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, Duong CPM, Flament C, Lepage P, Roberti MP, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016;45(4):931–43.PubMedCrossRef Daillère R, Vétizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, Duong CPM, Flament C, Lepage P, Roberti MP, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016;45(4):931–43.PubMedCrossRef
152.
go back to reference Wilson ID, Nicholson JK. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl Res. 2017;179:204–22.PubMedCrossRef Wilson ID, Nicholson JK. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl Res. 2017;179:204–22.PubMedCrossRef
153.
go back to reference Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967–70.PubMedPubMedCentralCrossRef Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967–70.PubMedPubMedCentralCrossRef
154.
go back to reference Gori S, Inno A, Belluomini L, Bocus P, Bisoffi Z, Russo A, Arcaro G. Gut microbiota and cancer: how gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit Rev Oncol Hematol. 2019;143:139–47.PubMedCrossRef Gori S, Inno A, Belluomini L, Bocus P, Bisoffi Z, Russo A, Arcaro G. Gut microbiota and cancer: how gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit Rev Oncol Hematol. 2019;143:139–47.PubMedCrossRef
155.
go back to reference van Vliet MJ, Tissing WJ, Dun CA, Meessen NE, Kamps WA, de Bont ES, Harmsen HJ. Chemotherapy treatment in pediatric patients with acute myeloid leukemia receiving antimicrobial prophylaxis leads to a relative increase of colonization with potentially pathogenic bacteria in the gut. Clin Infect Dis. 2009;49(2):262–70.PubMedCrossRef van Vliet MJ, Tissing WJ, Dun CA, Meessen NE, Kamps WA, de Bont ES, Harmsen HJ. Chemotherapy treatment in pediatric patients with acute myeloid leukemia receiving antimicrobial prophylaxis leads to a relative increase of colonization with potentially pathogenic bacteria in the gut. Clin Infect Dis. 2009;49(2):262–70.PubMedCrossRef
156.
go back to reference Yang J, Liu KX, Qu JM, Wang XD. The changes induced by cyclophosphamide in intestinal barrier and microflora in mice. Eur J Pharmacol. 2013;714(1–3):120–4.PubMedCrossRef Yang J, Liu KX, Qu JM, Wang XD. The changes induced by cyclophosphamide in intestinal barrier and microflora in mice. Eur J Pharmacol. 2013;714(1–3):120–4.PubMedCrossRef
157.
go back to reference Panebianco C, Andriulli A, Pazienza V. Pharmacomicrobiomics: exploiting the drug-microbiota interactions in anticancer therapies. Microbiome. 2018;6(1):92.PubMedPubMedCentralCrossRef Panebianco C, Andriulli A, Pazienza V. Pharmacomicrobiomics: exploiting the drug-microbiota interactions in anticancer therapies. Microbiome. 2018;6(1):92.PubMedPubMedCentralCrossRef
158.
go back to reference Ohigashi S, Sudo K, Kobayashi D, Takahashi T, Nomoto K, Onodera H. Significant changes in the intestinal environment after surgery in patients with colorectal cancer. J Gastrointest Surg. 2013;17(9):1657–64.PubMedCrossRef Ohigashi S, Sudo K, Kobayashi D, Takahashi T, Nomoto K, Onodera H. Significant changes in the intestinal environment after surgery in patients with colorectal cancer. J Gastrointest Surg. 2013;17(9):1657–64.PubMedCrossRef
159.
162.
go back to reference Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre ML, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084–9.PubMedPubMedCentralCrossRef Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre ML, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084–9.PubMedPubMedCentralCrossRef
163.
go back to reference Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.PubMedCrossRef Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–7.PubMedCrossRef
164.
go back to reference Mager LF, Burkhard R, Pett N, Cooke NCA, Brown K, Ramay H, Paik S, Stagg J, Groves RA, Gallo M, et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science. 2020;369(6510):1481–9.PubMedCrossRef Mager LF, Burkhard R, Pett N, Cooke NCA, Brown K, Ramay H, Paik S, Stagg J, Groves RA, Gallo M, et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science. 2020;369(6510):1481–9.PubMedCrossRef
165.
go back to reference Tanoue T, Morita S, Plichta DR, Skelly AN, Suda W, Sugiura Y, Narushima S, Vlamakis H, Motoo I, Sugita K, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019;565(7741):600–5.PubMedCrossRef Tanoue T, Morita S, Plichta DR, Skelly AN, Suda W, Sugiura Y, Narushima S, Vlamakis H, Motoo I, Sugita K, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019;565(7741):600–5.PubMedCrossRef
166.
go back to reference Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.PubMedCrossRef Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.PubMedCrossRef
167.
go back to reference Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8.PubMedPubMedCentralCrossRef Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–8.PubMedPubMedCentralCrossRef
169.
go back to reference Yi M, Jiao D, Qin S, Chu Q, Li A, Wu K. Manipulating gut microbiota composition to enhance the therapeutic effect of cancer immunotherapy. Integr Cancer Ther. 2019;18:1534735419876351.PubMedPubMedCentralCrossRef Yi M, Jiao D, Qin S, Chu Q, Li A, Wu K. Manipulating gut microbiota composition to enhance the therapeutic effect of cancer immunotherapy. Integr Cancer Ther. 2019;18:1534735419876351.PubMedPubMedCentralCrossRef
170.
171.
172.
go back to reference BouZerdan M, Niforatos S, Nasr S, Nasr D, Ombada M, John S, Dutta D, Lim SH. Fecal microbiota transplant for hematologic and oncologic diseases: principle and practice. Cancers (Basel). 2022;14(3):691.CrossRef BouZerdan M, Niforatos S, Nasr S, Nasr D, Ombada M, John S, Dutta D, Lim SH. Fecal microbiota transplant for hematologic and oncologic diseases: principle and practice. Cancers (Basel). 2022;14(3):691.CrossRef
173.
go back to reference van Nood E, Speelman P, Nieuwdorp M, Keller J. Fecal microbiota transplantation: facts and controversies. Curr Opin Gastroenterol. 2014;30(1):34–9.PubMedCrossRef van Nood E, Speelman P, Nieuwdorp M, Keller J. Fecal microbiota transplantation: facts and controversies. Curr Opin Gastroenterol. 2014;30(1):34–9.PubMedCrossRef
174.
go back to reference Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer. 2017;17(5):271–85.PubMedCrossRef Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer. 2017;17(5):271–85.PubMedCrossRef
175.
