Skip to main content
Top
Published in: Molecular Cancer 1/2019

Open Access 01-12-2019 | Hepatitis B | Review

The microbiota and microbiome in pancreatic cancer: more influential than expected

Authors: Miao-Yan Wei, Si Shi, Chen Liang, Qing-Cai Meng, Jie Hua, Yi-Yin Zhang, Jiang Liu, Bo Zhang, Jin Xu, Xian-Jun Yu

Published in: Molecular Cancer | Issue 1/2019

Login to get access

Abstract

Microbiota is just beginning to be recognized as an important player in carcinogenesis and the interplay among microbes is greater than expected. Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease for which mortality closely parallels incidence. Early detection would provide the best opportunity to increase survival rates. Specific well-studied oral, gastrointestinal, and intrapancreatic microbes and some kinds of hepatotropic viruses and bactibilia may have potential etiological roles in pancreatic carcinogenesis, or modulating individual responses to oncotherapy. Concrete mechanisms mainly involve perpetuating inflammation, regulating the immune system-microbe-tumor axis, affecting metabolism, and altering the tumor microenvironment. The revolutionary technology of omics has generated insight into cancer microbiomes. A better understanding of the microbiota in PDAC might lead to the establishment of screening or early-stage diagnosis methods, implementation of cancer bacteriotherapy, adjustment of therapeutic efficacy even alleviating the adverse effects, creating new opportunities and fostering hope for desperate PDAC patients.
Literature
1.
go back to reference Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–14.CrossRef Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–14.CrossRef
2.
go back to reference Rajpoot M, Sharma AK, Sharma A, Gupta GK. Understanding the microbiome: Emerging biomarkers for exploiting the microbiota for personalized medicine against cancer. Semin Cancer Biol. 2018;52:1–8.PubMedCrossRef Rajpoot M, Sharma AK, Sharma A, Gupta GK. Understanding the microbiome: Emerging biomarkers for exploiting the microbiota for personalized medicine against cancer. Semin Cancer Biol. 2018;52:1–8.PubMedCrossRef
3.
go back to reference Staley C, Weingarden AR, Khoruts A, Sadowsky MJ. Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl Microbiol Biotechnol. 2017;101:47–64.PubMedCrossRef Staley C, Weingarden AR, Khoruts A, Sadowsky MJ. Interaction of gut microbiota with bile acid metabolism and its influence on disease states. Appl Microbiol Biotechnol. 2017;101:47–64.PubMedCrossRef
4.
go back to reference Tilg H, Adolph TE. Beyond Digestion: The Pancreas Shapes Intestinal Microbiota and Immunity. Cell Metab. 2017;25:495–6.PubMedCrossRef Tilg H, Adolph TE. Beyond Digestion: The Pancreas Shapes Intestinal Microbiota and Immunity. Cell Metab. 2017;25:495–6.PubMedCrossRef
5.
go back to reference de Martel C, Ferlay J, Franceschi S, Vignat J, Bray F, Forman D, Plummer M. Global burden of cancers attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol. 2012;13:607–15.PubMedCrossRef de Martel C, Ferlay J, Franceschi S, Vignat J, Bray F, Forman D, Plummer M. Global burden of cancers attributable to infections in 2008: a review and synthetic analysis. Lancet Oncol. 2012;13:607–15.PubMedCrossRef
6.
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;15:1016–22.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;15:1016–22.PubMedPubMedCentralCrossRef
7.
go back to reference Housseau F, Sears CL. Enterotoxigenic Bacteroides fragilis (ETBF)-mediated colitis in Min (Apc+/−) mice: a human commensal-based murine model of colon carcinogenesis. Cell Cycle. 2010;9:3–5.PubMedCrossRef Housseau F, Sears CL. Enterotoxigenic Bacteroides fragilis (ETBF)-mediated colitis in Min (Apc+/−) mice: a human commensal-based murine model of colon carcinogenesis. Cell Cycle. 2010;9:3–5.PubMedCrossRef
10.
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;23:203–214 e205.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;23:203–214 e205.PubMedPubMedCentralCrossRef
11.
go back to reference Kwong TNY, Wang X, Nakatsu G, Chow TC, Tipoe T, Dai RZW, Tsoi KKK, Wong MCS, Tse G, Chan MTV, et al. Association Between Bacteremia From Specific Microbes and Subsequent Diagnosis of Colorectal Cancer. Gastroenterology. 2018;155:383–390 e388.PubMedCrossRef Kwong TNY, Wang X, Nakatsu G, Chow TC, Tipoe T, Dai RZW, Tsoi KKK, Wong MCS, Tse G, Chan MTV, et al. Association Between Bacteremia From Specific Microbes and Subsequent Diagnosis of Colorectal Cancer. Gastroenterology. 2018;155:383–390 e388.PubMedCrossRef
12.
go back to reference Li J, Sung CY, Lee N, Ni Y, Pihlajamaki J, Panagiotou G, El-Nezami H. Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice. Proc Natl Acad Sci U S A. 2016;113:E1306–15.PubMedPubMedCentralCrossRef Li J, Sung CY, Lee N, Ni Y, Pihlajamaki J, Panagiotou G, El-Nezami H. Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice. Proc Natl Acad Sci U S A. 2016;113:E1306–15.PubMedPubMedCentralCrossRef
13.
go back to reference Loo TM, Kamachi F, Watanabe Y, Yoshimoto S, Kanda H, Arai Y, Nakajima-Takagi Y, Iwama A, Koga T, Sugimoto Y, et al. Gut Microbiota Promotes Obesity-Associated Liver Cancer through PGE2-Mediated Suppression of Antitumor Immunity. Cancer Discov. 2017;7:522–38.PubMedCrossRef Loo TM, Kamachi F, Watanabe Y, Yoshimoto S, Kanda H, Arai Y, Nakajima-Takagi Y, Iwama A, Koga T, Sugimoto Y, et al. Gut Microbiota Promotes Obesity-Associated Liver Cancer through PGE2-Mediated Suppression of Antitumor Immunity. Cancer Discov. 2017;7:522–38.PubMedCrossRef
14.
15.
go back to reference Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, Agdashian D, Terabe M, Berzofsky JA, Fako V, et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 2018;360(6391):eaan5931.PubMedCrossRefPubMedCentral Ma C, Han M, Heinrich B, Fu Q, Zhang Q, Sandhu M, Agdashian D, Terabe M, Berzofsky JA, Fako V, et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science. 2018;360(6391):eaan5931.PubMedCrossRefPubMedCentral
16.
go back to reference Borrelli A, Bonelli P, Tuccillo FM, Goldfine ID, Evans JL, Buonaguro FM, Mancini A. Role of gut microbiota and oxidative stress in the progression of non-alcoholic fatty liver disease to hepatocarcinoma: Current and innovative therapeutic approaches. Redox Biol. 2018;15:467–79.PubMedPubMedCentralCrossRef Borrelli A, Bonelli P, Tuccillo FM, Goldfine ID, Evans JL, Buonaguro FM, Mancini A. Role of gut microbiota and oxidative stress in the progression of non-alcoholic fatty liver disease to hepatocarcinoma: Current and innovative therapeutic approaches. Redox Biol. 2018;15:467–79.PubMedPubMedCentralCrossRef
19.
go back to reference Ertz-Archambault N, Keim P, Von Hoff D. Microbiome and pancreatic cancer: A comprehensive topic review of literature. World J Gastroenterol. 2017;23:1899–908.PubMedPubMedCentralCrossRef Ertz-Archambault N, Keim P, Von Hoff D. Microbiome and pancreatic cancer: A comprehensive topic review of literature. World J Gastroenterol. 2017;23:1899–908.PubMedPubMedCentralCrossRef
20.
21.
22.
23.
go back to reference Jotwani R, Cutler CW. Adult periodontitis--specific bacterial infection or chronic inflammation? J Med Microbiol. 1998;47:187–8.PubMedCrossRef Jotwani R, Cutler CW. Adult periodontitis--specific bacterial infection or chronic inflammation? J Med Microbiol. 1998;47:187–8.PubMedCrossRef
24.
go back to reference Hujoel PP, Drangsholt M, Spiekerman C, Weiss NS. An exploration of the periodontitis-cancer association. Ann Epidemiol. 2003;13:312–6.PubMedCrossRef Hujoel PP, Drangsholt M, Spiekerman C, Weiss NS. An exploration of the periodontitis-cancer association. Ann Epidemiol. 2003;13:312–6.PubMedCrossRef
25.
go back to reference Michaud DS, Joshipura K, Giovannucci E, Fuchs CS. A prospective study of periodontal disease and pancreatic cancer in US male health professionals. J Natl Cancer Inst. 2007;99:171–5.PubMedCrossRef Michaud DS, Joshipura K, Giovannucci E, Fuchs CS. A prospective study of periodontal disease and pancreatic cancer in US male health professionals. J Natl Cancer Inst. 2007;99:171–5.PubMedCrossRef
26.
go back to reference Michaud DS, Liu Y, Meyer M, Giovannucci E, Joshipura K. Periodontal disease, tooth loss, and cancer risk in male health professionals: a prospective cohort study. Lancet Oncol. 2008;9:550–8.PubMedPubMedCentralCrossRef Michaud DS, Liu Y, Meyer M, Giovannucci E, Joshipura K. Periodontal disease, tooth loss, and cancer risk in male health professionals: a prospective cohort study. Lancet Oncol. 2008;9:550–8.PubMedPubMedCentralCrossRef
27.
