Elsevier

Surgical Oncology

Volume 26, Issue 4, December 2017, Pages 368-376
Surgical Oncology

The role of intestinal bacteria in the development and progression of gastrointestinal tract neoplasms

https://doi.org/10.1016/j.suronc.2017.07.011Get rights and content

Highlights

  • Microorganisms can influence host immunity and human diseases, including cancer.

  • Microbial dysbiosis may influence gastrointestinal tract tumor progression.

  • Gut microbiota may influence efficacies of cancer chemotherapy and immunotherapy.

  • Gut microbiota may influence complications after surgery for gastrointestinal tract cancers.

Abstract

More than 100 trillion microorganisms inhabit the human intestinal tract and play important roles in health conditions and diseases, including cancer. Accumulating evidence demonstrates that specific bacteria and bacterial dysbiosis in the gastrointestinal tract can potentiate the development and progression of gastrointestinal tract neoplasms by damaging DNA, activating oncogenic signaling pathways, producing tumor-promoting metabolites such as secondary bile acids, and suppressing antitumor immunity. Other bacterial species have been shown to produce short-chain fatty acids such as butyrate, which can suppress inflammation and carcinogenesis in the gastrointestinal tract. Consistent with these lines of evidence, clinical studies using metagenomic analyses have shown associations of specific bacteria and bacterial dysbiosis with gastrointestinal tract cancers, including esophageal, gastric, and colorectal cancers. Emerging data demonstrate that intestinal bacteria can modulate the efficacy of cancer chemotherapies and novel targeted immunotherapies such as anti-CTLA4 and anti-CD274 therapies, the process of absorption, and the occurrence of complications after gastrointestinal surgery. A better understanding of the mechanisms by which the gut microbiota influence tumor development and progression in the intestine would provide opportunities to develop new prevention and treatment strategies for patients with gastrointestinal tract cancers by targeting the intestinal microflora.

Introduction

Cancer in the gastrointestinal tract is a leading cause of death worldwide [1]. Accumulating evidence indicates that gastrointestinal tract cancers develop through the accumulation of genetic and epigenetic alterations, which are influenced by host immunity, diet, and environmental and microbial exposures [2].

The human intestinal microbiome encompasses at least 100 trillion microorganisms, which can influence the immune system and health conditions, including cancer [3], [4], [5]. A growing body of evidence indicates that diet, lifestyle, and drugs can influence the composition of the gut microbiota and that the gut microbiota can modulate the development and progression of gastrointestinal tract neoplasms [6], [7]. Recent data have shown that some bacterial species produce tumor-promoting metabolites such as secondary bile acids, which potentiate the development and progression of gastrointestinal tract neoplasms, whereas other species produce short-chain fatty acids (SCFAs) such as butyrate, which can suppress inflammation and carcinogenesis in the gastrointestinal tract [8], [9].

Here, we review clinical studies on intestinal bacteria in relation to gastrointestinal tract cancers, including esophageal, gastric, and colorectal cancers. In addition, we describe emerging evidence for roles of intestinal bacteria in the efficacy of cancer chemotherapies and novel targeted immunotherapies such as anti-CTLA4 and anti-CD274 therapies, the process of absorption, and the occurrence of complications after gastrointestinal surgery.

Section snippets

Bacterial genotoxins

Intestinal bacteria have been shown to potentiate carcinogenesis through specific toxins that induce DNA damage. Colibactin is encoded by the polyketide synthase island, which is expressed by Escherichia coli from phylogroup B2, and has been shown to induce DNA damage, affect genomic instability [10], [11], and promote colon carcinogenesis in Il10−/− mice [12], [13].

Enterococcus faecalis has been shown to produce extracellular superoxide that induces DNA damage and genomic instability in

Esophageal cancer

Esophageal carcinoma consists of two main histological types: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). ESCC constitutes the large majority of esophageal cancer cases worldwide and more than 90% of the cases in high-risk areas, such as China, Iran, and Japan [1]. EAC is one of the most rapidly increasing cancers in the United States [84]. Clinical studies have suggested associations of specific bacteria and bacterial dysbiosis (the condition of having

Gut microbiota, chemotherapy, and immunotherapy

Emerging data demonstrate that intestinal bacteria can modulate the efficacy of cancer chemotherapies. Colorectal cancers with unresectable distant metastases are treated with chemotherapy regimens that are based on oxaliplatin and irinotecan combined with molecularly targeted therapies such as a humanized monoclonal antibody against VEGFA (bevacizumab) and anti-EGFR antibodies (cetuximab or panitumumab) [134]. ROS are important for DNA damage and apoptosis in response to oxaliplatin [135]. The

Gut microbiota and surgery for gastrointestinal tract cancers

Surgery for gastrointestinal tract cancers and intestinal reconstructions has been shown to influence the composition of the gut microbiota. Patients after Roux-en-Y gastric bypass (RYGB) are at increased risk of malabsorption, trace element deficiency, and dumping syndrome [154]. In a mouse model of RYGB, the amount of Bacteroidetes, Verrucomicrobia, and Proteobacteria in stool increased, and transfer of the gut microbiota from RYGB-treated mice to non-operated, germ-free mice resulted in

Future directions

Accumulating evidence indicates that intestinal bacteria can influence the tumor development and progression in the gastrointestinal tract. Considering that diet, lifestyle, pharmacological factors (including antibiotics), and probiotics and prebiotics can influence the composition of the intestinal microbiota, future investigations may be warranted to examine the potential influences of these modifiable factors on the intestinal microflora and tumorigenic processes.