go back to reference Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T. Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res. 1996;56(16):3752–7.PubMed Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T. Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res. 1996;56(16):3752–7.PubMed
176.
go back to reference Wallace BD, Wang H, Lane KT, Scott JE, Orans J, Koo JS, Venkatesh M, Jobin C, Yeh LA, Mani S, et al. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science. 2010;330(6005):831–5.PubMedPubMedCentralCrossRef Wallace BD, Wang H, Lane KT, Scott JE, Orans J, Koo JS, Venkatesh M, Jobin C, Yeh LA, Mani S, et al. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science. 2010;330(6005):831–5.PubMedPubMedCentralCrossRef
177.
go back to reference Mego M, Chovanec J, Vochyanova-Andrezalova I, Konkolovsky P, Mikulova M, Reckova M, Miskovska V, Bystricky B, Beniak J, Medvecova L, et al. Prevention of irinotecan induced diarrhea by probiotics: a randomized double blind, placebo controlled pilot study. Complement Ther Med. 2015;23(3):356–62.PubMedCrossRef Mego M, Chovanec J, Vochyanova-Andrezalova I, Konkolovsky P, Mikulova M, Reckova M, Miskovska V, Bystricky B, Beniak J, Medvecova L, et al. Prevention of irinotecan induced diarrhea by probiotics: a randomized double blind, placebo controlled pilot study. Complement Ther Med. 2015;23(3):356–62.PubMedCrossRef
178.
go back to reference Chitapanarux I, Chitapanarux T, Traisathit P, Kudumpee S, Tharavichitkul E, Lorvidhaya V. Randomized controlled trial of live Lactobacillus acidophilus plus Bifidobacterium bifidum in prophylaxis of diarrhea during radiotherapy in cervical cancer patients. Radiat Oncol. 2010;5:31.PubMedPubMedCentralCrossRef Chitapanarux I, Chitapanarux T, Traisathit P, Kudumpee S, Tharavichitkul E, Lorvidhaya V. Randomized controlled trial of live Lactobacillus acidophilus plus Bifidobacterium bifidum in prophylaxis of diarrhea during radiotherapy in cervical cancer patients. Radiat Oncol. 2010;5:31.PubMedPubMedCentralCrossRef
179.
go back to reference Wang F, Yin Q, Chen L, Davis MM. Bifidobacterium can mitigate intestinal immunopathology in the context of CTLA-4 blockade. Proc Natl Acad Sci USA. 2018;115(1):157–61.PubMedCrossRef Wang F, Yin Q, Chen L, Davis MM. Bifidobacterium can mitigate intestinal immunopathology in the context of CTLA-4 blockade. Proc Natl Acad Sci USA. 2018;115(1):157–61.PubMedCrossRef
180.
go back to reference Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Huang K, Guevara F, Nelson T, et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. 2017;358(6369):1443–8.PubMedPubMedCentralCrossRef Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Huang K, Guevara F, Nelson T, et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. 2017;358(6369):1443–8.PubMedPubMedCentralCrossRef
181.
go back to reference Chen Y, Chen Y, Zhang J, Cao P, Su W, Deng Y, Zhan N, Fu X, Huang Y, Dong W. Fusobacterium nucleatum promotes metastasis in colorectal cancer by activating autophagy signaling via the upregulation of CARD3 expression. Theranostics. 2020;10(1):323–39.PubMedPubMedCentralCrossRef Chen Y, Chen Y, Zhang J, Cao P, Su W, Deng Y, Zhan N, Fu X, Huang Y, Dong W. Fusobacterium nucleatum promotes metastasis in colorectal cancer by activating autophagy signaling via the upregulation of CARD3 expression. Theranostics. 2020;10(1):323–39.PubMedPubMedCentralCrossRef
182.
go back to reference Gur C, Maalouf N, Shhadeh A, Berhani O, Singer BB, Bachrach G, Mandelboim O. Fusobacterium nucleatum supresses anti-tumor immunity by activating CEACAM1. Oncoimmunology. 2019;8(6): e1581531.PubMedPubMedCentralCrossRef Gur C, Maalouf N, Shhadeh A, Berhani O, Singer BB, Bachrach G, Mandelboim O. Fusobacterium nucleatum supresses anti-tumor immunity by activating CEACAM1. Oncoimmunology. 2019;8(6): e1581531.PubMedPubMedCentralCrossRef
184.
go back to reference Chaurasia AK, Thorat ND, Tandon A, Kim JH, Park SH, Kim KK. Coupling of radiofrequency with magnetic nanoparticles treatment as an alternative physical antibacterial strategy against multiple drug resistant bacteria. Sci Rep. 2016;6:33662.PubMedPubMedCentralCrossRef Chaurasia AK, Thorat ND, Tandon A, Kim JH, Park SH, Kim KK. Coupling of radiofrequency with magnetic nanoparticles treatment as an alternative physical antibacterial strategy against multiple drug resistant bacteria. Sci Rep. 2016;6:33662.PubMedPubMedCentralCrossRef
185.
go back to reference Ying M, Yu Q, Zheng B, Wang H, Wang J, Chen S, Nie S, Xie M. Cultured Cordyceps sinensis polysaccharides modulate intestinal mucosal immunity and gut microbiota in cyclophosphamide-treated mice. Carbohydr Polym. 2020;235: 115957.PubMedCrossRef Ying M, Yu Q, Zheng B, Wang H, Wang J, Chen S, Nie S, Xie M. Cultured Cordyceps sinensis polysaccharides modulate intestinal mucosal immunity and gut microbiota in cyclophosphamide-treated mice. Carbohydr Polym. 2020;235: 115957.PubMedCrossRef
186.