go back to reference Ahn J, Segers S, Hayes RB. Periodontal disease, Porphyromonas gingivalis serum antibody levels and orodigestive cancer mortality. Carcinogenesis. 2012;33:1055–8.PubMedPubMedCentralCrossRef Ahn J, Segers S, Hayes RB. Periodontal disease, Porphyromonas gingivalis serum antibody levels and orodigestive cancer mortality. Carcinogenesis. 2012;33:1055–8.PubMedPubMedCentralCrossRef
28.
go back to reference Michaud DS, Izard J, Wilhelm-Benartzi CS, You DH, Grote VA, Tjonneland A, Dahm CC, Overvad K, Jenab M, Fedirko V, et al. Plasma antibodies to oral bacteria and risk of pancreatic cancer in a large European prospective cohort study. Gut. 2013;62:1764–70.PubMedCrossRef Michaud DS, Izard J, Wilhelm-Benartzi CS, You DH, Grote VA, Tjonneland A, Dahm CC, Overvad K, Jenab M, Fedirko V, et al. Plasma antibodies to oral bacteria and risk of pancreatic cancer in a large European prospective cohort study. Gut. 2013;62:1764–70.PubMedCrossRef
29.
go back to reference Bracci PM. Oral Health and the Oral Microbiome in Pancreatic Cancer: An Overview of Epidemiological Studies. Cancer J. 2017;23:310–4.PubMedCrossRef Bracci PM. Oral Health and the Oral Microbiome in Pancreatic Cancer: An Overview of Epidemiological Studies. Cancer J. 2017;23:310–4.PubMedCrossRef
30.
go back to reference Heikkila P, But A, Sorsa T, Haukka J. Periodontitis and cancer mortality: Register-based cohort study of 68,273 adults in 10-year follow-up. Int J Cancer. 2018;142:2244–53.PubMedCrossRef Heikkila P, But A, Sorsa T, Haukka J. Periodontitis and cancer mortality: Register-based cohort study of 68,273 adults in 10-year follow-up. Int J Cancer. 2018;142:2244–53.PubMedCrossRef
31.
go back to reference Tezal M, Sullivan MA, Hyland A, Marshall JR, Stoler D, Reid ME, Loree TR, Rigual NR, Merzianu M, Hauck L, et al. Chronic periodontitis and the incidence of head and neck squamous cell carcinoma. Cancer Epidemiol Biomark Prev. 2009;18:2406–12.CrossRef Tezal M, Sullivan MA, Hyland A, Marshall JR, Stoler D, Reid ME, Loree TR, Rigual NR, Merzianu M, Hauck L, et al. Chronic periodontitis and the incidence of head and neck squamous cell carcinoma. Cancer Epidemiol Biomark Prev. 2009;18:2406–12.CrossRef
32.
go back to reference Mai X, LaMonte MJ, Hovey KM, Nwizu N, Freudenheim JL, Tezal M, Scannapieco F, Hyland A, Andrews CA, Genco RJ, Wactawski-Wende J. History of periodontal disease diagnosis and lung cancer incidence in the Women's Health Initiative Observational Study. Cancer Causes Control. 2014;25:1045–53.PubMedPubMedCentralCrossRef Mai X, LaMonte MJ, Hovey KM, Nwizu N, Freudenheim JL, Tezal M, Scannapieco F, Hyland A, Andrews CA, Genco RJ, Wactawski-Wende J. History of periodontal disease diagnosis and lung cancer incidence in the Women's Health Initiative Observational Study. Cancer Causes Control. 2014;25:1045–53.PubMedPubMedCentralCrossRef
33.
go back to reference Urbaniak C, Cummins J, Brackstone M, Macklaim JM, Gloor GB, Baban CK, Scott L, O'Hanlon DM, Burton JP, Francis KP, et al. Microbiota of human breast tissue. Appl Environ Microbiol. 2014;80:3007–14.PubMedPubMedCentralCrossRef Urbaniak C, Cummins J, Brackstone M, Macklaim JM, Gloor GB, Baban CK, Scott L, O'Hanlon DM, Burton JP, Francis KP, et al. Microbiota of human breast tissue. Appl Environ Microbiol. 2014;80:3007–14.PubMedPubMedCentralCrossRef
34.
go back to reference Freudenheim JL, Genco RJ, LaMonte MJ, Millen AE, Hovey KM, Mai X, Nwizu N, Andrews CA, Wactawski-Wende J. Periodontal Disease and Breast Cancer: Prospective Cohort Study of Postmenopausal Women. Cancer Epidemiol Biomark Prev. 2016;25:43–50.CrossRef Freudenheim JL, Genco RJ, LaMonte MJ, Millen AE, Hovey KM, Mai X, Nwizu N, Andrews CA, Wactawski-Wende J. Periodontal Disease and Breast Cancer: Prospective Cohort Study of Postmenopausal Women. Cancer Epidemiol Biomark Prev. 2016;25:43–50.CrossRef
35.
go back to reference Nwizu NN, Marshall JR, Moysich K, Genco RJ, Hovey KM, Mai X, LaMonte MJ, Freudenheim JL, Wactawski-Wende J. Periodontal Disease and Incident Cancer Risk among Postmenopausal Women: Results from the Women's Health Initiative Observational Cohort. Cancer Epidemiol Biomark Prev. 2017;26:1255–65.CrossRef Nwizu NN, Marshall JR, Moysich K, Genco RJ, Hovey KM, Mai X, LaMonte MJ, Freudenheim JL, Wactawski-Wende J. Periodontal Disease and Incident Cancer Risk among Postmenopausal Women: Results from the Women's Health Initiative Observational Cohort. Cancer Epidemiol Biomark Prev. 2017;26:1255–65.CrossRef
36.
go back to reference Farrell JJ, Zhang L, Zhou H, Chia D, Elashoff D, Akin D, Paster BJ, Joshipura K, Wong DT. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut. 2012;61:582–8.PubMedCrossRef Farrell JJ, Zhang L, Zhou H, Chia D, Elashoff D, Akin D, Paster BJ, Joshipura K, Wong DT. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut. 2012;61:582–8.PubMedCrossRef
37.
go back to reference Meyer MS, Joshipura K, Giovannucci E, Michaud DS. A review of the relationship between tooth loss, periodontal disease, and cancer. Cancer Causes Control. 2008;19:895–907.PubMedPubMedCentralCrossRef Meyer MS, Joshipura K, Giovannucci E, Michaud DS. A review of the relationship between tooth loss, periodontal disease, and cancer. Cancer Causes Control. 2008;19:895–907.PubMedPubMedCentralCrossRef
38.
go back to reference Stolzenberg-Solomon RZ, Dodd KW, Blaser MJ, Virtamo J, Taylor PR, Albanes D. Tooth loss, pancreatic cancer, and Helicobacter pylori. Am J Clin Nutr. 2003;78:176–81.CrossRefPubMed Stolzenberg-Solomon RZ, Dodd KW, Blaser MJ, Virtamo J, Taylor PR, Albanes D. Tooth loss, pancreatic cancer, and Helicobacter pylori. Am J Clin Nutr. 2003;78:176–81.CrossRefPubMed
39.
go back to reference Hiraki A, Matsuo K, Suzuki T, Kawase T, Tajima K. Teeth loss and risk of cancer at 14 common sites in Japanese. Cancer Epidemiol Biomark Prev. 2008;17:1222–7.CrossRef Hiraki A, Matsuo K, Suzuki T, Kawase T, Tajima K. Teeth loss and risk of cancer at 14 common sites in Japanese. Cancer Epidemiol Biomark Prev. 2008;17:1222–7.CrossRef
40.
go back to reference Hayashi C, Gudino CV, Gibson FC 3rd, Genco CA. Review: Pathogen-induced inflammation at sites distant from oral infection: bacterial persistence and induction of cell-specific innate immune inflammatory pathways. Mol Oral Microbiol. 2010;25:305–16.PubMedPubMedCentralCrossRef Hayashi C, Gudino CV, Gibson FC 3rd, Genco CA. Review: Pathogen-induced inflammation at sites distant from oral infection: bacterial persistence and induction of cell-specific innate immune inflammatory pathways. Mol Oral Microbiol. 2010;25:305–16.PubMedPubMedCentralCrossRef
41.
go back to reference Brook I, Frazier EH. Microbiological analysis of pancreatic abscess. Clin Infect Dis. 1996;22:384–5.PubMedCrossRef Brook I, Frazier EH. Microbiological analysis of pancreatic abscess. Clin Infect Dis. 1996;22:384–5.PubMedCrossRef
42.
go back to reference Jacob JA. Study Links Periodontal Disease Bacteria to Pancreatic Cancer Risk. JAMA. 2016;315:2653–4.PubMedCrossRef Jacob JA. Study Links Periodontal Disease Bacteria to Pancreatic Cancer Risk. JAMA. 2016;315:2653–4.PubMedCrossRef
43.