The main cause of

Funding

This work was supported by grants from the U.S. National Institutes of Health (NIH) [R35 CA197735 to S.O.] and the Nodal Award (to S.O.) of the Dana-Farber/Harvard Cancer Center. K.M. is supported by grants from the Takeda Science Foundation, Kanae Foundation for the Promotion of Medical Science, YOKOYAMA Foundation for Clinical Pharmacology, and JSPS KAKENHI Grant Number 17H05094. The content is solely the responsibility of the authors and does not necessarily represent the official views of

Author contributions

Kosuke Mima and Shuji Ogino contributed equally as co-first authors. All authors contributed to review and revision. Kosuke Mima, Shuji Ogino, Yoichi Yamashita, Naoya Yoshida, Akira Chikamoto, Takatoshi Ishiko, and Hideo Baba developed the main concept and designed the study. Kosuke Mima, Shuji Ogino, and Hideo Baba wrote grant applications. Kosuke Mima and Shuji Ogino drafted the manuscript. Shigeki Nakagawa, Hiroshi Sawayama, Koichi Kinoshita, Ryuichi Krashima, Takatsugu Ishimoto, Katsunori

Potential competing interests

All authors declare that they have no competing financial interests.

Use of standardized official symbols

We use HUGO (Human Genome Organisation)-approved official symbols for genes and gene products, including AKT, BRAF, CCL20, CD274, CDH1, CTLA4, EGFR, IL6, IL10, IL17, IL18, IL22, IL23, MAPK, MMP9, NFKB, PDCD1, PIK3CA, PTGS2, STAT3, TLR2, TLR4, TP53, VEGFA, and WNT; all are described at www.genenames.org.

References (167)

  • M. Quante et al.

    Bile acid and inflammation activate gastric cardia stem cells in a mouse model of Barrett-like metaplasia

    Cancer Cell

    (2012)
  • N. Singh et al.

    Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis

    Immunity

    (2014)
  • D.R. Donohoe et al.

    The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation

    Mol. Cell

    (2012)
  • A. Belcheva et al.

    Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells

    Cell

    (2014)
  • H. Tye et al.

    STAT3-driven upregulation of TLR2 promotes gastric tumorigenesis independent of tumor inflammation

    Cancer Cell

    (2012)
  • J. Dupaul-Chicoine et al.

    The Nlrp3 inflammasome suppresses colorectal cancer metastatic growth in the liver by promoting natural killer cell tumoricidal activity

    Immunity

    (2015)
  • A.D. Kostic et al.

    Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment

    Cell Host Microbe

    (2013)
  • M. Amieva et al.

    Pathobiology of Helicobacter pylori-Induced gastric cancer

    Gastroenterology

    (2016)
  • R.L. Siegel et al.

    Cancer statistics, 2017

    CA Cancer J. Clin.

    (2017)
  • M. Song et al.

    Nutrients, foods, and colorectal cancer prevention

    Gastroenterology

    (2015)
  • W.S. Garrett

    Cancer and the microbiota

    Science

    (2015)
  • M.G. Rooks et al.

    Gut microbiota, metabolites and host immunity

    Nat. Rev. Immunol.

    (2016)
  • J. Wang et al.

    Metagenome-wide association studies: fine-mining the microbiome

    Nat. Rev. Microbiol.

    (2016)
  • E.D. Sonnenburg et al.

    Diet-induced extinctions in the gut microbiota compound over generations

    Nature

    (2016)
  • A. Zhernakova et al.

    Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity

    Science

    (2016)
  • P. Louis et al.

    The gut microbiota, bacterial metabolites and colorectal cancer

    Nat. Rev. Microbiol.

    (2014)
  • S.J. O'Keefe

    Diet, microorganisms and their metabolites, and colon cancer

    Nat. Rev. Gastroenterol. Hepatol.

    (2016)
  • J.P. Nougayrede et al.

    Escherichia coli induces DNA double-strand breaks in eukaryotic cells

    Science

    (2006)
  • G. Cuevas-Ramos et al.

    Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2010)
  • J.C. Arthur et al.

    Intestinal inflammation targets cancer-inducing activity of the microbiota

    Science

    (2012)
  • J.C. Arthur et al.

    Microbial genomic analysis reveals the essential role of inflammation in bacteria-induced colorectal cancer

    Nat. Commun.

    (2014)
  • M.M. Huycke et al.

    Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA

    Carcinogenesis

    (2002)
  • X. Wang et al.