187.
go back to reference Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin JM, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 2021;371(6529):595–602.PubMedPubMedCentralCrossRef Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin JM, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 2021;371(6529):595–602.PubMedPubMedCentralCrossRef
188.
go back to reference Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371(6529):602–9.PubMedCrossRef Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371(6529):602–9.PubMedCrossRef
190.
go back to reference Quemener V, Blanchard Y, Chamaillard L, Havouis R, Cipolla B, Moulinoux JP. Polyamine deprivation: a new tool in cancer treatment. Anticancer Res. 1994;14(2a):443–8.PubMed Quemener V, Blanchard Y, Chamaillard L, Havouis R, Cipolla B, Moulinoux JP. Polyamine deprivation: a new tool in cancer treatment. Anticancer Res. 1994;14(2a):443–8.PubMed
191.
go back to reference Tian T, Zhao Y, Yang Y, Wang T, Jin S, Guo J, Liu Z. The protective role of short-chain fatty acids acting as signal molecules in chemotherapy- or radiation-induced intestinal inflammation. Am J Cancer Res. 2020;10(11):3508–31.PubMedPubMedCentral Tian T, Zhao Y, Yang Y, Wang T, Jin S, Guo J, Liu Z. The protective role of short-chain fatty acids acting as signal molecules in chemotherapy- or radiation-induced intestinal inflammation. Am J Cancer Res. 2020;10(11):3508–31.PubMedPubMedCentral
192.
193.
go back to reference Jaye K, Li CG, Chang D, Bhuyan DJ. The role of key gut microbial metabolites in the development and treatment of cancer. Gut Microbes. 2022;14(1):2038865.PubMedPubMedCentralCrossRef Jaye K, Li CG, Chang D, Bhuyan DJ. The role of key gut microbial metabolites in the development and treatment of cancer. Gut Microbes. 2022;14(1):2038865.PubMedPubMedCentralCrossRef
195.
go back to reference Sieow BF, Wun KS, Yong WP, Hwang IY, Chang MW. Tweak to treat: reprograming bacteria for cancer treatment. Trends Cancer. 2021;7(5):447–64.PubMedCrossRef Sieow BF, Wun KS, Yong WP, Hwang IY, Chang MW. Tweak to treat: reprograming bacteria for cancer treatment. Trends Cancer. 2021;7(5):447–64.PubMedCrossRef
197.
go back to reference Jessup JM, Stewart A, Greene FL, Minsky BD. Adjuvant chemotherapy for stage III colon cancer: implications of race/ethnicity, age, and differentiation. JAMA. 2005;294(21):2703–11.PubMedCrossRef Jessup JM, Stewart A, Greene FL, Minsky BD. Adjuvant chemotherapy for stage III colon cancer: implications of race/ethnicity, age, and differentiation. JAMA. 2005;294(21):2703–11.PubMedCrossRef
198.
go back to reference Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, Diaz LA Jr. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 2019;16(6):361–75.PubMedPubMedCentralCrossRef Ganesh K, Stadler ZK, Cercek A, Mendelsohn RB, Shia J, Segal NH, Diaz LA Jr. Immunotherapy in colorectal cancer: rationale, challenges and potential. Nat Rev Gastroenterol Hepatol. 2019;16(6):361–75.PubMedPubMedCentralCrossRef
199.
go back to reference Newsome RC, Yang Y, Jobin C. The microbiome, gastrointestinal cancer, and immunotherapy. J Gastroenterol Hepatol. 2022;37(2):263–72.PubMedCrossRef Newsome RC, Yang Y, Jobin C. The microbiome, gastrointestinal cancer, and immunotherapy. J Gastroenterol Hepatol. 2022;37(2):263–72.PubMedCrossRef
200.
go back to reference Som A, Mandaliya R, Alsaadi D, Farshidpour M, Charabaty A, Malhotra N, Mattar MC. Immune checkpoint inhibitor-induced colitis: a comprehensive review. World J Clin Cases. 2019;7(4):405–18.PubMedPubMedCentralCrossRef Som A, Mandaliya R, Alsaadi D, Farshidpour M, Charabaty A, Malhotra N, Mattar MC. Immune checkpoint inhibitor-induced colitis: a comprehensive review. World J Clin Cases. 2019;7(4):405–18.PubMedPubMedCentralCrossRef
201.
go back to reference Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, Boselli L, Routier E, Cassard L, Collins M, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368–79.PubMedCrossRef Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, Boselli L, Routier E, Cassard L, Collins M, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368–79.PubMedCrossRef
202.
go back to reference Nomura M, Nagatomo R, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Inoue K, Muto M. Association of short-chain fatty acids in the gut microbiome with clinical response to treatment with nivolumab or pembrolizumab in patients with solid cancer tumors. JAMA Netw Open. 2020;3(4): e202895.PubMedPubMedCentralCrossRef Nomura M, Nagatomo R, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Inoue K, Muto M. Association of short-chain fatty acids in the gut microbiome with clinical response to treatment with nivolumab or pembrolizumab in patients with solid cancer tumors. JAMA Netw Open. 2020;3(4): e202895.PubMedPubMedCentralCrossRef
204.
go back to reference Veziant J, Villéger R, Barnich N, Bonnet M. Gut microbiota as potential biomarker and/or therapeutic target to improve the management of cancer: focus on colibactin-producing Escherichia coli in colorectal cancer. Cancers (Basel). 2021;13(9):2215.PubMedPubMedCentralCrossRef Veziant J, Villéger R, Barnich N, Bonnet M. Gut microbiota as potential biomarker and/or therapeutic target to improve the management of cancer: focus on colibactin-producing Escherichia coli in colorectal cancer. Cancers (Basel). 2021;13(9):2215.PubMedPubMedCentralCrossRef
205.
go back to reference Chen C, Niu M, Pan J, Du N, Liu S, Li H, He Q, Mao J, Duan Y, Du Y. Bacteroides, butyric acid and t10, c12-CLA changes in colorectal adenomatous polyp patients. Gut Pathog. 2021;13(1):1.PubMedPubMedCentralCrossRef Chen C, Niu M, Pan J, Du N, Liu S, Li H, He Q, Mao J, Duan Y, Du Y. Bacteroides, butyric acid and t10, c12-CLA changes in colorectal adenomatous polyp patients. Gut Pathog. 2021;13(1):1.PubMedPubMedCentralCrossRef
206.
go back to reference Xi Y, Yuefen P, Wei W, Quan Q, Jing Z, Jiamin X, Shuwen H. Analysis of prognosis, genome, microbiome, and microbial metabolome in different sites of colorectal cancer. J Transl Med. 2019;17(1):353.PubMedPubMedCentralCrossRef Xi Y, Yuefen P, Wei W, Quan Q, Jing Z, Jiamin X, Shuwen H. Analysis of prognosis, genome, microbiome, and microbial metabolome in different sites of colorectal cancer. J Transl Med. 2019;17(1):353.PubMedPubMedCentralCrossRef
207.