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;67:120–7.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;67:120–7.PubMedCrossRef
44.
go back to reference Barton CM, Staddon SL, Hughes CM, Hall PA, O'Sullivan C, Kloppel G, Theis B, Russell RC, Neoptolemos J, Williamson RC, et al. Abnormalities of the p53 tumour suppressor gene in human pancreatic cancer. Br J Cancer. 1991;64:1076–82.PubMedPubMedCentralCrossRef Barton CM, Staddon SL, Hughes CM, Hall PA, O'Sullivan C, Kloppel G, Theis B, Russell RC, Neoptolemos J, Williamson RC, et al. Abnormalities of the p53 tumour suppressor gene in human pancreatic cancer. Br J Cancer. 1991;64:1076–82.PubMedPubMedCentralCrossRef
45.
go back to reference Liu L, Wang K, Zhu ZM, Shao JH. Associations between P53 Arg72Pro and development of digestive tract cancers: a meta-analysis. Arch Med Res. 2011;42:60–9.PubMedCrossRef Liu L, Wang K, Zhu ZM, Shao JH. Associations between P53 Arg72Pro and development of digestive tract cancers: a meta-analysis. Arch Med Res. 2011;42:60–9.PubMedCrossRef
46.
go back to reference Prassolov VS, Sakamoto H, Nishimura S, Terada M, Sugimura T. Activation of c-Ki-ras gene in human pancreatic cancer. Jpn J Cancer Res. 1985;76:792–5.PubMed Prassolov VS, Sakamoto H, Nishimura S, Terada M, Sugimura T. Activation of c-Ki-ras gene in human pancreatic cancer. Jpn J Cancer Res. 1985;76:792–5.PubMed
47.
go back to reference Watanabe H, Miyagi C, Yamaguchi Y, Satomura Y, Ohta H, Motoo Y, Okai T, Yoshimura T, Tsuji Y, Sawabu N. Detection of K-ras point mutations at codon 12 in pancreatic juice for the diagnosis of pancreatic cancer by hybridization protection assay: a simple method for the determination of the types of point mutation. Jpn J Cancer Res. 1996;87:466–74.PubMedPubMedCentralCrossRef Watanabe H, Miyagi C, Yamaguchi Y, Satomura Y, Ohta H, Motoo Y, Okai T, Yoshimura T, Tsuji Y, Sawabu N. Detection of K-ras point mutations at codon 12 in pancreatic juice for the diagnosis of pancreatic cancer by hybridization protection assay: a simple method for the determination of the types of point mutation. Jpn J Cancer Res. 1996;87:466–74.PubMedPubMedCentralCrossRef
48.
go back to reference Evans T, Faircloth M, Deery A, Thomas V, Turner A, Dalgleish A. Analysis of K-ras gene mutations in human pancreatic cancer cell lines and in bile samples from patients with pancreatic and biliary cancers. Oncol Rep. 1997;4:1373–81.PubMed Evans T, Faircloth M, Deery A, Thomas V, Turner A, Dalgleish A. Analysis of K-ras gene mutations in human pancreatic cancer cell lines and in bile samples from patients with pancreatic and biliary cancers. Oncol Rep. 1997;4:1373–81.PubMed
49.
go back to reference Sinn BV, Striefler JK, Rudl MA, Lehmann A, Bahra M, Denkert C, Sinn M, Stieler J, Klauschen F, Budczies J, et al. KRAS mutations in codon 12 or 13 are associated with worse prognosis in pancreatic ductal adenocarcinoma. Pancreas. 2014;43:578–83.PubMedCrossRef Sinn BV, Striefler JK, Rudl MA, Lehmann A, Bahra M, Denkert C, Sinn M, Stieler J, Klauschen F, Budczies J, et al. KRAS mutations in codon 12 or 13 are associated with worse prognosis in pancreatic ductal adenocarcinoma. Pancreas. 2014;43:578–83.PubMedCrossRef
51.
53.
go back to reference Tribble GD, Kerr JE, Wang BY. Genetic diversity in the oral pathogen Porphyromonas gingivalis: molecular mechanisms and biological consequences. Future Microbiol. 2013;8:607–20.PubMedCrossRef Tribble GD, Kerr JE, Wang BY. Genetic diversity in the oral pathogen Porphyromonas gingivalis: molecular mechanisms and biological consequences. Future Microbiol. 2013;8:607–20.PubMedCrossRef
54.
go back to reference Aliyu SH, Marriott RK, Curran MD, Parmar S, Bentley N, Brown NM, Brazier JS, Ludlam H. Real-time PCR investigation into the importance of Fusobacterium necrophorum as a cause of acute pharyngitis in general practice. J Med Microbiol. 2004;53:1029–35.PubMedCrossRef Aliyu SH, Marriott RK, Curran MD, Parmar S, Bentley N, Brown NM, Brazier JS, Ludlam H. Real-time PCR investigation into the importance of Fusobacterium necrophorum as a cause of acute pharyngitis in general practice. J Med Microbiol. 2004;53:1029–35.PubMedCrossRef
55.
go back to reference Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14:207–15.PubMedPubMedCentralCrossRef Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14:207–15.PubMedPubMedCentralCrossRef
56.
go back to reference Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, et al. Fusobacterium nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy. Cell. 2017;170:548–563 e516.PubMedPubMedCentralCrossRef Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, et al. Fusobacterium nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy. Cell. 2017;170:548–563 e516.PubMedPubMedCentralCrossRef
57.
go back to reference Maisonneuve P, Lowenfels AB. Risk factors for pancreatic cancer: a summary review of meta-analytical studies. Int J Epidemiol. 2015;44:186–98.PubMedCrossRef Maisonneuve P, Lowenfels AB. Risk factors for pancreatic cancer: a summary review of meta-analytical studies. Int J Epidemiol. 2015;44:186–98.PubMedCrossRef
58.
go back to reference Mishra AK, Dubey V, Ghosh AR. Obesity: An overview of possible role(s) of gut hormones, lipid sensing and gut microbiota. Metabolism. 2016;65:48–65.PubMedCrossRef Mishra AK, Dubey V, Ghosh AR. Obesity: An overview of possible role(s) of gut hormones, lipid sensing and gut microbiota. Metabolism. 2016;65:48–65.PubMedCrossRef
59.
go back to reference Fritz S, Hackert T, Hartwig W, Rossmanith F, Strobel O, Schneider L, Will-Schweiger K, Kommerell M, Buchler MW, Werner J. Bacterial translocation and infected pancreatic necrosis in acute necrotizing pancreatitis derives from small bowel rather than from colon. Am J Surg. 2010;200:111–7.PubMedCrossRef Fritz S, Hackert T, Hartwig W, Rossmanith F, Strobel O, Schneider L, Will-Schweiger K, Kommerell M, Buchler MW, Werner J. Bacterial translocation and infected pancreatic necrosis in acute necrotizing pancreatitis derives from small bowel rather than from colon. Am J Surg. 2010;200:111–7.PubMedCrossRef
60.
go back to reference Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, Mohan N, Aykut B, Usyk M, Torres LE, et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov. 2018;8:403–16.PubMedPubMedCentralCrossRef Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, Mohan N, Aykut B, Usyk M, Torres LE, et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov. 2018;8:403–16.PubMedPubMedCentralCrossRef
61.
go back to reference Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, Zhang L, Sharma U, Giri B, Garg B, et al. Gut Microbiota Promotes Tumor Growth in Mice by Modulating Immune Response. Gastroenterology. 2018;155:33–37 e36.PubMedCrossRef Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, Zhang L, Sharma U, Giri B, Garg B, et al. Gut Microbiota Promotes Tumor Growth in Mice by Modulating Immune Response. Gastroenterology. 2018;155:33–37 e36.PubMedCrossRef
62.
go back to reference Raderer M, Wrba F, Kornek G, Maca T, Koller DY, Weinlaender G, Hejna M, Scheithauer W. Association between Helicobacter pylori infection and pancreatic cancer. Oncology. 1998;55:16–9.PubMedCrossRef Raderer M, Wrba F, Kornek G, Maca T, Koller DY, Weinlaender G, Hejna M, Scheithauer W. Association between Helicobacter pylori infection and pancreatic cancer. Oncology. 1998;55:16–9.PubMedCrossRef
63.
go back to reference Stolzenberg-Solomon RZ, Blaser MJ, Limburg PJ, Perez-Perez G, Taylor PR, Virtamo J, Albanes D, Study A. Helicobacter pylori seropositivity as a risk factor for pancreatic cancer. J Natl Cancer Inst. 2001;93:937–41.PubMedCrossRef Stolzenberg-Solomon RZ, Blaser MJ, Limburg PJ, Perez-Perez G, Taylor PR, Virtamo J, Albanes D, Study A. Helicobacter pylori seropositivity as a risk factor for pancreatic cancer. J Natl Cancer Inst. 2001;93:937–41.PubMedCrossRef
64.
go back to reference Risch HA, Yu H, Lu L, Kidd MS. ABO blood group, Helicobacter pylori seropositivity, and risk of pancreatic cancer: a case-control study. J Natl Cancer Inst. 2010;102:502–5.PubMedPubMedCentralCrossRef Risch HA, Yu H, Lu L, Kidd MS. ABO blood group, Helicobacter pylori seropositivity, and risk of pancreatic cancer: a case-control study. J Natl Cancer Inst. 2010;102:502–5.PubMedPubMedCentralCrossRef
65.
go back to reference Lindkvist B, Johansen D, Borgstrom A, Manjer J. A prospective study of Helicobacter pylori in relation to the risk for pancreatic cancer. BMC Cancer. 2008;8:321.PubMedPubMedCentralCrossRef Lindkvist B, Johansen D, Borgstrom A, Manjer J. A prospective study of Helicobacter pylori in relation to the risk for pancreatic cancer. BMC Cancer. 2008;8:321.PubMedPubMedCentralCrossRef
66.
go back to reference Bao Y, Spiegelman D, Li R, Giovannucci E, Fuchs CS, Michaud DS. History of peptic ulcer disease and pancreatic cancer risk in men. Gastroenterology. 2010;138:541–9.PubMedCrossRef Bao Y, Spiegelman D, Li R, Giovannucci E, Fuchs CS, Michaud DS. History of peptic ulcer disease and pancreatic cancer risk in men. Gastroenterology. 2010;138:541–9.PubMedCrossRef
67.
go back to reference Trikudanathan G, Philip A, Dasanu CA, Baker WL. Association between Helicobacter pylori infection and pancreatic cancer. A cumulative meta-analysis. JOP. 2011;12:26–31.PubMed Trikudanathan G, Philip A, Dasanu CA, Baker WL. Association between Helicobacter pylori infection and pancreatic cancer. A cumulative meta-analysis. JOP. 2011;12:26–31.PubMed
68.