    Enterococcus faecalis induces aneuploidy and tetraploidy in colonic epithelial cells through a bystander effect

    Cancer Res.

    (2008)
  • X. Wang et al.

    4-hydroxy-2-nonenal mediates genotoxicity and bystander effects caused by Enterococcus faecalis-infected macrophages

    Gastroenterology

    (2012)
  • X. Wang et al.

    Cyclooxygenase-2 generates the endogenous mutagen trans-4-hydroxy-2-nonenal in Enterococcus faecalis-infected macrophages

    Cancer Prev. Res. (Phila)

    (2013)
  • J.A. Strickertsson et al.

    Enterococcus faecalis infection causes inflammation, intracellular oxphos-independent ROS production, and DNA damage in human gastric cancer cells

    PLoS One

    (2013)
  • J.A. Strickertsson et al.
    (2014)
  • D. Nesic et al.

    Assembly and function of a bacterial genotoxin

    Nature

    (2004)
  • Z. Ge et al.

    Bacterial cytolethal distending toxin promotes the development of dysplasia in a model of microbially induced hepatocarcinogenesis

    Cell Microbiol.

    (2007)
  • R. Guidi et al.

    Chronic exposure to the cytolethal distending toxins of Gram-negative bacteria promotes genomic instability and altered DNA damage response

    Cell Microbiol.

    (2013)
  • V. Graillot et al.

    Genotoxicity of cytolethal distending toxin (CDT) on isogenic human colorectal cell lines: potential promoting effects for colorectal carcinogenesis

    Front. Cell Infect. Microbiol.

    (2016)
  • N. Murata-Kamiya et al.

    Helicobacter pylori CagA interacts with E-cadherin and deregulates the beta-catenin signal that promotes intestinal transdifferentiation in gastric epithelial cells

    Oncogene

    (2007)
  • S. Brandt et al.

    NF-kappaB activation and potentiation of proinflammatory responses by the Helicobacter pylori CagA protein

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • J. Wei et al.

    Bacterial CagA protein induces degradation of p53 protein in a p14ARF-dependent manner

    Gut

    (2015)
  • E. Bessede et al.

    Helicobacter pylori generates cells with cancer stem cell properties via epithelial-mesenchymal transition-like changes

    Oncogene

    (2014)
  • J. Kountouras et al.

    Helicobacter pylori might contribute to cancer and/or bone marrow-derived stem cell-related gastrointestinal oncogenesis

    Oncogene

    (2015)
  • E. Bessede et al.

    Helicobacter pylori infection and stem cells at the origin of gastric cancer

    Oncogene

    (2015)
  • S. Bartfeld et al.

    In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection

    Gastroenterology

    (2015)
  • K.W. McCracken et al.

    Modelling human development and disease in pluripotent stem-cell-derived gastric organoids

    Nature

    (2014)
  • R. Cristescu et al.

    Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes

    Nat. Med.

    (2015)
  • Cited by (58)

    • Effects of melittin on laying performance and intestinal barrier function of quails

      2023, Poultry Science
      Citation Excerpt :

      The intestinal microflora forms a protective layer by combining with the intestinal epithelial surface of intestinal cells (Yegani and Korver, 2008), which protects the host from pathogens. Altering the composition of intestinal flora has been shown to cause intestinal inflammation (Khalif et al., 2005; Deriu et al., 2016; Mima et al., 2017). In the present study, 0.12 g/kg melittin had the best effect on intestinal health.

    • Role and potential clinical utility of ARID1A in gastrointestinal malignancy

      2021, Mutation Research - Reviews in Mutation Research
      Citation Excerpt :

      The human microbiome is an ecosystem containing many microbiota interacting continuously with their microenvironment, particularly with the immune system [89–92]. Aberrant changes in this microecosystem can induce chronic inflammation [93–100] that plays exogenous and endogenous roles in the pathogenesis of tumors [101–104]. Some intestinal bacteria can activate oncogenic signaling pathways [105], while others cause microsatellite instability (MSI), CpG island methylation phenotype (CIMP), BRAF, and PIK3CA mutations.

    • Review of short-chain fatty acids effects on the immune system and cancer

      2020, Food Bioscience
      Citation Excerpt :

      The microbiome of the human gastrointestinal (GI) tract (T) consists of >~100 million microorganisms known as gut microflora/microbiota. The ecosystem of this bacterial, fungal, and viral population can produce a large number of bio-functional compounds like lactic acid, short-chain fatty acids (SCFA), linolic acid, vitamins, and some peptides influencing their ecosystem and the host (Mima et al., 2017; Qamar et al., 2017; Sivaprakasam et al., 2017; Tlaskalova-Hogenova et al., 2011). Among these products, SCFA produced by anaerobic fermentation of indigestible fiber (as prebiotics) by the gut bacterium and have various roles in cancer prevalence and influences the immune system (Dahl et al., 2017; Esgalhado et al., 2017).

    View all citing articles on Scopus
    1

    K.M. and S.O. contributed equally.

    View full text