go back to reference Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS ONE. 2013;8(8): e70803.PubMedPubMedCentralCrossRef Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS ONE. 2013;8(8): e70803.PubMedPubMedCentralCrossRef
208.
go back to reference Yang Y, Misra BB, Liang L, Bi D, Weng W, Wu W, Cai S, Qin H, Goel A, Li X, et al. Integrated microbiome and metabolome analysis reveals a novel interplay between commensal bacteria and metabolites in colorectal cancer. Theranostics. 2019;9(14):4101–14.PubMedPubMedCentralCrossRef Yang Y, Misra BB, Liang L, Bi D, Weng W, Wu W, Cai S, Qin H, Goel A, Li X, et al. Integrated microbiome and metabolome analysis reveals a novel interplay between commensal bacteria and metabolites in colorectal cancer. Theranostics. 2019;9(14):4101–14.PubMedPubMedCentralCrossRef
210.
go back to reference Villéger R, Lopès A, Veziant J, Gagnière J, Barnich N, Billard E, Boucher D, Bonnet M. Microbial markers in colorectal cancer detection and/or prognosis. World J Gastroenterol. 2018;24(22):2327–47.PubMedPubMedCentralCrossRef Villéger R, Lopès A, Veziant J, Gagnière J, Barnich N, Billard E, Boucher D, Bonnet M. Microbial markers in colorectal cancer detection and/or prognosis. World J Gastroenterol. 2018;24(22):2327–47.PubMedPubMedCentralCrossRef
211.
212.
go back to reference Di Domenico EG, Cavallo I, Pontone M, Toma L, Ensoli F. Biofilm producing Salmonella Typhi: chronic colonization and development of gallbladder cancer. Int J Mol Sci. 2017;18(9):1887.PubMedCentralCrossRef Di Domenico EG, Cavallo I, Pontone M, Toma L, Ensoli F. Biofilm producing Salmonella Typhi: chronic colonization and development of gallbladder cancer. Int J Mol Sci. 2017;18(9):1887.PubMedCentralCrossRef
213.
go back to reference Wang F, Meng W, Wang B, Qiao L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 2014;345(2):196–202.PubMedCrossRef Wang F, Meng W, Wang B, Qiao L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 2014;345(2):196–202.PubMedCrossRef
214.
go back to reference Elkahwaji JE, Hauke RJ, Brawner CM. Chronic bacterial inflammation induces prostatic intraepithelial neoplasia in mouse prostate. Br J Cancer. 2009;101(10):1740–8.PubMedPubMedCentralCrossRef Elkahwaji JE, Hauke RJ, Brawner CM. Chronic bacterial inflammation induces prostatic intraepithelial neoplasia in mouse prostate. Br J Cancer. 2009;101(10):1740–8.PubMedPubMedCentralCrossRef
215.
go back to reference Simons BW, Durham NM, Bruno TC, Grosso JF, Schaeffer AJ, Ross AE, Hurley PJ, Berman DM, Drake CG, Thumbikat P, et al. A human prostatic bacterial isolate alters the prostatic microenvironment and accelerates prostate cancer progression. J Pathol. 2015;235(3):478–89.PubMedPubMedCentralCrossRef Simons BW, Durham NM, Bruno TC, Grosso JF, Schaeffer AJ, Ross AE, Hurley PJ, Berman DM, Drake CG, Thumbikat P, et al. A human prostatic bacterial isolate alters the prostatic microenvironment and accelerates prostate cancer progression. J Pathol. 2015;235(3):478–89.PubMedPubMedCentralCrossRef
216.
go back to reference El-Mosalamy H, Salman TM, Ashmawey AM, Osama N. Role of chronic E. coli infection in the process of bladder cancer—an experimental study. Infect Agent Cancer. 2012;7(1):19.PubMedPubMedCentralCrossRef El-Mosalamy H, Salman TM, Ashmawey AM, Osama N. Role of chronic E. coli infection in the process of bladder cancer—an experimental study. Infect Agent Cancer. 2012;7(1):19.PubMedPubMedCentralCrossRef
217.
go back to reference Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Abnet CC, Stolzenberg-Solomon R, Miller G, et al. Human oral microbiome and prospective risk for pancreatic cancer: a population-based nested case-control study. Gut. 2018;67(1):120–7.PubMedCrossRef Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Abnet CC, Stolzenberg-Solomon R, Miller G, et al. Human oral microbiome and prospective risk for pancreatic cancer: a population-based nested case-control study. Gut. 2018;67(1):120–7.PubMedCrossRef
218.
219.
go back to reference Zamani S, Taslimi R, Sarabi A, Jasemi S, Sechi LA, Feizabadi MM. Enterotoxigenic Bacteroides fragilis: a possible etiological candidate for bacterially-induced colorectal precancerous and cancerous lesions. Front Cell Infect Microbiol. 2019;9:449.PubMedCrossRef Zamani S, Taslimi R, Sarabi A, Jasemi S, Sechi LA, Feizabadi MM. Enterotoxigenic Bacteroides fragilis: a possible etiological candidate for bacterially-induced colorectal precancerous and cancerous lesions. Front Cell Infect Microbiol. 2019;9:449.PubMedCrossRef
220.
go back to reference Chen S, Su T, Zhang Y, Lee A, He J, Ge Q, Wang L, Si J, Zhuo W, Wang L. Fusobacterium nucleatum promotes colorectal cancer metastasis by modulating KRT7-AS/KRT7. Gut Microbes. 2020;11(3):511–25.PubMedPubMedCentralCrossRef Chen S, Su T, Zhang Y, Lee A, He J, Ge Q, Wang L, Si J, Zhuo W, Wang L. Fusobacterium nucleatum promotes colorectal cancer metastasis by modulating KRT7-AS/KRT7. Gut Microbes. 2020;11(3):511–25.PubMedPubMedCentralCrossRef
221.
go back to reference Chang C, Geng F, Shi X, Li Y, Zhang X, Zhao X, Pan Y. The prevalence rate of periodontal pathogens and its association with oral squamous cell carcinoma. Appl Microbiol Biotechnol. 2019;103(3):1393–404.PubMedCrossRef Chang C, Geng F, Shi X, Li Y, Zhang X, Zhao X, Pan Y. The prevalence rate of periodontal pathogens and its association with oral squamous cell carcinoma. Appl Microbiol Biotechnol. 2019;103(3):1393–404.PubMedCrossRef
222.