69.
go back to reference Schulte A, Pandeya N, Fawcett J, Fritschi L, Risch HA, Webb PM, Whiteman DC, Neale RE. Association between Helicobacter pylori and pancreatic cancer risk: a meta-analysis. Cancer Causes Control. 2015;26:1027–35.PubMedCrossRef Schulte A, Pandeya N, Fawcett J, Fritschi L, Risch HA, Webb PM, Whiteman DC, Neale RE. Association between Helicobacter pylori and pancreatic cancer risk: a meta-analysis. Cancer Causes Control. 2015;26:1027–35.PubMedCrossRef
70.
go back to reference de Martel C, Llosa AE, Friedman GD, Vogelman JH, Orentreich N, Stolzenberg-Solomon RZ, Parsonnet J. Helicobacter pylori infection and development of pancreatic cancer. Cancer Epidemiol Biomark Prev. 2008;17:1188–94.CrossRef de Martel C, Llosa AE, Friedman GD, Vogelman JH, Orentreich N, Stolzenberg-Solomon RZ, Parsonnet J. Helicobacter pylori infection and development of pancreatic cancer. Cancer Epidemiol Biomark Prev. 2008;17:1188–94.CrossRef
71.
go back to reference Gawin A, Wex T, Lawniczak M, Malfertheiner P, Starzynska T. Helicobacter pylori infection in pancreatic cancer. Pol Merkur Lekarski. 2012;32:103–7.PubMed Gawin A, Wex T, Lawniczak M, Malfertheiner P, Starzynska T. Helicobacter pylori infection in pancreatic cancer. Pol Merkur Lekarski. 2012;32:103–7.PubMed
72.
go back to reference Yu G, Murphy G, Michel A, Weinstein SJ, Mannisto S, Albanes D, Pawlita M, Stolzenberg-Solomon RZ. Seropositivity to Helicobacter pylori and risk of pancreatic cancer. Cancer Epidemiol Biomark Prev. 2013;22:2416–9.CrossRef Yu G, Murphy G, Michel A, Weinstein SJ, Mannisto S, Albanes D, Pawlita M, Stolzenberg-Solomon RZ. Seropositivity to Helicobacter pylori and risk of pancreatic cancer. Cancer Epidemiol Biomark Prev. 2013;22:2416–9.CrossRef
73.
go back to reference Chen XZ, Wang R, Chen HN, Hu JK. Cytotoxin-Associated Gene A-Negative Strains of Helicobacter pylori as a Potential Risk Factor of Pancreatic Cancer: A Meta-Analysis Based on Nested Case-Control Studies. Pancreas. 2015;44:1340–4.PubMedCrossRef Chen XZ, Wang R, Chen HN, Hu JK. Cytotoxin-Associated Gene A-Negative Strains of Helicobacter pylori as a Potential Risk Factor of Pancreatic Cancer: A Meta-Analysis Based on Nested Case-Control Studies. Pancreas. 2015;44:1340–4.PubMedCrossRef
74.
go back to reference Wang Y, Zhang FC, Wang YJ. Helicobacter pylori and pancreatic cancer risk: a meta- analysis based on 2,049 cases and 2,861 controls. Asian Pac J Cancer Prev. 2014;15:4449–54.PubMedCrossRef Wang Y, Zhang FC, Wang YJ. Helicobacter pylori and pancreatic cancer risk: a meta- analysis based on 2,049 cases and 2,861 controls. Asian Pac J Cancer Prev. 2014;15:4449–54.PubMedCrossRef
75.
go back to reference Risch HA, Lu L, Kidd MS, Wang J, Zhang W, Ni Q, Gao YT, Yu H. Helicobacter pylori seropositivities and risk of pancreatic carcinoma. Cancer Epidemiol Biomark Prev. 2014;23:172–8.CrossRef Risch HA, Lu L, Kidd MS, Wang J, Zhang W, Ni Q, Gao YT, Yu H. Helicobacter pylori seropositivities and risk of pancreatic carcinoma. Cancer Epidemiol Biomark Prev. 2014;23:172–8.CrossRef
76.
go back to reference Kalaf EA, Al-Khafaji ZM, Yassen NY, Al-Abbudi FA, Sadwen SN. Study of the cytoxin-associated gene a (CagA gene) in Helicobacter pylori using gastric biopsies of Iraqi patients. Saudi J Gastroenterol. 2013;19:69–74.PubMedPubMedCentralCrossRef Kalaf EA, Al-Khafaji ZM, Yassen NY, Al-Abbudi FA, Sadwen SN. Study of the cytoxin-associated gene a (CagA gene) in Helicobacter pylori using gastric biopsies of Iraqi patients. Saudi J Gastroenterol. 2013;19:69–74.PubMedPubMedCentralCrossRef
77.
go back to reference Chen S, Duan G, Zhang R, Fan Q. Helicobacter pylori cytotoxin-associated gene A protein upregulates alpha-enolase expression via Src/MEK/ERK pathway: implication for progression of gastric cancer. Int J Oncol. 2014;45:764–70.PubMedCrossRef Chen S, Duan G, Zhang R, Fan Q. Helicobacter pylori cytotoxin-associated gene A protein upregulates alpha-enolase expression via Src/MEK/ERK pathway: implication for progression of gastric cancer. Int J Oncol. 2014;45:764–70.PubMedCrossRef
78.
go back to reference Risch HA. Pancreatic cancer: Helicobacter pylori colonization, N-nitrosamine exposures, and ABO blood group. Mol Carcinog. 2012;51:109–18.PubMedCrossRef Risch HA. Pancreatic cancer: Helicobacter pylori colonization, N-nitrosamine exposures, and ABO blood group. Mol Carcinog. 2012;51:109–18.PubMedCrossRef
79.
go back to reference Jesnowski R, Isaksson B, Mohrcke C, Bertsch C, Bulajic M, Schneider-Brachert W, Kloppel G, Lowenfels AB, Maisonneuve P, Lohr JM. Helicobacter pylori in autoimmune pancreatitis and pancreatic carcinoma. Pancreatology. 2010;10:462–6.PubMedCrossRef Jesnowski R, Isaksson B, Mohrcke C, Bertsch C, Bulajic M, Schneider-Brachert W, Kloppel G, Lowenfels AB, Maisonneuve P, Lohr JM. Helicobacter pylori in autoimmune pancreatitis and pancreatic carcinoma. Pancreatology. 2010;10:462–6.PubMedCrossRef
80.
go back to reference Luo J, Nordenvall C, Nyren O, Adami HO, Permert J, Ye W. The risk of pancreatic cancer in patients with gastric or duodenal ulcer disease. Int J Cancer. 2007;120:368–72.PubMedCrossRef Luo J, Nordenvall C, Nyren O, Adami HO, Permert J, Ye W. The risk of pancreatic cancer in patients with gastric or duodenal ulcer disease. Int J Cancer. 2007;120:368–72.PubMedCrossRef
81.
go back to reference Shimoda T, Shikata T, Karasawa T, Tsukagoshi S, Yoshimura M, Sakurai I. Light microscopic localization of hepatitis B virus antigens in the human pancreas. Possibility of multiplication of hepatitis B virus in the human pancreas. Gastroenterology. 1981;81:998–1005.PubMed Shimoda T, Shikata T, Karasawa T, Tsukagoshi S, Yoshimura M, Sakurai I. Light microscopic localization of hepatitis B virus antigens in the human pancreas. Possibility of multiplication of hepatitis B virus in the human pancreas. Gastroenterology. 1981;81:998–1005.PubMed
82.
go back to reference Yoshimura M, Sakurai I, Shimoda T, Abe K, Okano T, Shikata T. Detection of HBsAg in the pancreas. Acta Pathol Jpn. 1981;31:711–7.PubMed Yoshimura M, Sakurai I, Shimoda T, Abe K, Okano T, Shikata T. Detection of HBsAg in the pancreas. Acta Pathol Jpn. 1981;31:711–7.PubMed
83.
go back to reference Dejean A, Lugassy C, Zafrani S, Tiollais P, Brechot C. Detection of hepatitis B virus DNA in pancreas, kidney and skin of two human carriers of the virus. J Gen Virol. 1984;65 ( Pt 3:651–5.PubMedCrossRef Dejean A, Lugassy C, Zafrani S, Tiollais P, Brechot C. Detection of hepatitis B virus DNA in pancreas, kidney and skin of two human carriers of the virus. J Gen Virol. 1984;65 ( Pt 3:651–5.PubMedCrossRef
84.
go back to reference Hohenberger P. The pancreas as target organ for hepatitis B virus--immunohistological detection of HBsAg in pancreatic carcinoma and chronic pancreatitis. Leber Magen Darm. 1985;15:58–63.PubMed Hohenberger P. The pancreas as target organ for hepatitis B virus--immunohistological detection of HBsAg in pancreatic carcinoma and chronic pancreatitis. Leber Magen Darm. 1985;15:58–63.PubMed
85.