go back to reference Youssef O, Lahti L, Kokkola A, Karla T, Tikkanen M, Ehsan H, Carpelan-Holmström M, Koskensalo S, Böhling T, Rautelin H, et al. Stool microbiota composition differs in patients with stomach, colon, and rectal neoplasms. Dig Dis Sci. 2018;63(11):2950–8.PubMedPubMedCentralCrossRef Youssef O, Lahti L, Kokkola A, Karla T, Tikkanen M, Ehsan H, Carpelan-Holmström M, Koskensalo S, Böhling T, Rautelin H, et al. Stool microbiota composition differs in patients with stomach, colon, and rectal neoplasms. Dig Dis Sci. 2018;63(11):2950–8.PubMedPubMedCentralCrossRef
223.
go back to reference Yan X, Yang M, Liu J, Gao R, Hu J, Li J, Zhang L, Shi Y, Guo H, Cheng J, et al. Discovery and validation of potential bacterial biomarkers for lung cancer. Am J Cancer Res. 2015;5(10):3111–22.PubMedPubMedCentral Yan X, Yang M, Liu J, Gao R, Hu J, Li J, Zhang L, Shi Y, Guo H, Cheng J, et al. Discovery and validation of potential bacterial biomarkers for lung cancer. Am J Cancer Res. 2015;5(10):3111–22.PubMedPubMedCentral
224.
go back to reference Ohnishi N, Yuasa H, Tanaka S, Sawa H, Miura M, Matsui A, Higashi H, Musashi M, Iwabuchi K, Suzuki M, et al. Transgenic expression of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in mouse. Proc Natl Acad Sci USA. 2008;105(3):1003–8.PubMedPubMedCentralCrossRef Ohnishi N, Yuasa H, Tanaka S, Sawa H, Miura M, Matsui A, Higashi H, Musashi M, Iwabuchi K, Suzuki M, et al. Transgenic expression of Helicobacter pylori CagA induces gastrointestinal and hematopoietic neoplasms in mouse. Proc Natl Acad Sci USA. 2008;105(3):1003–8.PubMedPubMedCentralCrossRef
225.
go back to reference Higashi H, Tsutsumi R, Muto S, Sugiyama T, Azuma T, Asaka M, Hatakeyama M. SHP-2 tyrosine phosphatase as an intracellular target of Helicobacter pylori CagA protein. Science. 2002;295(5555):683–6.PubMedCrossRef Higashi H, Tsutsumi R, Muto S, Sugiyama T, Azuma T, Asaka M, Hatakeyama M. SHP-2 tyrosine phosphatase as an intracellular target of Helicobacter pylori CagA protein. Science. 2002;295(5555):683–6.PubMedCrossRef
226.
go back to reference Buti L, Ruiz-Puig C, Sangberg D, Leissing TM, Brewer RC, Owen RP, Sgromo B, Royer C, Ebner D, Lu X. CagA-ASPP2 complex mediates loss of cell polarity and favors H. pylori colonization of human gastric organoids. Proc Natl Acad Sci USA. 2020;117(5):2645–55.PubMedPubMedCentralCrossRef Buti L, Ruiz-Puig C, Sangberg D, Leissing TM, Brewer RC, Owen RP, Sgromo B, Royer C, Ebner D, Lu X. CagA-ASPP2 complex mediates loss of cell polarity and favors H. pylori colonization of human gastric organoids. Proc Natl Acad Sci USA. 2020;117(5):2645–55.PubMedPubMedCentralCrossRef
227.
go back to reference Chang YJ, Wu MS, Lin JT, Pestell RG, Blaser MJ, Chen CC. Mechanisms for Helicobacter pylori CagA-induced cyclin D1 expression that affect cell cycle. Cell Microbiol. 2006;8(11):1740–52.PubMedCrossRef Chang YJ, Wu MS, Lin JT, Pestell RG, Blaser MJ, Chen CC. Mechanisms for Helicobacter pylori CagA-induced cyclin D1 expression that affect cell cycle. Cell Microbiol. 2006;8(11):1740–52.PubMedCrossRef
228.
go back to reference Mimuro H, Suzuki T, Tanaka J, Asahi M, Haas R, Sasakawa C. Grb2 is a key mediator of Helicobacter pylori CagA protein activities. Mol Cell. 2002;10(4):745–55.PubMedCrossRef Mimuro H, Suzuki T, Tanaka J, Asahi M, Haas R, Sasakawa C. Grb2 is a key mediator of Helicobacter pylori CagA protein activities. Mol Cell. 2002;10(4):745–55.PubMedCrossRef
229.
go back to reference Costa AM, Ferreira RM, Pinto-Ribeiro I, Sougleri IS, Oliveira MJ, Carreto L, Santos MA, Sgouras DN, Carneiro F, Leite M, et al. Helicobacter pylori activates matrix metalloproteinase 10 in gastric epithelial cells via EGFR and ERK-mediated pathways. J Infect Dis. 2016;213(11):1767–76.PubMedCrossRef Costa AM, Ferreira RM, Pinto-Ribeiro I, Sougleri IS, Oliveira MJ, Carreto L, Santos MA, Sgouras DN, Carneiro F, Leite M, et al. Helicobacter pylori activates matrix metalloproteinase 10 in gastric epithelial cells via EGFR and ERK-mediated pathways. J Infect Dis. 2016;213(11):1767–76.PubMedCrossRef
230.
go back to reference Palrasu M, Zaika E, El-Rifai W, Garcia-Buitrago M, Piazuelo MB, Wilson KT, Peek RM Jr, Zaika AI. Bacterial CagA protein compromises tumor suppressor mechanisms in gastric epithelial cells. J Clin Invest. 2020;130(5):2422–34.PubMedPubMedCentralCrossRef Palrasu M, Zaika E, El-Rifai W, Garcia-Buitrago M, Piazuelo MB, Wilson KT, Peek RM Jr, Zaika AI. Bacterial CagA protein compromises tumor suppressor mechanisms in gastric epithelial cells. J Clin Invest. 2020;130(5):2422–34.PubMedPubMedCentralCrossRef
231.
go back to reference Liu B, Li X, Sun F, Tong X, Bai Y, Jin K, Liu L, Dai F, Li N. HP-CagA+ regulates the expression of CDK4/CyclinD1 via reg3 to change cell cycle and promote cell proliferation. Int J Mol Sci. 2019;21(1):224.PubMedCentralCrossRef Liu B, Li X, Sun F, Tong X, Bai Y, Jin K, Liu L, Dai F, Li N. HP-CagA+ regulates the expression of CDK4/CyclinD1 via reg3 to change cell cycle and promote cell proliferation. Int J Mol Sci. 2019;21(1):224.PubMedCentralCrossRef
232.
go back to reference Kuck D, Kolmerer B, Iking-Konert C, Krammer PH, Stremmel W, Rudi J. Vacuolating cytotoxin of Helicobacter pylori induces apoptosis in the human gastric epithelial cell line AGS. Infect Immun. 2001;69(8):5080–7.PubMedPubMedCentralCrossRef Kuck D, Kolmerer B, Iking-Konert C, Krammer PH, Stremmel W, Rudi J. Vacuolating cytotoxin of Helicobacter pylori induces apoptosis in the human gastric epithelial cell line AGS. Infect Immun. 2001;69(8):5080–7.PubMedPubMedCentralCrossRef
233.
go back to reference Seto K, Hayashi-Kuwabara Y, Yoneta T, Suda H, Tamaki H. Vacuolation induced by cytotoxin from Helicobacter pylori is mediated by the EGF receptor in HeLa cells. FEBS Lett. 1998;431(3):347–50.PubMedCrossRef Seto K, Hayashi-Kuwabara Y, Yoneta T, Suda H, Tamaki H. Vacuolation induced by cytotoxin from Helicobacter pylori is mediated by the EGF receptor in HeLa cells. FEBS Lett. 1998;431(3):347–50.PubMedCrossRef
234.
235.
go back to reference Wang F, Xia P, Wu F, Wang D, Wang W, Ward T, Liu Y, Aikhionbare F, Guo Z, Powell M, et al. Helicobacter pylori VacA disrupts apical membrane-cytoskeletal interactions in gastric parietal cells. J Biol Chem. 2008;283(39):26714–25.PubMedPubMedCentralCrossRef Wang F, Xia P, Wu F, Wang D, Wang W, Ward T, Liu Y, Aikhionbare F, Guo Z, Powell M, et al. Helicobacter pylori VacA disrupts apical membrane-cytoskeletal interactions in gastric parietal cells. J Biol Chem. 2008;283(39):26714–25.PubMedPubMedCentralCrossRef
236.
go back to reference Kobayashi H, Kamiya S, Suzuki T, Kohda K, Muramatsu S, Kurumada T, Ohta U, Miyazawa M, Kimura N, Mutoh N, et al. The effect of Helicobacter pylori on gastric acid secretion by isolated parietal cells from a guinea pig. Association with production of vacuolating toxin by H. pylori. Scand J Gastroenterol. 1996;31(5):428–33.PubMedCrossRef Kobayashi H, Kamiya S, Suzuki T, Kohda K, Muramatsu S, Kurumada T, Ohta U, Miyazawa M, Kimura N, Mutoh N, et al. The effect of Helicobacter pylori on gastric acid secretion by isolated parietal cells from a guinea pig. Association with production of vacuolating toxin by H. pylori. Scand J Gastroenterol. 1996;31(5):428–33.PubMedCrossRef
237.
go back to reference Gebert B, Fischer W, Weiss E, Hoffmann R, Haas R. Helicobacter pylori vacuolating cytotoxin inhibits T lymphocyte activation. Science. 2003;301(5636):1099–102.PubMedCrossRef Gebert B, Fischer W, Weiss E, Hoffmann R, Haas R. Helicobacter pylori vacuolating cytotoxin inhibits T lymphocyte activation. Science. 2003;301(5636):1099–102.PubMedCrossRef
238.
go back to reference Montecucco C, de Bernard M. Immunosuppressive and proinflammatory activities of the VacA toxin of Helicobacter pylori. J Exp Med. 2003;198(12):1767–71.PubMedPubMedCentralCrossRef Montecucco C, de Bernard M. Immunosuppressive and proinflammatory activities of the VacA toxin of Helicobacter pylori. J Exp Med. 2003;198(12):1767–71.PubMedPubMedCentralCrossRef
239.
go back to reference Radin JN, González-Rivera C, Ivie SE, McClain MS, Cover TL. Helicobacter pylori VacA induces programmed necrosis in gastric epithelial cells. Infect Immun. 2011;79(7):2535–43.PubMedPubMedCentralCrossRef Radin JN, González-Rivera C, Ivie SE, McClain MS, Cover TL. Helicobacter pylori VacA induces programmed necrosis in gastric epithelial cells. Infect Immun. 2011;79(7):2535–43.PubMedPubMedCentralCrossRef
240.
go back to reference Cover TL, Krishna US, Israel DA, Peek RM Jr. Induction of gastric epithelial cell apoptosis by Helicobacter pylori vacuolating cytotoxin. Cancer Res. 2003;63(5):951–7.PubMed Cover TL, Krishna US, Israel DA, Peek RM Jr. Induction of gastric epithelial cell apoptosis by Helicobacter pylori vacuolating cytotoxin. Cancer Res. 2003;63(5):951–7.PubMed
241.
go back to reference Radin JN, González-Rivera C, Frick-Cheng AE, Sheng J, Gaddy JA, Rubin DH, Algood HM, McClain MS, Cover TL. Role of connexin 43 in Helicobacter pylori VacA-induced cell death. Infect Immun. 2014;82(1):423–32.PubMedPubMedCentralCrossRef Radin JN, González-Rivera C, Frick-Cheng AE, Sheng J, Gaddy JA, Rubin DH, Algood HM, McClain MS, Cover TL. Role of connexin 43 in Helicobacter pylori VacA-induced cell death. Infect Immun. 2014;82(1):423–32.PubMedPubMedCentralCrossRef
242.
go back to reference Boncristiano M, Paccani SR, Barone S, Ulivieri C, Patrussi L, Ilver D, Amedei A, D’Elios MM, Telford JL, Baldari CT. The Helicobacter pylori vacuolating toxin inhibits T cell activation by two independent mechanisms. J Exp Med. 2003;198(12):1887–97.PubMedPubMedCentralCrossRef Boncristiano M, Paccani SR, Barone S, Ulivieri C, Patrussi L, Ilver D, Amedei A, D’Elios MM, Telford JL, Baldari CT. The Helicobacter pylori vacuolating toxin inhibits T cell activation by two independent mechanisms. J Exp Med. 2003;198(12):1887–97.PubMedPubMedCentralCrossRef
243.