go back to reference Jin Y, Gao H, Chen H, Wang J, Chen M, Li G, Wang L, Gu J, Tu H. Identification and impact of hepatitis B virus DNA and antigens in pancreatic cancer tissues and adjacent non-cancerous tissues. Cancer Lett. 2013;335:447–54.PubMedCrossRef Jin Y, Gao H, Chen H, Wang J, Chen M, Li G, Wang L, Gu J, Tu H. Identification and impact of hepatitis B virus DNA and antigens in pancreatic cancer tissues and adjacent non-cancerous tissues. Cancer Lett. 2013;335:447–54.PubMedCrossRef
86.
go back to reference Chen Y, Bai X, Zhang Q, Wen L, Su W, Fu Q, Sun X, Lou Y, Yang J, Zhang J, et al. The hepatitis B virus X protein promotes pancreatic cancer through modulation of the PI3K/AKT signaling pathway. Cancer Lett. 2016;380:98–105.PubMedCrossRef Chen Y, Bai X, Zhang Q, Wen L, Su W, Fu Q, Sun X, Lou Y, Yang J, Zhang J, et al. The hepatitis B virus X protein promotes pancreatic cancer through modulation of the PI3K/AKT signaling pathway. Cancer Lett. 2016;380:98–105.PubMedCrossRef
87.
go back to reference Fiorino S, Visani M, Acquaviva G, Fornelli A, Masetti M, Cuppini A, Bacchi-Reggiani ML, Jovine E, Tallini G, Pession A, de Biase D. Search for HBV and HCV Genome in Cancer Cells of Pancreatic Tumors. Pancreas. 2016;45:e12–4.PubMedCrossRef Fiorino S, Visani M, Acquaviva G, Fornelli A, Masetti M, Cuppini A, Bacchi-Reggiani ML, Jovine E, Tallini G, Pession A, de Biase D. Search for HBV and HCV Genome in Cancer Cells of Pancreatic Tumors. Pancreas. 2016;45:e12–4.PubMedCrossRef
88.
go back to reference Sundquist K, Sundquist J, Ji J. Risk of hepatocellular carcinoma and cancers at other sites among patients diagnosed with chronic hepatitis B virus infection in Sweden. J Med Virol. 2014;86:18–22.PubMedCrossRef Sundquist K, Sundquist J, Ji J. Risk of hepatocellular carcinoma and cancers at other sites among patients diagnosed with chronic hepatitis B virus infection in Sweden. J Med Virol. 2014;86:18–22.PubMedCrossRef
89.
go back to reference Kamiza AB, Su FH, Wang WC, Sung FC, Chang SN, Yeh CC. Chronic hepatitis infection is associated with extrahepatic cancer development: a nationwide population-based study in Taiwan. BMC Cancer. 2016;16:861.PubMedPubMedCentralCrossRef Kamiza AB, Su FH, Wang WC, Sung FC, Chang SN, Yeh CC. Chronic hepatitis infection is associated with extrahepatic cancer development: a nationwide population-based study in Taiwan. BMC Cancer. 2016;16:861.PubMedPubMedCentralCrossRef
90.
go back to reference Kwok RM, Tran TT. Hepatitis B and Risk of Non-Hepatocellular Carcinoma Malignancy. Clin Liver Dis. 2016;20:693–702.PubMedCrossRef Kwok RM, Tran TT. Hepatitis B and Risk of Non-Hepatocellular Carcinoma Malignancy. Clin Liver Dis. 2016;20:693–702.PubMedCrossRef
91.
go back to reference Wei XL, Luo HY, Li CF, Jin Y, Zeng ZL, Ju HQ, Wu QN, Wang Y, Mao MJ, Liu WL, et al. Hepatitis B virus infection is associated with younger median age at diagnosis and death in cancers. Int J Cancer. 2017;141:152–9.PubMedCrossRef Wei XL, Luo HY, Li CF, Jin Y, Zeng ZL, Ju HQ, Wu QN, Wang Y, Mao MJ, Liu WL, et al. Hepatitis B virus infection is associated with younger median age at diagnosis and death in cancers. Int J Cancer. 2017;141:152–9.PubMedCrossRef
92.
go back to reference Fiorino S. HBV and HCV infection and pancreatic ductal adenocarcinoma. Hepatobiliary Pancreat Dis Int. 2013;12:570–4.PubMedCrossRef Fiorino S. HBV and HCV infection and pancreatic ductal adenocarcinoma. Hepatobiliary Pancreat Dis Int. 2013;12:570–4.PubMedCrossRef
93.
go back to reference Taranto D, Carrato A, Romano M, Maio G, Izzo CM, Del Vecchio Blanco C. Mild pancreatic damage in acute viral hepatitis. Digestion. 1989;42:93–7.PubMedCrossRef Taranto D, Carrato A, Romano M, Maio G, Izzo CM, Del Vecchio Blanco C. Mild pancreatic damage in acute viral hepatitis. Digestion. 1989;42:93–7.PubMedCrossRef
94.
go back to reference Katakura Y, Yotsuyanagi H, Hashizume K, Okuse C, Okuse N, Nishikawa K, Suzuki M, Iino S, Itoh F. Pancreatic involvement in chronic viral hepatitis. World J Gastroenterol. 2005;11:3508–13.PubMedPubMedCentralCrossRef Katakura Y, Yotsuyanagi H, Hashizume K, Okuse C, Okuse N, Nishikawa K, Suzuki M, Iino S, Itoh F. Pancreatic involvement in chronic viral hepatitis. World J Gastroenterol. 2005;11:3508–13.PubMedPubMedCentralCrossRef
95.
go back to reference Hoefs JC, Renner IG, Askhcavai M, Redeker AG. Hepatitis B surface antigen in pancreatic and biliary secretions. Gastroenterology. 1980;79:191–4.PubMedCrossRef Hoefs JC, Renner IG, Askhcavai M, Redeker AG. Hepatitis B surface antigen in pancreatic and biliary secretions. Gastroenterology. 1980;79:191–4.PubMedCrossRef
96.
go back to reference Ben Q, Li Z, Liu C, Cai Q, Yuan Y, Wang K, Xiao L, Gao J, Zhang H. Hepatitis B virus status and risk of pancreatic ductal adenocarcinoma: a case-control study from China. Pancreas. 2012;41:435–40.PubMedCrossRef Ben Q, Li Z, Liu C, Cai Q, Yuan Y, Wang K, Xiao L, Gao J, Zhang H. Hepatitis B virus status and risk of pancreatic ductal adenocarcinoma: a case-control study from China. Pancreas. 2012;41:435–40.PubMedCrossRef
97.
go back to reference Hassan MM, Li D, El-Deeb AS, Wolff RA, Bondy ML, Davila M, Abbruzzese JL. Association between hepatitis B virus and pancreatic cancer. J Clin Oncol. 2008;26:4557–62.PubMedPubMedCentralCrossRef Hassan MM, Li D, El-Deeb AS, Wolff RA, Bondy ML, Davila M, Abbruzzese JL. Association between hepatitis B virus and pancreatic cancer. J Clin Oncol. 2008;26:4557–62.PubMedPubMedCentralCrossRef
98.
go back to reference Fiorino S, Chili E, Bacchi-Reggiani L, Masetti M, Deleonardi G, Grondona AG, Silvestri T, Magrini E, Zanini N, Cuppini A, et al. Association between hepatitis B or hepatitis C virus infection and risk of pancreatic adenocarcinoma development: a systematic review and meta-analysis. Pancreatology. 2013;13:147–60.PubMedCrossRef Fiorino S, Chili E, Bacchi-Reggiani L, Masetti M, Deleonardi G, Grondona AG, Silvestri T, Magrini E, Zanini N, Cuppini A, et al. Association between hepatitis B or hepatitis C virus infection and risk of pancreatic adenocarcinoma development: a systematic review and meta-analysis. Pancreatology. 2013;13:147–60.PubMedCrossRef
99.
go back to reference Li L, Wu B, Yang LB, Yin GC, Liu JY. Chronic hepatitis B virus infection and risk of pancreatic cancer: a meta-analysis. Asian Pac J Cancer Prev. 2013;14:275–9.PubMedCrossRef Li L, Wu B, Yang LB, Yin GC, Liu JY. Chronic hepatitis B virus infection and risk of pancreatic cancer: a meta-analysis. Asian Pac J Cancer Prev. 2013;14:275–9.PubMedCrossRef
100.
go back to reference Luo G, Hao NB, Hu CJ, Yong X, Lu MH, Cheng BJ, Zhang Y, Yang SM. HBV infection increases the risk of pancreatic cancer: a meta-analysis. Cancer Causes Control. 2013;24:529–37.PubMedCrossRef Luo G, Hao NB, Hu CJ, Yong X, Lu MH, Cheng BJ, Zhang Y, Yang SM. HBV infection increases the risk of pancreatic cancer: a meta-analysis. Cancer Causes Control. 2013;24:529–37.PubMedCrossRef
101.
go back to reference Wang Y, Yang S, Song F, Cao S, Yin X, Xie J, Tu X, Xu J, Xu X, Dong X, Lu Z. Hepatitis B virus status and the risk of pancreatic cancer: a meta-analysis. Eur J Cancer Prev. 2013;22:328–34.PubMedCrossRef Wang Y, Yang S, Song F, Cao S, Yin X, Xie J, Tu X, Xu J, Xu X, Dong X, Lu Z. Hepatitis B virus status and the risk of pancreatic cancer: a meta-analysis. Eur J Cancer Prev. 2013;22:328–34.PubMedCrossRef
102.