go back to reference Seo B, Choy EW, Maudsley S, Miller WE, Wilson BA, Luttrell LM. Pasteurella multocida toxin stimulates mitogen-activated protein kinase via Gq/11-dependent transactivation of the epidermal growth factor receptor. J Biol Chem. 2000;275(3):2239–45.PubMedCrossRef Seo B, Choy EW, Maudsley S, Miller WE, Wilson BA, Luttrell LM. Pasteurella multocida toxin stimulates mitogen-activated protein kinase via Gq/11-dependent transactivation of the epidermal growth factor receptor. J Biol Chem. 2000;275(3):2239–45.PubMedCrossRef
244.
go back to reference Rozengurt E, Higgins T, Chanter N, Lax AJ, Staddon JM. Pasteurella multocida toxin: potent mitogen for cultured fibroblasts. Proc Natl Acad Sci USA. 1990;87(1):123–7.PubMedPubMedCentralCrossRef Rozengurt E, Higgins T, Chanter N, Lax AJ, Staddon JM. Pasteurella multocida toxin: potent mitogen for cultured fibroblasts. Proc Natl Acad Sci USA. 1990;87(1):123–7.PubMedPubMedCentralCrossRef
245.
go back to reference Preuss I, Hildebrand D, Orth JH, Aktories K, Kubatzky KF. Pasteurella multocida toxin is a potent activator of anti-apoptotic signalling pathways. Cell Microbiol. 2010;12(8):1174–85.PubMedCrossRef Preuss I, Hildebrand D, Orth JH, Aktories K, Kubatzky KF. Pasteurella multocida toxin is a potent activator of anti-apoptotic signalling pathways. Cell Microbiol. 2010;12(8):1174–85.PubMedCrossRef
246.
go back to reference Orth JH, Aktories K, Kubatzky KF. Modulation of host cell gene expression through activation of STAT transcription factors by Pasteurella multocida toxin. J Biol Chem. 2007;282(5):3050–7.PubMedCrossRef Orth JH, Aktories K, Kubatzky KF. Modulation of host cell gene expression through activation of STAT transcription factors by Pasteurella multocida toxin. J Biol Chem. 2007;282(5):3050–7.PubMedCrossRef
247.
go back to reference Carlini F, Maroccia Z, Fiorentini C, Travaglione S, Fabbri A. Effects of the Escherichia coli bacterial toxin cytotoxic necrotizing factor 1 on different human and animal cells: a systematic review. Int J Mol Sci. 2021;22(22):12610.PubMedPubMedCentralCrossRef Carlini F, Maroccia Z, Fiorentini C, Travaglione S, Fabbri A. Effects of the Escherichia coli bacterial toxin cytotoxic necrotizing factor 1 on different human and animal cells: a systematic review. Int J Mol Sci. 2021;22(22):12610.PubMedPubMedCentralCrossRef
248.
go back to reference Travaglione S, Fabbri A, Fiorentini C. The Rho-activating CNF1 toxin from pathogenic E. coli: a risk factor for human cancer development? Infect Agent Cancer. 2008;3:4.PubMedPubMedCentralCrossRef Travaglione S, Fabbri A, Fiorentini C. The Rho-activating CNF1 toxin from pathogenic E. coli: a risk factor for human cancer development? Infect Agent Cancer. 2008;3:4.PubMedPubMedCentralCrossRef
249.
go back to reference Zhang Z, Aung KM, Uhlin BE, Wai SN. Reversible senescence of human colon cancer cells after blockage of mitosis/cytokinesis caused by the CNF1 cyclomodulin from Escherichia coli. Sci Rep. 2018;8(1):17780.PubMedPubMedCentralCrossRef Zhang Z, Aung KM, Uhlin BE, Wai SN. Reversible senescence of human colon cancer cells after blockage of mitosis/cytokinesis caused by the CNF1 cyclomodulin from Escherichia coli. Sci Rep. 2018;8(1):17780.PubMedPubMedCentralCrossRef
250.
go back to reference Guo Y, Wang J, Zhou K, Lv J, Wang L, Gao S, Keller ET, Zhang ZS, Wang Q, Yao Z. Cytotoxic necrotizing factor 1 promotes bladder cancer angiogenesis through activating RhoC. Faseb J. 2020;34(6):7927–40.PubMedCrossRef Guo Y, Wang J, Zhou K, Lv J, Wang L, Gao S, Keller ET, Zhang ZS, Wang Q, Yao Z. Cytotoxic necrotizing factor 1 promotes bladder cancer angiogenesis through activating RhoC. Faseb J. 2020;34(6):7927–40.PubMedCrossRef
251.
go back to reference Guo Y, Zhang Z, Wei H, Wang J, Lv J, Zhang K, Keller ET, Yao Z, Wang Q. Cytotoxic necrotizing factor 1 promotes prostate cancer progression through activating the Cdc42-PAK1 axis. J Pathol. 2017;243(2):208–19.PubMedCrossRef Guo Y, Zhang Z, Wei H, Wang J, Lv J, Zhang K, Keller ET, Yao Z, Wang Q. Cytotoxic necrotizing factor 1 promotes prostate cancer progression through activating the Cdc42-PAK1 axis. J Pathol. 2017;243(2):208–19.PubMedCrossRef
252.
go back to reference Fabbri A, Travaglione S, Rosadi F, Ballan G, Maroccia Z, Giambenedetti M, Guidotti M, Ødum N, Krejsgaard T, Fiorentini C. The Escherichia coli protein toxin cytotoxic necrotizing factor 1 induces epithelial mesenchymal transition. Cell Microbiol. 2020;22(2): e13138.PubMedCrossRef Fabbri A, Travaglione S, Rosadi F, Ballan G, Maroccia Z, Giambenedetti M, Guidotti M, Ødum N, Krejsgaard T, Fiorentini C. The Escherichia coli protein toxin cytotoxic necrotizing factor 1 induces epithelial mesenchymal transition. Cell Microbiol. 2020;22(2): e13138.PubMedCrossRef
253.