go back to reference Xing S, Li ZW, Tian YF, Zhang LM, Li MQ, Zhou P. Chronic hepatitis virus infection increases the risk of pancreatic cancer: a meta-analysis. Hepatobiliary Pancreat Dis Int. 2013;12:575–83.PubMedCrossRef Xing S, Li ZW, Tian YF, Zhang LM, Li MQ, Zhou P. Chronic hepatitis virus infection increases the risk of pancreatic cancer: a meta-analysis. Hepatobiliary Pancreat Dis Int. 2013;12:575–83.PubMedCrossRef
103.
go back to reference Xu JH, Fu JJ, Wang XL, Zhu JY, Ye XH, Chen SD. Hepatitis B or C viral infection and risk of pancreatic cancer: a meta-analysis of observational studies. World J Gastroenterol. 2013;19:4234–41.PubMedPubMedCentralCrossRef Xu JH, Fu JJ, Wang XL, Zhu JY, Ye XH, Chen SD. Hepatitis B or C viral infection and risk of pancreatic cancer: a meta-analysis of observational studies. World J Gastroenterol. 2013;19:4234–41.PubMedPubMedCentralCrossRef
104.
go back to reference Majumder S, Bockorny B, Baker WL, Dasanu CA. Association between HBsAg positivity and pancreatic cancer: a meta-analysis. J Gastrointest Cancer. 2014;45:347–52.PubMedCrossRef Majumder S, Bockorny B, Baker WL, Dasanu CA. Association between HBsAg positivity and pancreatic cancer: a meta-analysis. J Gastrointest Cancer. 2014;45:347–52.PubMedCrossRef
105.
go back to reference Iloeje UH, Yang HI, Jen CL, Su J, Wang LY, You SL, Lu SN, Chen CJ. Risk of pancreatic cancer in chronic hepatitis B virus infection: data from the REVEAL-HBV cohort study. Liver Int. 2010;30:423–9.PubMedCrossRef Iloeje UH, Yang HI, Jen CL, Su J, Wang LY, You SL, Lu SN, Chen CJ. Risk of pancreatic cancer in chronic hepatitis B virus infection: data from the REVEAL-HBV cohort study. Liver Int. 2010;30:423–9.PubMedCrossRef
106.
go back to reference Wei XL, Qiu MZ, Chen WW, Jin Y, Ren C, Wang F, Luo HY, Wang ZQ, Zhang DS, Wang FH, et al. The status of HBV infection influences metastatic pattern and survival in Chinese patients with pancreatic cancer. J Transl Med. 2013;11:249.PubMedPubMedCentralCrossRef Wei XL, Qiu MZ, Chen WW, Jin Y, Ren C, Wang F, Luo HY, Wang ZQ, Zhang DS, Wang FH, et al. The status of HBV infection influences metastatic pattern and survival in Chinese patients with pancreatic cancer. J Transl Med. 2013;11:249.PubMedPubMedCentralCrossRef
107.
go back to reference Wang DS, Chen DL, Ren C, Wang ZQ, Qiu MZ, Luo HY, Zhang DS, Wang FH, Li YH, Xu RH. ABO blood group, hepatitis B viral infection and risk of pancreatic cancer. Int J Cancer. 2012;131:461–8.PubMedCrossRef Wang DS, Chen DL, Ren C, Wang ZQ, Qiu MZ, Luo HY, Zhang DS, Wang FH, Li YH, Xu RH. ABO blood group, hepatitis B viral infection and risk of pancreatic cancer. Int J Cancer. 2012;131:461–8.PubMedCrossRef
108.
go back to reference Andersen ES, Omland LH, Jepsen P, Krarup H, Christensen PB, Obel N, Weis N, Study DC. Risk of all-type cancer, hepatocellular carcinoma, non-Hodgkin lymphoma and pancreatic cancer in patients infected with hepatitis B virus. J Viral Hepat. 2015;22:828–34.PubMedCrossRef Andersen ES, Omland LH, Jepsen P, Krarup H, Christensen PB, Obel N, Weis N, Study DC. Risk of all-type cancer, hepatocellular carcinoma, non-Hodgkin lymphoma and pancreatic cancer in patients infected with hepatitis B virus. J Viral Hepat. 2015;22:828–34.PubMedCrossRef
109.
go back to reference Fiorino S, Lorenzini S, Masetti M, Deleonardi G, Grondona AG, Silvestri T, Chili E, Del Prete P, Bacchi-Reggiani L, Cuppini A, Jovine E. Hepatitis B and C virus infections as possible risk factor for pancreatic adenocarcinoma. Med Hypotheses. 2012;79:678–97.PubMedCrossRef Fiorino S, Lorenzini S, Masetti M, Deleonardi G, Grondona AG, Silvestri T, Chili E, Del Prete P, Bacchi-Reggiani L, Cuppini A, Jovine E. Hepatitis B and C virus infections as possible risk factor for pancreatic adenocarcinoma. Med Hypotheses. 2012;79:678–97.PubMedCrossRef
110.
go back to reference Fiorino S, Bacchi-Reggiani L, Pontoriero L, Gallo C, Chili E, Masetti M, Zanini N, Grondona A, Silvestri T, Deleonardi G, et al. Tensegrity model hypothesis: may this paradigm be useful to explain hepatic and pancreatic carcinogenesis in patients with persistent hepatitis B or hepatitis C virus infection? JOP. 2014;15:151–64.PubMed Fiorino S, Bacchi-Reggiani L, Pontoriero L, Gallo C, Chili E, Masetti M, Zanini N, Grondona A, Silvestri T, Deleonardi G, et al. Tensegrity model hypothesis: may this paradigm be useful to explain hepatic and pancreatic carcinogenesis in patients with persistent hepatitis B or hepatitis C virus infection? JOP. 2014;15:151–64.PubMed
111.
go back to reference Sangiorgio L, Attardo T, Gangemi R, Rubino C, Barone M, Lunetta M. Increased frequency of HCV and HBV infection in type 2 diabetic patients. Diabetes Res Clin Pract. 2000;48:147–51.PubMedCrossRef Sangiorgio L, Attardo T, Gangemi R, Rubino C, Barone M, Lunetta M. Increased frequency of HCV and HBV infection in type 2 diabetic patients. Diabetes Res Clin Pract. 2000;48:147–51.PubMedCrossRef
112.
go back to reference Maekawa T, Fukaya R, Takamatsu S, Itoyama S, Fukuoka T, Yamada M, Hata T, Nagaoka S, Kawamoto K, Eguchi H, et al. Possible involvement of Enterococcus infection in the pathogenesis of chronic pancreatitis and cancer. Biochem Biophys Res Commun. 2018;506:962-969.PubMedCrossRef Maekawa T, Fukaya R, Takamatsu S, Itoyama S, Fukuoka T, Yamada M, Hata T, Nagaoka S, Kawamoto K, Eguchi H, et al. Possible involvement of Enterococcus infection in the pathogenesis of chronic pancreatitis and cancer. Biochem Biophys Res Commun. 2018;506:962-969.PubMedCrossRef
113.
go back to reference Serra N, Di Carlo P, Gulotta G, d' Arpa F, Giammanco A, Colomba C, Melfa G, Fasciana T, Sergi C. Bactibilia in women affected with diseases of the biliary tract and pancreas. A STROBE guidelines-adherent cross-sectional study in Southern Italy. J Med Microbiol. 2018;67:1090–5.PubMedCrossRef Serra N, Di Carlo P, Gulotta G, d' Arpa F, Giammanco A, Colomba C, Melfa G, Fasciana T, Sergi C. Bactibilia in women affected with diseases of the biliary tract and pancreas. A STROBE guidelines-adherent cross-sectional study in Southern Italy. J Med Microbiol. 2018;67:1090–5.PubMedCrossRef
114.
go back to reference Tomasiewicz K, Modrzewska R, Lyczak A, Krawczuk G. TT virus infection and pancreatic cancer: relationship or accidental coexistence. World J Gastroenterol. 2005;11:2847–9.PubMedPubMedCentralCrossRef Tomasiewicz K, Modrzewska R, Lyczak A, Krawczuk G. TT virus infection and pancreatic cancer: relationship or accidental coexistence. World J Gastroenterol. 2005;11:2847–9.PubMedPubMedCentralCrossRef
115.
116.
go back to reference Olson SH, Satagopan J, Xu Y, Ling L, Leong S, Orlow I, Saldia A, Li P, Nunes P, Madonia V, et al. The oral microbiota in patients with pancreatic cancer, patients with IPMNs, and controls: a pilot study. Cancer Causes Control. 2017;28:959–69.PubMedPubMedCentralCrossRef Olson SH, Satagopan J, Xu Y, Ling L, Leong S, Orlow I, Saldia A, Li P, Nunes P, Madonia V, et al. The oral microbiota in patients with pancreatic cancer, patients with IPMNs, and controls: a pilot study. Cancer Causes Control. 2017;28:959–69.PubMedPubMedCentralCrossRef
117.
go back to reference Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, Gavert N, Zwang Y, Cooper ZA, Shee K, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357:1156–60.PubMedPubMedCentralCrossRef Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, Gavert N, Zwang Y, Cooper ZA, Shee K, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357:1156–60.PubMedPubMedCentralCrossRef
118.
go back to reference Nilsson HO, Stenram U, Ihse I, Wadstrom T. Helicobacter species ribosomal DNA in the pancreas, stomach and duodenum of pancreatic cancer patients. World J Gastroenterol. 2006;12:3038–43.PubMedPubMedCentralCrossRef Nilsson HO, Stenram U, Ihse I, Wadstrom T. Helicobacter species ribosomal DNA in the pancreas, stomach and duodenum of pancreatic cancer patients. World J Gastroenterol. 2006;12:3038–43.PubMedPubMedCentralCrossRef
119.