go back to reference Cuevas-Ramos G, Petit CR, Marcq I, Boury M, Oswald E, Nougayrède JP. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc Natl Acad Sci USA. 2010;107(25):11537–42.PubMedPubMedCentralCrossRef Cuevas-Ramos G, Petit CR, Marcq I, Boury M, Oswald E, Nougayrède JP. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc Natl Acad Sci USA. 2010;107(25):11537–42.PubMedPubMedCentralCrossRef
254.
go back to reference Dziubańska-Kusibab PJ, Berger H, Battistini F, Bouwman BAM, Iftekhar A, Katainen R, Cajuso T, Crosetto N, Orozco M, Aaltonen LA, et al. Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat Med. 2020;26(7):1063–9.PubMedCrossRef Dziubańska-Kusibab PJ, Berger H, Battistini F, Bouwman BAM, Iftekhar A, Katainen R, Cajuso T, Crosetto N, Orozco M, Aaltonen LA, et al. Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat Med. 2020;26(7):1063–9.PubMedCrossRef
255.
go back to reference Lopès A, Billard E, Casse AH, Villéger R, Veziant J, Roche G, Carrier G, Sauvanet P, Briat A, Pagès F, et al. Colibactin-positive Escherichia coli induce a procarcinogenic immune environment leading to immunotherapy resistance in colorectal cancer. Int J Cancer. 2020;146(11):3147–59.PubMedCrossRef Lopès A, Billard E, Casse AH, Villéger R, Veziant J, Roche G, Carrier G, Sauvanet P, Briat A, Pagès F, et al. Colibactin-positive Escherichia coli induce a procarcinogenic immune environment leading to immunotherapy resistance in colorectal cancer. Int J Cancer. 2020;146(11):3147–59.PubMedCrossRef
256.
257.
go back to reference Toprak NU, Yagci A, Gulluoglu BM, Akin ML, Demirkalem P, Celenk T, Soyletir G. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer. Clin Microbiol Infect. 2006;12(8):782–6.PubMedCrossRef Toprak NU, Yagci A, Gulluoglu BM, Akin ML, Demirkalem P, Celenk T, Soyletir G. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer. Clin Microbiol Infect. 2006;12(8):782–6.PubMedCrossRef
258.
go back to reference Obiso RJ Jr, Azghani AO, Wilkins TD. The Bacteroides fragilis toxin fragilysin disrupts the paracellular barrier of epithelial cells. Infect Immun. 1997;65(4):1431–9.PubMedPubMedCentralCrossRef Obiso RJ Jr, Azghani AO, Wilkins TD. The Bacteroides fragilis toxin fragilysin disrupts the paracellular barrier of epithelial cells. Infect Immun. 1997;65(4):1431–9.PubMedPubMedCentralCrossRef
260.
go back to reference Wu S, Powell J, Mathioudakis N, Kane S, Fernandez E, Sears CL. Bacteroides fragilis enterotoxin induces intestinal epithelial cell secretion of interleukin-8 through mitogen-activated protein kinases and a tyrosine kinase-regulated nuclear factor-kappaB pathway. Infect Immun. 2004;72(10):5832–9.PubMedPubMedCentralCrossRef Wu S, Powell J, Mathioudakis N, Kane S, Fernandez E, Sears CL. Bacteroides fragilis enterotoxin induces intestinal epithelial cell secretion of interleukin-8 through mitogen-activated protein kinases and a tyrosine kinase-regulated nuclear factor-kappaB pathway. Infect Immun. 2004;72(10):5832–9.PubMedPubMedCentralCrossRef
261.
go back to reference Chung L, Thiele Orberg E, Geis AL, Chan JL, Fu K, DeStefano Shields CE, Dejea CM, Fathi P, Chen J, Finard BB, et al. Bacteroides fragilis toxin coordinates a pro-carcinogenic inflammatory cascade via targeting of colonic epithelial cells. Cell Host Microbe. 2018;23(2):203-14.e5.PubMedPubMedCentralCrossRef Chung L, Thiele Orberg E, Geis AL, Chan JL, Fu K, DeStefano Shields CE, Dejea CM, Fathi P, Chen J, Finard BB, et al. Bacteroides fragilis toxin coordinates a pro-carcinogenic inflammatory cascade via targeting of colonic epithelial cells. Cell Host Microbe. 2018;23(2):203-14.e5.PubMedPubMedCentralCrossRef
262.
go back to reference Li J, Zhang AH, Wu FF, Wang XJ. Alterations in the gut microbiota and their metabolites in colorectal cancer: recent progress and future prospects. Front Oncol. 2022;12:841552.PubMedPubMedCentralCrossRef Li J, Zhang AH, Wu FF, Wang XJ. Alterations in the gut microbiota and their metabolites in colorectal cancer: recent progress and future prospects. Front Oncol. 2022;12:841552.PubMedPubMedCentralCrossRef
263.
go back to reference Bultman SJ. The microbiome and its potential as a cancer preventive intervention. Semin Oncol. 2016;43(1):97–106.PubMedCrossRef Bultman SJ. The microbiome and its potential as a cancer preventive intervention. Semin Oncol. 2016;43(1):97–106.PubMedCrossRef
264.
go back to reference Cardona F, Andrés-Lacueva C, Tulipani S, Tinahones FJ, Queipo-Ortuño MI. Benefits of polyphenols on gut microbiota and implications in human health. J Nutr Biochem. 2013;24(8):1415–22.PubMedCrossRef Cardona F, Andrés-Lacueva C, Tulipani S, Tinahones FJ, Queipo-Ortuño MI. Benefits of polyphenols on gut microbiota and implications in human health. J Nutr Biochem. 2013;24(8):1415–22.PubMedCrossRef
265.
go back to reference Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, Koh AY. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia. 2017;19(10):848–55.PubMedPubMedCentralCrossRef Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, Koh AY. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia. 2017;19(10):848–55.PubMedPubMedCentralCrossRef
Metadata
Title
Emerging role of human microbiome in cancer development and response to therapy: special focus on intestinal microflora
Authors
Hourieh Sadrekarimi
Zhanna R. Gardanova
Morteza Bakhshesh
Farnoosh Ebrahimzadeh
Amirhossein Fakhre Yaseri
Lakshmi Thangavelu
Zahra Hasanpoor
Firoozeh Abolhasani Zadeh
Mohammad Saeed Kahrizi
Publication date
01-12-2022
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2022
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-022-03492-7

Other articles of this Issue 1/2022

Journal of Translational Medicine 1/2022 Go to the issue