go back to reference Takayama S, Takahashi H, Matsuo Y, Okada Y, Manabe T. Effects of Helicobacter pylori infection on human pancreatic cancer cell line. Hepatogastroenterology. 2007;54:2387–91.PubMed Takayama S, Takahashi H, Matsuo Y, Okada Y, Manabe T. Effects of Helicobacter pylori infection on human pancreatic cancer cell line. Hepatogastroenterology. 2007;54:2387–91.PubMed
120.
go back to reference Mitsuhashi K, Nosho K, Sukawa Y, Matsunaga Y, Ito M, Kurihara H, Kanno S, Igarashi H, Naito T, Adachi Y, et al. Association of Fusobacterium species in pancreatic cancer tissues with molecular features and prognosis. Oncotarget. 2015;6:7209–20.PubMedPubMedCentral Mitsuhashi K, Nosho K, Sukawa Y, Matsunaga Y, Ito M, Kurihara H, Kanno S, Igarashi H, Naito T, Adachi Y, et al. Association of Fusobacterium species in pancreatic cancer tissues with molecular features and prognosis. Oncotarget. 2015;6:7209–20.PubMedPubMedCentral
122.
go back to reference Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, Dimagno EP, Andren-Sandberg A, Domellof L. Pancreatitis and the risk of pancreatic cancer. International Pancreatitis Study Group. N Engl J Med. 1993;328:1433–7.PubMedCrossRef Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, Dimagno EP, Andren-Sandberg A, Domellof L. Pancreatitis and the risk of pancreatic cancer. International Pancreatitis Study Group. N Engl J Med. 1993;328:1433–7.PubMedCrossRef
124.
go back to reference Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44.PubMedCrossRef Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature. 2008;454:436–44.PubMedCrossRef
125.
go back to reference Morgillo F, Dallio M, Della Corte CM, Gravina AG, Viscardi G, Loguercio C, Ciardiello F, Federico A. Carcinogenesis as a Result of Multiple Inflammatory and Oxidative Hits: a Comprehensive Review from Tumor Microenvironment to Gut Microbiota. Neoplasia. 2018;20:721–33.PubMedPubMedCentralCrossRef Morgillo F, Dallio M, Della Corte CM, Gravina AG, Viscardi G, Loguercio C, Ciardiello F, Federico A. Carcinogenesis as a Result of Multiple Inflammatory and Oxidative Hits: a Comprehensive Review from Tumor Microenvironment to Gut Microbiota. Neoplasia. 2018;20:721–33.PubMedPubMedCentralCrossRef
126.
go back to reference Guerra C, Schuhmacher AJ, Canamero M, Grippo PJ, Verdaguer L, Perez-Gallego L, Dubus P, Sandgren EP, Barbacid M. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007;11:291–302.PubMedCrossRef Guerra C, Schuhmacher AJ, Canamero M, Grippo PJ, Verdaguer L, Perez-Gallego L, Dubus P, Sandgren EP, Barbacid M. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007;11:291–302.PubMedCrossRef
127.
go back to reference Sparmann A, Bar-Sagi D. Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell. 2004;6:447–58.PubMedCrossRef Sparmann A, Bar-Sagi D. Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis. Cancer Cell. 2004;6:447–58.PubMedCrossRef
128.
go back to reference Schulz MD, Atay C, Heringer J, Romrig FK, Schwitalla S, Aydin B, Ziegler PK, Varga J, Reindl W, Pommerenke C, et al. High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature. 2014;514:508–12.PubMedPubMedCentralCrossRef Schulz MD, Atay C, Heringer J, Romrig FK, Schwitalla S, Aydin B, Ziegler PK, Varga J, Reindl W, Pommerenke C, et al. High-fat-diet-mediated dysbiosis promotes intestinal carcinogenesis independently of obesity. Nature. 2014;514:508–12.PubMedPubMedCentralCrossRef
129.
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;342:967–70.PubMedCrossRefPubMedCentral 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;342:967–70.PubMedCrossRefPubMedCentral
130.
132.
go back to reference Sistigu A, Viaud S, Chaput N, Bracci L, Proietti E, Zitvogel L. Immunomodulatory effects of cyclophosphamide and implementations for vaccine design. Semin Immunopathol. 2011;33:369–83.PubMedCrossRef Sistigu A, Viaud S, Chaput N, Bracci L, Proietti E, Zitvogel L. Immunomodulatory effects of cyclophosphamide and implementations for vaccine design. Semin Immunopathol. 2011;33:369–83.PubMedCrossRef
133.
go back to reference Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere 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;342:971–6.PubMedPubMedCentralCrossRef Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere 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;342:971–6.PubMedPubMedCentralCrossRef
134.
go back to reference Vetizou M, Pitt JM, Daillere 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;350:1079–84.PubMedPubMedCentralCrossRef Vetizou M, Pitt JM, Daillere 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;350:1079–84.PubMedPubMedCentralCrossRef
135.
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;350:1084–9.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;350:1084–9.PubMedPubMedCentralCrossRef
136.
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;359:97–103.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;359:97–103.PubMedCrossRef
137.
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;359:104–8.PubMedCrossRefPubMedCentral 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;359:104–8.PubMedCrossRefPubMedCentral
138.
go back to reference Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere 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;359:91–7.PubMedCrossRef Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere 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;359:91–7.PubMedCrossRef
139.
140.
go back to reference Ochi A, Nguyen AH, Bedrosian AS, Mushlin HM, Zarbakhsh S, Barilla R, Zambirinis CP, Fallon NC, Rehman A, Pylayeva-Gupta Y, et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. J Exp Med. 2012;209:1671–87.PubMedPubMedCentralCrossRef Ochi A, Nguyen AH, Bedrosian AS, Mushlin HM, Zarbakhsh S, Barilla R, Zambirinis CP, Fallon NC, Rehman A, Pylayeva-Gupta Y, et al. MyD88 inhibition amplifies dendritic cell capacity to promote pancreatic carcinogenesis via Th2 cells. J Exp Med. 2012;209:1671–87.PubMedPubMedCentralCrossRef
141.
go back to reference Ochi A, Graffeo CS, Zambirinis CP, Rehman A, Hackman M, Fallon N, Barilla RM, Henning JR, Jamal M, Rao R, et al. Toll-like receptor 7 regulates pancreatic carcinogenesis in mice and humans. J Clin Invest. 2012;122:4118–29.PubMedPubMedCentralCrossRef Ochi A, Graffeo CS, Zambirinis CP, Rehman A, Hackman M, Fallon N, Barilla RM, Henning JR, Jamal M, Rao R, et al. Toll-like receptor 7 regulates pancreatic carcinogenesis in mice and humans. J Clin Invest. 2012;122:4118–29.PubMedPubMedCentralCrossRef
142.
go back to reference Ikebe M, Kitaura Y, Nakamura M, Tanaka H, Yamasaki A, Nagai S, Wada J, Yanai K, Koga K, Sato N, et al. Lipopolysaccharide (LPS) increases the invasive ability of pancreatic cancer cells through the TLR4/MyD88 signaling pathway. J Surg Oncol. 2009;100:725–31.PubMedCrossRef Ikebe M, Kitaura Y, Nakamura M, Tanaka H, Yamasaki A, Nagai S, Wada J, Yanai K, Koga K, Sato N, et al. Lipopolysaccharide (LPS) increases the invasive ability of pancreatic cancer cells through the TLR4/MyD88 signaling pathway. J Surg Oncol. 2009;100:725–31.PubMedCrossRef
143.
go back to reference Zhang JJ, Wu HS, Wang L, Tian Y, Zhang JH, Wu HL. Expression and significance of TLR4 and HIF-1alpha in pancreatic ductal adenocarcinoma. World J Gastroenterol. 2010;16:2881–8.PubMedPubMedCentralCrossRef Zhang JJ, Wu HS, Wang L, Tian Y, Zhang JH, Wu HL. Expression and significance of TLR4 and HIF-1alpha in pancreatic ductal adenocarcinoma. World J Gastroenterol. 2010;16:2881–8.PubMedPubMedCentralCrossRef
144.
go back to reference Ding SP, Li JC, Jin C. A mouse model of severe acute pancreatitis induced with caerulein and lipopolysaccharide. World J Gastroenterol. 2003;9:584–9.PubMedPubMedCentralCrossRef Ding SP, Li JC, Jin C. A mouse model of severe acute pancreatitis induced with caerulein and lipopolysaccharide. World J Gastroenterol. 2003;9:584–9.PubMedPubMedCentralCrossRef
145.
go back to reference Sharif R, Dawra R, Wasiluk K, Phillips P, Dudeja V, Kurt-Jones E, Finberg R, Saluja A. Impact of toll-like receptor 4 on the severity of acute pancreatitis and pancreatitis-associated lung injury in mice. Gut. 2009;58:813–9.PubMedCrossRef Sharif R, Dawra R, Wasiluk K, Phillips P, Dudeja V, Kurt-Jones E, Finberg R, Saluja A. Impact of toll-like receptor 4 on the severity of acute pancreatitis and pancreatitis-associated lung injury in mice. Gut. 2009;58:813–9.PubMedCrossRef
146.
go back to reference Daniluk J, Liu Y, Deng D, Chu J, Huang H, Gaiser S, Cruz-Monserrate Z, Wang H, Ji B, Logsdon CD. An NF-kappaB pathway-mediated positive feedback loop amplifies Ras activity to pathological levels in mice. J Clin Invest. 2012;122:1519–28.PubMedPubMedCentralCrossRef Daniluk J, Liu Y, Deng D, Chu J, Huang H, Gaiser S, Cruz-Monserrate Z, Wang H, Ji B, Logsdon CD. An NF-kappaB pathway-mediated positive feedback loop amplifies Ras activity to pathological levels in mice. J Clin Invest. 2012;122:1519–28.PubMedPubMedCentralCrossRef
147.
go back to reference Riquelme E, Maitra A, McAllister F. Immunotherapy for Pancreatic Cancer: More Than Just a Gut Feeling. Cancer Discov. 2018;8:386–8.PubMedCrossRef Riquelme E, Maitra A, McAllister F. Immunotherapy for Pancreatic Cancer: More Than Just a Gut Feeling. Cancer Discov. 2018;8:386–8.PubMedCrossRef
148.
go back to reference De Monte L, Reni M, Tassi E, Clavenna D, Papa I, Recalde H, Braga M, Di Carlo V, Doglioni C, Protti MP. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208:469–78.PubMedPubMedCentralCrossRef De Monte L, Reni M, Tassi E, Clavenna D, Papa I, Recalde H, Braga M, Di Carlo V, Doglioni C, Protti MP. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208:469–78.PubMedPubMedCentralCrossRef
149.
go back to reference Benakanakere MR, Li Q, Eskan MA, Singh AV, Zhao J, Galicia JC, Stathopoulou P, Knudsen TB, Kinane DF. Modulation of TLR2 protein expression by miR-105 in human oral keratinocytes. J Biol Chem. 2009;284:23107–15.PubMedPubMedCentralCrossRef Benakanakere MR, Li Q, Eskan MA, Singh AV, Zhao J, Galicia JC, Stathopoulou P, Knudsen TB, Kinane DF. Modulation of TLR2 protein expression by miR-105 in human oral keratinocytes. J Biol Chem. 2009;284:23107–15.PubMedPubMedCentralCrossRef
150.
go back to reference Moffatt CE, Lamont RJ. Porphyromonas gingivalis induction of microRNA-203 expression controls suppressor of cytokine signaling 3 in gingival epithelial cells. Infect Immun. 2011;79:2632–7.PubMedPubMedCentralCrossRef Moffatt CE, Lamont RJ. Porphyromonas gingivalis induction of microRNA-203 expression controls suppressor of cytokine signaling 3 in gingival epithelial cells. Infect Immun. 2011;79:2632–7.PubMedPubMedCentralCrossRef
151.
go back to reference Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7.PubMedCrossRef Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, Pettersson S. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7.PubMedCrossRef
152.
go back to reference Donohoe DR, Collins LB, Wali A, Bigler R, Sun W, Bultman SJ. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol Cell. 2012;48:612–26.PubMedPubMedCentralCrossRef Donohoe DR, Collins LB, Wali A, Bigler R, Sun W, Bultman SJ. The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation. Mol Cell. 2012;48:612–26.PubMedPubMedCentralCrossRef
153.
go back to reference Donohoe DR, Holley D, Collins LB, Montgomery SA, Whitmore AC, Hillhouse A, Curry KP, Renner SW, Greenwalt A, Ryan EP, et al. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov. 2014;4:1387–97.PubMedPubMedCentralCrossRef Donohoe DR, Holley D, Collins LB, Montgomery SA, Whitmore AC, Hillhouse A, Curry KP, Renner SW, Greenwalt A, Ryan EP, et al. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov. 2014;4:1387–97.PubMedPubMedCentralCrossRef
154.
go back to reference Clarke JM, Topping DL, Bird AR, Young GP, Cobiac L. Effects of high-amylose maize starch and butyrylated high-amylose maize starch on azoxymethane-induced intestinal cancer in rats. Carcinogenesis. 2008;29:2190–4.PubMedPubMedCentralCrossRef Clarke JM, Topping DL, Bird AR, Young GP, Cobiac L. Effects of high-amylose maize starch and butyrylated high-amylose maize starch on azoxymethane-induced intestinal cancer in rats. Carcinogenesis. 2008;29:2190–4.PubMedPubMedCentralCrossRef
155.
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;52:729–34.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;52:729–34.PubMedPubMedCentralCrossRef
156.
go back to reference Miko E, Vida A, Kovacs T, Ujlaki G, Trencsenyi G, Marton J, Sari Z, Kovacs P, Boratko A, Hujber Z, et al. Lithocholic acid, a bacterial metabolite reduces breast cancer cell proliferation and aggressiveness. Biochim Biophys Acta Bioenerg. 2018;1859:958–74.PubMedCrossRef Miko E, Vida A, Kovacs T, Ujlaki G, Trencsenyi G, Marton J, Sari Z, Kovacs P, Boratko A, Hujber Z, et al. Lithocholic acid, a bacterial metabolite reduces breast cancer cell proliferation and aggressiveness. Biochim Biophys Acta Bioenerg. 2018;1859:958–74.PubMedCrossRef
157.
go back to reference Rogers CJ, Prabhu KS, Vijay-Kumar M. The microbiome and obesity-an established risk for certain types of cancer. Cancer J. 2014;20:176–80.PubMedCrossRef Rogers CJ, Prabhu KS, Vijay-Kumar M. The microbiome and obesity-an established risk for certain types of cancer. Cancer J. 2014;20:176–80.PubMedCrossRef
158.
go back to reference Djuric Z. Obesity-associated cancer risk: the role of intestinal microbiota in the etiology of the host proinflammatory state. Transl Res. 2017;179:155–67.PubMedCrossRef Djuric Z. Obesity-associated cancer risk: the role of intestinal microbiota in the etiology of the host proinflammatory state. Transl Res. 2017;179:155–67.PubMedCrossRef
159.
go back to reference Parekh PJ, Balart LA, Johnson DA. The Influence of the Gut Microbiome on Obesity, Metabolic Syndrome and Gastrointestinal Disease. Clin Transl Gastroenterol. 2015;6:e91.PubMedPubMedCentralCrossRef Parekh PJ, Balart LA, Johnson DA. The Influence of the Gut Microbiome on Obesity, Metabolic Syndrome and Gastrointestinal Disease. Clin Transl Gastroenterol. 2015;6:e91.PubMedPubMedCentralCrossRef
160.
go back to reference Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, Iwakura Y, Oshima K, Morita H, Hattori M, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 2013;499:97–101.PubMedCrossRef Yoshimoto S, Loo TM, Atarashi K, Kanda H, Sato S, Oyadomari S, Iwakura Y, Oshima K, Morita H, Hattori M, et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature. 2013;499:97–101.PubMedCrossRef
161.
go back to reference Eibl G, Rozengurt E. KRAS, YAP, and obesity in pancreatic cancer: A signaling network with multiple loops. Semin Cancer Biol. 2019;54:50-62.PubMedCrossRef Eibl G, Rozengurt E. KRAS, YAP, and obesity in pancreatic cancer: A signaling network with multiple loops. Semin Cancer Biol. 2019;54:50-62.PubMedCrossRef
162.
go back to reference Thomas H. Pancreatic cancer: Intra-tumour bacteria promote gemcitabine resistance in pancreatic adenocarcinoma. Nat Rev Gastroenterol Hepatol. 2017;14:632.PubMedCrossRef Thomas H. Pancreatic cancer: Intra-tumour bacteria promote gemcitabine resistance in pancreatic adenocarcinoma. Nat Rev Gastroenterol Hepatol. 2017;14:632.PubMedCrossRef
163.
go back to reference Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015;25:198–213.PubMedCrossRef Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015;25:198–213.PubMedCrossRef
165.
go back to reference Apte MV, Wilson JS, Lugea A, Pandol SJ. A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology. 2013;144:1210–9.PubMedCrossRef Apte MV, Wilson JS, Lugea A, Pandol SJ. A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology. 2013;144:1210–9.PubMedCrossRef
166.
go back to reference Tang D, Wang D, Yuan Z, Xue X, Zhang Y, An Y, Chen J, Tu M, Lu Z, Wei J, et al. Persistent activation of pancreatic stellate cells creates a microenvironment favorable for the malignant behavior of pancreatic ductal adenocarcinoma. Int J Cancer. 2013;132:993–1003.PubMedCrossRef Tang D, Wang D, Yuan Z, Xue X, Zhang Y, An Y, Chen J, Tu M, Lu Z, Wei J, et al. Persistent activation of pancreatic stellate cells creates a microenvironment favorable for the malignant behavior of pancreatic ductal adenocarcinoma. Int J Cancer. 2013;132:993–1003.PubMedCrossRef
167.
go back to reference Barbour SE, Nakashima K, Zhang JB, Tangada S, Hahn CL, Schenkein HA, Tew JG. Tobacco and smoking: environmental factors that modify the host response (immune system) and have an impact on periodontal health. Crit Rev Oral Biol Med. 1997;8:437–60.PubMedCrossRef Barbour SE, Nakashima K, Zhang JB, Tangada S, Hahn CL, Schenkein HA, Tew JG. Tobacco and smoking: environmental factors that modify the host response (immune system) and have an impact on periodontal health. Crit Rev Oral Biol Med. 1997;8:437–60.PubMedCrossRef
Metadata
Title
The microbiota and microbiome in pancreatic cancer: more influential than expected
Authors
Miao-Yan Wei
Si Shi
Chen Liang
Qing-Cai Meng
Jie Hua
Yi-Yin Zhang
Jiang Liu
Bo Zhang
Jin Xu
Xian-Jun Yu
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2019
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-019-1008-0

Other articles of this Issue 1/2019

Molecular Cancer 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine