Skip to main content
Clinics in Colon and Rectal Surgery logoLink to Clinics in Colon and Rectal Surgery
. 2023 Jan 25;36(2):105–111. doi: 10.1055/s-0042-1760679

Role of Bacteria in the Development of Colorectal Cancer

Ryan M Thomas 1,
PMCID: PMC9946721  PMID: 36844716

Abstract

Colorectal cancer (CRC) is the second leading cause of cancer-related death in the United States. Once limited to older populations, the incidence of CRC in patients under the age of 50 years is increasing and the etiology for this is uncertain. One hypothesis lies on the impact of the intestinal microbiome. The intestinal microbiome, composed primarily of bacteria but also viruses, fungi, and archaea, has been shown to regulate CRC development and progression both in vitro and in vivo. In this review, the role and intersection of the bacterial microbiome in various stages of clinical CRC development and management are discussed beginning with CRC screening. Various mechanisms whereby the microbiome has been shown to modulate CRC development including the influence of diet on the microbiome, bacterial-induced injury to the colonic epithelium, bacterial-produced toxins, and alteration of normal cancer immunosurveillance by the microbiome are discussed. Finally, the influence of microbiome on the response of CRC to treatment is discussed while highlighting ongoing clinical trials. The complexities of the microbiome and its role in CRC development and progression have become apparent and will require ongoing commitment to translate laboratory findings into meaningful clinical results that will aid more than 150,000 patients that develop CRC every year.

Keywords: microbiome, oncobiome, colorectal cancer, colon cancer, chemotherapy, immunotherapy


The importance of the microbiome, the collection of microbes (bacteria, viruses, fungi, and archaea) that have a symbiotic, commensal, and pathobiont relationship with its host, in cancer development has been clarified and expounded upon in recent years. Notably, the influence of the microbiome on cancer development, progression, and treatment response has been documented for a variety of cancers. However, the role of microbiome in colorectal cancer (CRC), the second leading cause of cancer-related death in the United States, remains the seminal body of work on this topic. The reason is likely because of the logical reasoning that local microbes could have an impact on CRC and the ease from which samples can be collected to test this hypothesis.

While only Helicobacter pylori (gastric cancer, gastric lymphoma), Human papillomavirus (cervical, oropharyngeal, and anogenital cancers), Hepatitis B and C viruses (hepatocellular cancer), Epstein-Barr virus (nasopharyngeal cancer and Burkitt lymphoma), Human herpesvirus type 8 (Kaposi sarcoma), Schistosoma haematobium (bladder carcinoma), Human T-cell lymphotropic virus (adult T-cell leukemia and lymphoma), Opisthorchis viverrine (cholangiocarcinoma), and Clonorchis sinensis (cholangiocarcinoma) have been recognized by the World Health Organization as directly contributing to cancer development, 1 microbes have been implicated in the development of CRC through direct cellular injury, production of toxic substances, and alterations of the host immune response as well. Determining what mechanism(s) is the primary driver, as opposed to a single causative microbe, is likely to be more impactful as it is likely that the status of the bacterial community structure will be the instigator of CRC development rather than a single microorganism.

In this review, the preceding topics are explored and presented with a potential clinical focus and applicability in mind. As the majority of studies involve the bacterial microbiome, this will be the focus of discussion, recognizing that the influence of the virome, fungome, and other lesser microorganisms remains relatively unstudied but may yield just as important information as the microbiome. Given that the incidence of CRC has been increasing and that the median age at diagnosis has been decreasing, affecting younger patients, this is an important and germane topic for clinicians to be aware of. 2 3

Microbiome and Colorectal Cancer Screening

Current recommendations for CRC screening take into account risk factors for CRC development based on age, family history, prior polyps, and inflammatory bowel disease (IBD), among others. 3 For low-risk individuals, the use of tumor DNA detection techniques has provided a patient-friendly method of screening and early detection but does rely on the presence of malignancy. However, the false positive rate of these tests is upward of 76%, necessitating a diagnostic colonoscopy. 4 Determining associations of microbiome community structure changes or the presence of specific microbes may provide another screening methodology to detect high-risk individuals who have yet to develop CRC. Colorectal carcinoma is accepted to develop in a stepwise fashion beginning with hyperplastic polyps that accumulate genetic aberrations in a progressive fashion to cancer. 5 Prior research has demonstrated that as these polyps progress into carcinoma, the microbial community structure changes and provides a potential opportunity to detect by non-invasive means premalignant lesions before the onset of frank carcinoma. 6 7 For example, species richness was reduced in stool samples from patients with conventional adenomatous polyps compared with controls without any polyps. However, there was no difference in diversity or overall composition in stool from patients with sessile serrated adenoma or hyperplastic polyps did not differ from control, albeit the authors admitted they had a small sample size. 8 Additionally, Rezasoltani and colleagues reported that specific bacterial species ( Fusobacterium nucleatum, Enterococcus faecalis, Streptococcus bovis, Enterotoxigenic Bacteroides fragilis [ETFB], and Porphyromonas spp ) were more prevalent in adenomatous polyp cases compared with sessile serrated polyp, hyperplastic polyp, and normal control cases. 9 Fusobacterium spp. has also been shown to be more prevalent in adenomas compared with adjacent normal tissue control. 10 The theoretical ability to detect cancer at its earliest stages has also been confirmed recently in pancreatic cancer. 11 Several clinical trials seek to exploit these noted changes to identify the process of colorectal carcinogenesis earlier ( Table 1 ). How polyps develop and accumulate their mutational burden is not well-delineated, but these studies provide some initial insight. The microbiota community structure is dictated by many influences, diet being one of them. Dietary differences between individuals and geographical locations may therefore explain differences in CRC incidence through alterations of the microbiome. 12

Table 1. Selected trials evaluating the role of the microbiome in colorectal cancer screening.

Study name Clinical trials id Study type Microbiome source
Pilot study of the fecal microbiome in the Shanghai population NCT01778595 Observational Feces
Analysis of intestinal microflora combined with DNA methylation in stool to detect colorectal cancer (IMMDC) NCT04302363 Observational Feces
Gut microbiota-based tool for the detection of colorectal cancer in positive patients for the fecal occult blood test NCT04662853 Observational Feces
Microbiome test for the detection of colorectal polyps and cancer NCT02141945 Observational Feces

Dietary Influence on the Microbiome and CRC Development

Much work has focused on the role of diet in CRC development. 13 14 15 Dietary intake secondarily impact the intestinal microbiome and likely is an upstream mediator of CRC development, acting through modulation of gut microbiota. Additionally, dietary differences associate with microbiome changes, giving credence to the theory that dietary causation is likely upstream to the actual inciting carcinogenic event of the microbiome. For example, microbes can metabolize dietary sulfur into hydrogen sulfide (H 2 S), a known carcinogen 16 and capable of disrupting the protective colonic mucosal layer. 17 Nguyen and colleagues reported on the effect of a high microbial sulfur diet, that is predicted to enrich for sulfur-metabolizing bacteria, on colonic polyp formation. The high microbial sulfur diet was defined by intake of processed meats, liquor, and low-calorie drinks. They demonstrated that a high sulfur microbial diet was associated with an increased risk to develop tubulovillous/villous and tubular adenomas (age-adjusted odds ratio [OR] of 1.61 [ p  = 0.04] and 1.27 [ p  = 0.05], respectively) before the age of 50. 18 Additionally, this high sulfur diet was associated with increased risk of conventional adenoma formation in the proximal colon (OR 1.6, p  = 0.009) but not distal colon or rectum. A follow-up study demonstrated that a high microbial sulfur diet resulted in an increased risk of CRC (age-adjusted OR 1.21, p  = 0.008) which was driven by an increase in distal colon and rectal cancers (age-adjusted OR 1.53, p  < 0.0001). Interestingly, Bilophila wadsworthia and Erysipelotrichaceae bacterium 21_3 , both sulfur-metabolizing bacteria and shown to be associated with CRC, were enriched in higher sulfur microbial diets. 15 19 20 21

An inverse relationship has been shown between dietary fiber intake and the incidence of CRC and colon adenoma formation. 22 23 A notable increase in the incidence of CRC in patients under the age of 50 is suspected to be partly related to reduced fiber intake. This appears secondary to the effects that fiber has on the gut microbiota. 24 Bacteria ferment dietary fiber into the short-chain fatty acids (SCFA): butyrate, propionate, and acetate. These SCFAs have critical roles in colonic mucosal health, immune function, and serve as a colonocyte energy source. 25 26 27 Notably, butyrate has been demonstrated to have potent anti-cancer properties through inhibition of proliferation and induction of apoptosis in vitro. 28 29 Similar to butyrate, the SCFAs acetate and propionate have similar, although to a lesser extent, anti-inflammatory properties and the ability to inhibit histone deacetylase which results in inhibition of cancer cell proliferation and apoptosis induction. 30 Interestingly, compared with non-cancer controls, patients with CRC had a decrease in bacteria capable of fermenting fiber into butyrate, notably Clostridium , Roseburia , and Eubacterium species ( p <0.001), and a concomitant decrease in SCFA levels. 31 Recently, the butyrate-producing bacteria, Clostridium butyricum , was demonstrated to inhibit colonic tumor formation in a spontaneous model of CRC, the APC min/+ mouse model. 32 This appears likely through inhibition of the oncogenic Wnt/β-catenin pathway and alteration of the microbiome to increase the richness of SCFA-producing bacteria. Given the above findings, non-pharmaceutical manipulation of the microbiome through dietary modulation or probiotic/bacterial supplementation is an appealing intervention to reduce CRC incidence that could have broad population health implications. Ongoing clinical trials seek to modify the diet for the sake of microbiome manipulation to reduce CRC risk ( Table 2 ).

Table 2. Selected trials studying the effects of diet on the microbiome and colorectal cancer.

Study name Clinical trials ID Study type
Meat-based versus Pesco-vegetarian diet and colorectal cancer (MeaTIc) NCT03416777 Interventional
Randomized
Exploring the effects of a high chlorophyll dietary intervention to reduce colon cancer risk in adults (M3G) NCT03582306 Interventional
Randomized
Diet modulation of bacterial sulfur and bile acid metabolism and colon cancer risk NCT03550885 Interventional
Randomized
Fiber to reduce colon cancer in Alaska native people NCT03028831 Interventional
Randomized

Microbiome-Mediated Injury to the Intestinal Mucosal Barrier

The colonic epithelium and associated mucosal lining are in constant contact with ingested toxins and aim to help establish a symbiont and pathobiont relationship with the host and its microbiota. The colon mucus layer is actually composed of two layers, with the inner layer being devoid of bacteria and in direct contact with the colonic epithelium. 33 34 Disruption of this barrier can result in repeated epithelial injury that over time can lead to genetic aberrations that are likely to progress to CRC. The role of the microbiota in this process has gained much attention recently as changes in the microbiome of patients with IBD have been shown to induce inflammation. 35 Interestingly, germ-free mice (mice born and raised without exposure to microbes) do not develop IBD 36 and transplantation of human stool from healthy volunteers into rats with dextran sodium sulfate-induced colitis reduced the inflammatory state of the colon and restored the natural diversity of the intestinal microbiota. 37 Building off the importance of SCFAs, butyrate has been shown to activate the mucin-encoding gene, MUC-2, through regulation of prostaglandin synthesis. This increased mucin production provides a protective barrier to the colonic epithelium and alterations in the intestinal microbiota result in reduced mucosal protection and allowance of bacteria and antigens to cross the epithelial barrier. 38 Dietary emulsifiers, present in processed foods, have been shown to increase the ability of Escherichia coli to translocate across an in vitro model of the membranous barrier of Peyer's patches (known as M cells). 39 Additionally, the dietary emulsifiers, carboxymethylcellulose, and polysorbate-80, induced colitis in mice predisposed to this condition. 40 Even in wild-type mice, these emulsifiers were able to increase gut permeability as confirmed with increased serum antibody levels of lipopolysaccharide (LPS) and flagellin, both components of gram negative bacteria. Finally, the bile acid deoxycholic acid (DCA) is increased in patients consuming a high fat diet, a recognized risk factor for CRC development. Not only is DCA capable of disrupting the integrity of the colonic mucosal and accelerating carcinogenesis in the APC min/+ mouse model of colon cancer, 41 but elevated levels of DCA are associated with microbiota changes found in CRC. 42 Taken together, the intestinal microbiome interacts directly with the protective mucosal layer of the colonic epithelium. Disruptions to either homeostasis of the microbiome or integrity of the mucosa can lead to increased inflammation and colon cancer formation, demonstrating the fine balance between these host factors.

Interplay of the Microbiome on Immunosurveillance of Colorectal Cancer

An important role of the host immune system is malignant cell surveillance but a hallmark of cancer is the ability of neoplastic cells to avoid immunologic detection and destruction. 43 To balance these antagonistic processes, the intestinal microbiome has been shown to have a multifaceted impact on the immune system. The intestinal wall is rife with immune cells, particularly in Peyer's patches located within the lamina propria. In this location, both innate and adaptive immune cells reside and participate in immunosurveillance by secretion of antimicrobial peptides and pattern recognition receptors (such as Toll-like receptor and Nod-like receptor) that recognize pathogen-associated molecular patterns and damage-associated molecular patterns, such as LPS and flagellin mentioned previously. 44 45 46 The response of the local colonic immune system to resident bacteria is dependent on multiple factors that can alter microbiome-specific T-cell responses, including specific bacteria. Yu and colleagues demonstrated that two mouse colonies with distinct microbiome communities developed colon cancer at different rates using the azoxy-methane-dextran sodium sulfate (AOM-DSS) model of colon cancer secondary to alterations in the intratumoral immune environment. 47 In these distinct tumors, it was noted the intratumoral T cells in mice with increased tumors had increased exhaustion. Furthermore, these mice had increased CD8 + IFNγ + T cells in the lamina propria prior to tumor formation but reduced CD8 + IFNγ + T cells in tumors and adjacent tissues compared with mice with fewer tumors. In a hallmark study, Ivanov and colleagues demonstrated that segmented filamentous bacteria (SFB) were able to induce Th17 cells (CD4 + T-cells characterized by IL-17 and IL-22 production) in the lamina propria of the small intestine. 48 Colonization with SFB was associated with increased expression of inflammatory and anti-microbial defensive genes. This study demonstrated the ability of bacteria to regulate mucosal immunity and inflammation. Recently, Helicobacter hepaticus has been recognized as an immunogenic intestinal bacterium capable of mitigating CRC formation in a mouse model. 49 Mice colonized with this bacterium had reduced colon cancer formation in the AOM-DSS model of colon cancer. The mechanism for this reduction appears related to the ability of H. hepaticus to induce T follicular helper cell infiltration into colon tumors and expansion of local lymphoid structures adjacent to colon tumors to facilitate clearance. The intestinal microbiome therefore has the capability to alter the immune surveillance of cells in the lamina propria as well as modulate intratumoral immune cell infiltration and activity. Such information will likely prove instrumental in identifying tumors that will be responsive to immunotherapy.

Toxigenic Bacteria in Colorectal Cancer Development

Given the abundance of bacteria in intestinal lumen and their direct contact to the colonic epithelium, the ability of microbes to cause direct injury to epithelium is a natural theoretical extension and one that has been well studied. Three specific bacteria have been identified that have unique mechanisms of direct cellular injury. First, E. coli is prevalent but of low abundance in the gastrointestinal tract. While a variety of strains exist that cause a human disease, E. coli that harbor the polyketide synthase ( pks ) gene island encodes 19 genes responsible for production of colibactin, a potent genotoxin. Colibactin is comprised of dual cyclopropane motifs which induce double-strand DNA breaks and cross links through alkylating adenine residues. 50 This induction of DNA damage results in colibactin-mediated mutagenesis. E. coli harboring the pks genotoxin island have accelerated colon carcinogenesis in a mouse model of CRC. 51 52 Recently, exposure of colonic organoids with pks + E. coli has been shown to result in a distinct mutational signature that mirrors the mutational signature from patient CRC specimens. 53 Of note, the adenomatosis polyposis coli ( APC ) gene, the most commonly mutated gene in CRC, harbored the most mutations that matched this signature (>5%).

Second, ETBF has been shown to function to promote colon carcinogenesis through its B. fragilis toxin (BFT). 54 While there are both non-toxigenic and toxigenic strains of B. fragilis , the pathogenicity of ETBF is based on its BFT which is a matrix metalloprotease consisting of three isoforms. Notably, isoforms BFT-1 and BFT-2 have been detected in CRC tumor specimens and impact colon carcinogenesis by three distinct mechanisms. 55 56 The BFT is also capable to induce reactive oxygen species that subsequently cause direct DNA damage. Next, BFT has been shown to upregulate the transcription factors STAT3 and NF-κB. This results in immunoregulation of anti-tumor function, through IL-17 mediated tumorigenesis, upregulation of TNFα and IL-8, and increased recruitment of protumorigenic myeloid cells that suppress antitumor immunity. Finally, BFT directly activates the known oncogenic pathways of Ras/mTOR, p38, and c-myc resulting in increased intestinal cell proliferation, tumor growth, decreased tumor apoptosis, and increased intestinal cell permeability.

Third, Fusobacterium nucleatum , while not specifically secreting toxic compounds as pks + E. coli or BFT-producing ETBF, has been associated with a variety of cancers, including CRC. 57 58 59 The adhesion molecule, FadA, expressed by F. nucleatum appears to direct the carcinogenic profile of this bacteria. Notably, the FadA adhesion molecule is capable of binding E-cadherin whereby activation of the β-catenin and Wnt pathways occurs, resulting in increased cellular proliferation. 60 Additionally, Fap2 is part of a family of secretion systems that gram negative bacteria, such as F. nucleatum , utilize to secrete proteins. There are nine families of secretions systems but F. nucleatum only possesses type V, and not all strains express it. 56 Recruitment of F. nucleatum to cancer cells is facilitated by the expression of d-galactose-β(1–3)- N -acetyl-d-galactosamine (Gal-GalNAc ) on cancer cells whereby Fap2 is able to bind to and incite a proinflammatory response. 61 Additionally, Fap2 is capable of binding to the immune checkpoint inhibitor, TIGIT. Binding to this receptor results in impaired anti-tumor function of immune cells that express TIGIT: CD4, CD8, and natural killer cells. This results in decreased immune-mediated cytotoxicity to CRC and increased immune cell apoptosis. 57 Finally, LPS from F. nucleatum and gram negative bacteria in general, can act as a potent virulence factor for carcinogenesis. Utilizing colon cancer cell lines in vitro and a CRC mouse model, LPS interacts with the Toll-like receptor 4 (TLR4) pattern recognition receptor to upregulate microRNA-21, resulting in increased cancer cell and tumor proliferation and growth. 62 The LPS-TLR4 interaction also been shown to increase autophagy and chemotherapeutic resistance in a CRC model. 63 The toxigenic properties of F. nucleatum thus appear to bridge the gap of many carcinogenic processes including anti-tumor immunosuppression, activation of oncogenic pathways, autophagy, and cellular recruitment.

Microbiome and Response of Colorectal Cancer to Treatment

Much interest has been generated recently regarding the role of the microbiome in response to treatment of cancers. Hallmark studies demonstrating the role of microbiome in immunotherapy response to CTLA4 and PD-1/PD-L1 inhibition in melanoma and lung cancer support the critical interaction between these systems. 64 65 66 From a clinical standpoint, immune checkpoint blockade has limited use in CRC, as microsatellite-stable tumors (the majority of CRC) do not respond to these therapies in contrast to microsatellite-instability high (MSI high ) tumors. 67 How the microbiome can influence anti-tumor response in all CRC, independent of MSI status, is an area of active interest ( Table 3 ). Recent research has demonstrated that Bifidobacterium pseudolongum , isolated from mouse tumors that responded to immune checkpoint blockade, was able to increase intratumoral IFNγ-expressing CD4 cells in mice with colon cancer tumors. 68 In the mice colonized with B. pseudolongum , metabolite analysis revealed an increase in inosine which was also seen in mice colonized with an 11-member consortium of bacteria isolated feces from healthy human donor feces that was able to elicit an increase in IFNγ CD8 lymphocytes. This consortium resulted in improved response to immune checkpoint blockade of MC38 colon cancer xenografts with concomitant increased inosine in the stool of these animals. These studies demonstrate that the microbiome can alter response of colon cancer to immunotherapy and may provide an entry point for future studies. The microbiome has also been shown to impact the response to more traditional chemotherapy used in CRC. 69 The chemotherapy irinotecan has been shown to have activity in CRC and a component of the common FOLFIRI chemotherapy regimen. It has been demonstrated that the gut microbiome can convert byproducts of irinotecan metabolism into their original active form. This results in dose-limiting diarrhea that prevents appropriate dosing and timing of therapy for patients with CRC. 70 71 In a more direct manner, F. nucleatum has been shown to be involved in resistance to both 5- f luorouracil (5- F U) and ox aliplatin therapy, components of both the FOL F OX and FOL F IRI CRC chemotherapeutic regimens. The mechanism of this resistance is based on activation of autophagy through the Toll-like receptor and MyD88 pathways. 56 63 These examples are a growing list of members of the intestinal microbiome that can impact treatment efficacy in CRC. Many other examples exist in non-CRC malignancies which demonstrate the important interaction that the microbiome has with medications, an emerging field of research termed pharmacomicrobiomics. 72 This active area of investigation seeks to harness the microbiome for purposes of drug metabolism, efficacy, and side effect mitigation.

Table 3. Selected trials studying the interaction of the microbiome in treatment of colorectal cancer.

Study name Clinical trials Id Study type
Microbiotic product to promote microbiome health and improve chemotherapy delivery NCT05296681 Interventional
Randomized
FMT combined with immune checkpoint inhibitor and TKI in the treatment of CRC patients with advanced stage NCT05279677 Interventional
COLON-MD: Colon cancer longitudinal study (COLON-MD) NCT04751448 Observational
Gut microbiome in colorectal cancer (GO) NCT04054908 Observational
Treatment of colorectal liver metastases with immunotherapy and bevacizumab (CLIMB) NCT03698461 Interventional

Conclusion

The importance of the microbiome–cancer relationship, termed as oncobiome, 73 is no longer an area of debate given the variety of cancers that the microbiome has been shown to play a role in. Progress in a variety of technologies and reductions in their cost, such as sequencing, has facilitated the rapid growth in this arena. Beyond mere associations, the intestinal microbiome is now accepted to have an intimate relationship with CRC development. While a single causative microbe is not likely to be identified that results in CRC development, a better understanding of the intestinal microbiome community structure will yield important details on the pathogenesis of this disease. From a clinical standpoint, it is important to be familiar with the concepts presented in this review as rapid progress is being made to harness and modify the microbiome of each patient for CRC screening, prevention, and treatment. Additionally, clinicians will care for patients who take probiotics but must realize that this unregulated field has not, at present, shown benefit in CRC prevention or treatment. In fact, some probiotics contain the E. coli strain, Nissle 1917 , which harbors a pks island that is responsible for colibactin as described earlier. Current gaps in knowledge regarding the microbiome and the care of CRC patients include a greater insight into bacterial metabolites that modulate CRC development, progression, and treatment response. Additionally, progress in bioreactor technology is needed to recapitulate the complex microbiome communities that potentiate or mitigate CRC. With these bioreactors, a greater understanding of the community interaction will be acquired for patient microbiome manipulation and potential treatment. Finally, success of immunotherapy in other cancers has not necessarily applied to CRC despite the known involvement of the immune system. Further knowledge is needed regarding how the microbiome alters the CRC tumor microenvironment and immune response. Taken together, research into the role of microbiome and CRC development has made enormous strides over the past 20+ years and aims to be one of the first cancers whereby treatment, and cure, is dictated by individual microbiomes.

Footnotes

Conflict of Interest None declared.

References

  • 1.de Martel C, Georges D, Bray F, Ferlay J, Clifford G M. Global burden of cancer attributable to infections in 2018: a worldwide incidence analysis. Lancet Glob Health. 2020;8(02):e180–e190. doi: 10.1016/S2214-109X(19)30488-7. [DOI] [PubMed] [Google Scholar]
  • 2.Akimoto N, Ugai T, Zhong R. Rising incidence of early-onset colorectal cancer – a call to action. Nat Rev Clin Oncol. 2021;18(04):230–243. doi: 10.1038/s41571-020-00445-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.US Preventive Services Task Force . Davidson K W, Barry M J, Mangione C M. Screening for colorectal cancer: US preventive services task force recommendation statement. JAMA. 2021;325(19):1965–1977. doi: 10.1001/jama.2021.6238. [DOI] [PubMed] [Google Scholar]
  • 4.Imperiale T F, Ransohoff D F, Itzkowitz S H. Multitarget stool DNA testing for colorectal-cancer screening. N Engl J Med. 2014;370(14):1287–1297. doi: 10.1056/NEJMoa1311194. [DOI] [PubMed] [Google Scholar]
  • 5.Vogelstein B, Fearon E R, Hamilton S R. Genetic alterations during colorectal-tumor development. N Engl J Med. 1988;319(09):525–532. doi: 10.1056/NEJM198809013190901. [DOI] [PubMed] [Google Scholar]
  • 6.Nakatsu G, Li X, Zhou H. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat Commun. 2015;6:8727. doi: 10.1038/ncomms9727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Zeller G, Tap J, Voigt A Y. Potential of fecal microbiota for early-stage detection of colorectal cancer. Mol Syst Biol. 2014;10(11):766. doi: 10.15252/msb.20145645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Peters B A, Dominianni C, Shapiro J A. The gut microbiota in conventional and serrated precursors of colorectal cancer. Microbiome. 2016;4(01):69. doi: 10.1186/s40168-016-0218-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rezasoltani S, Asadzadeh Aghdaei H, Dabiri H, Akhavan Sepahi A, Modarressi M H, Nazemalhosseini Mojarad E. The association between fecal microbiota and different types of colorectal polyp as precursors of colorectal cancer. Microb Pathog. 2018;124:244–249. doi: 10.1016/j.micpath.2018.08.035. [DOI] [PubMed] [Google Scholar]
  • 10.Kostic A D, Chun E, Robertson L. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 2013;14(02):207–215. doi: 10.1016/j.chom.2013.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.MAGIC Study investigators ; PanGenEU Study investigators . Kartal E, Schmidt T SB, Molina-Montes E. A faecal microbiota signature with high specificity for pancreatic cancer. Gut. 2022;71(07):1359–1372. doi: 10.1136/gutjnl-2021-324755. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.O'Keefe S JD, Li J V, Lahti L. Fat, fibre and cancer risk in African Americans and rural Africans. Nat Commun. 2015;6:6342. doi: 10.1038/ncomms7342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Song M, Garrett W S, Chan A T. Nutrients, foods, and colorectal cancer prevention. Gastroenterology. 2015;148(06):1244–6.0E17. doi: 10.1053/j.gastro.2014.12.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Arayici M E, Mert-Ozupek N, Yalcin F, Basbinar Y, Ellidokuz H. Soluble and insoluble dietary fiber consumption and colorectal cancer risk: a systematic review and meta-analysis. Nutr Cancer. 2022;74(07):2412–2425. doi: 10.1080/01635581.2021.2008990. [DOI] [PubMed] [Google Scholar]
  • 15.Feng Q, Liang S, Jia H. Gut microbiome development along the colorectal adenoma-carcinoma sequence. Nat Commun. 2015;6:6528. doi: 10.1038/ncomms7528. [DOI] [PubMed] [Google Scholar]
  • 16.Attene-Ramos M S, Wagner E D, Gaskins H R, Plewa M J. Hydrogen sulfide induces direct radical-associated DNA damage. Mol Cancer Res. 2007;5(05):455–459. doi: 10.1158/1541-7786.MCR-06-0439. [DOI] [PubMed] [Google Scholar]
  • 17.Ijssennagger N, van der Meer R, van Mil S WC. Sulfide as a mucus barrier-breaker in inflammatory bowel disease? Trends Mol Med. 2016;22(03):190–199. doi: 10.1016/j.molmed.2016.01.002. [DOI] [PubMed] [Google Scholar]
  • 18.Nguyen L H, Cao Y, Hur J. The sulfur microbial diet is associated with increased risk of early-onset colorectal cancer precursors. Gastroenterology. 2021;161(05):1423–1.432E7. doi: 10.1053/j.gastro.2021.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Chen W, Liu F, Ling Z, Tong X, Xiang C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS One. 2012;7(06):e39743. doi: 10.1371/journal.pone.0039743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yazici C, Wolf P G, Kim H. Race-dependent association of sulfidogenic bacteria with colorectal cancer. Gut. 2017;66(11):1983–1994. doi: 10.1136/gutjnl-2016-313321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Nguyen L H, Ma W, Wang D D. Association between sulfur-metabolizing bacterial communities in stool and risk of distal colorectal cancer in men. Gastroenterology. 2020;158(05):1313–1325. doi: 10.1053/j.gastro.2019.12.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.European Prospective Investigation into Cancer and Nutrition Bingham S A, Day N E, Luben R.Dietary fibre in food and protection against colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC): an observational study Lancet 2003361(9368):1496–1501. [DOI] [PubMed] [Google Scholar]
  • 23.Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial Project Team Peters U, Sinha R, Chatterjee N.Dietary fibre and colorectal adenoma in a colorectal cancer early detection programme Lancet 2003361(9368):1491–1495. [DOI] [PubMed] [Google Scholar]
  • 24.Song M, Chan A T. Environmental factors, gut microbiota, and colorectal cancer prevention. Clin Gastroenterol Hepatol. 2019;17(02):275–289. doi: 10.1016/j.cgh.2018.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Roediger W E. Utilization of nutrients by isolated epithelial cells of the rat colon. Gastroenterology. 1982;83(02):424–429. [PubMed] [Google Scholar]
  • 26.Macfarlane G T, Englyst H N. Starch utilization by the human large intestinal microflora. J Appl Bacteriol. 1986;60(03):195–201. doi: 10.1111/j.1365-2672.1986.tb01073.x. [DOI] [PubMed] [Google Scholar]
  • 27.Corrêa-Oliveira R, Fachi J L, Vieira A, Sato F T, Vinolo M AR. Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunology. 2016;5(04):e73. doi: 10.1038/cti.2016.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bordonaro M, Lazarova D L, Sartorelli A C. Butyrate and Wnt signaling: a possible solution to the puzzle of dietary fiber and colon cancer risk? Cell Cycle. 2008;7(09):1178–1183. doi: 10.4161/cc.7.9.5818. [DOI] [PubMed] [Google Scholar]
  • 29.Bultman S J. Molecular pathways: gene-environment interactions regulating dietary fiber induction of proliferation and apoptosis via butyrate for cancer prevention. Clin Cancer Res. 2014;20(04):799–803. doi: 10.1158/1078-0432.CCR-13-2483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Waldecker M, Kautenburger T, Daumann H, Busch C, Schrenk D. Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J Nutr Biochem. 2008;19(09):587–593. doi: 10.1016/j.jnutbio.2007.08.002. [DOI] [PubMed] [Google Scholar]
  • 31.Chen H-M, Yu Y N, Wang J L. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013;97(05):1044–1052. doi: 10.3945/ajcn.112.046607. [DOI] [PubMed] [Google Scholar]
  • 32.Chen D, Jin D, Huang S. Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota. Cancer Lett. 2020;469:456–467. doi: 10.1016/j.canlet.2019.11.019. [DOI] [PubMed] [Google Scholar]
  • 33.Johansson M EV, Larsson J MH, Hansson G C. The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host-microbial interactions. Proc Natl Acad Sci U S A. 2011;108 01:4659–4665. doi: 10.1073/pnas.1006451107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Johansson M EV, Phillipson M, Petersson J, Velcich A, Holm L, Hansson G C. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc Natl Acad Sci U S A. 2008;105(39):15064–15069. doi: 10.1073/pnas.0803124105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ott S J, Musfeldt M, Wenderoth D F. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut. 2004;53(05):685–693. doi: 10.1136/gut.2003.025403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Sartor R B. Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology. 2004;126(06):1620–1633. doi: 10.1053/j.gastro.2004.03.024. [DOI] [PubMed] [Google Scholar]
  • 37.Lleal M, Sarrabayrouse G, Willamil J, Santiago A, Pozuelo M, Manichanh C. A single faecal microbiota transplantation modulates the microbiome and improves clinical manifestations in a rat model of colitis. EBioMedicine. 2019;48:630–641. doi: 10.1016/j.ebiom.2019.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Willemsen L EM, Koetsier M A, van Deventer S JH, van Tol E AF. Short chain fatty acids stimulate epithelial mucin 2 expression through differential effects on prostaglandin E(1) and E(2) production by intestinal myofibroblasts. Gut. 2003;52(10):1442–1447. doi: 10.1136/gut.52.10.1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Roberts C L, Keita A V, Duncan S H. Translocation of Crohn's disease Escherichia coli across M-cells: contrasting effects of soluble plant fibres and emulsifiers . Gut. 2010;59(10):1331–1339. doi: 10.1136/gut.2009.195370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Chassaing B, Koren O, Goodrich J K.Corrigendum: dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome Nature 2016536(7615):238. [DOI] [PubMed] [Google Scholar]
  • 41.Liu L, Dong W, Wang S. Deoxycholic acid disrupts the intestinal mucosal barrier and promotes intestinal tumorigenesis. Food Funct. 2018;9(11):5588–5597. doi: 10.1039/c8fo01143e. [DOI] [PubMed] [Google Scholar]
  • 42.Ocvirk S, O'Keefe S J. Influence of bile acids on colorectal cancer risk: potential mechanisms mediated by diet – gut microbiota interactions. Curr Nutr Rep. 2017;6(04):315–322. doi: 10.1007/s13668-017-0219-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Vesely M D, Kershaw M H, Schreiber R D, Smyth M J. Natural innate and adaptive immunity to cancer. Annu Rev Immunol. 2011;29:235–271. doi: 10.1146/annurev-immunol-031210-101324. [DOI] [PubMed] [Google Scholar]
  • 44.Takiishi T, Fenero C IM, Câmara N OS. Intestinal barrier and gut microbiota: shaping our immune responses throughout life. Tissue Barriers. 2017;5(04):e1373208. doi: 10.1080/21688370.2017.1373208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Elphick D A, Mahida Y R. Paneth cells: their role in innate immunity and inflammatory disease. Gut. 2005;54(12):1802–1809. doi: 10.1136/gut.2005.068601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Aghamajidi A, Maleki Vareki S. The effect of the gut microbiota on systemic and anti-tumor immunity and response to systemic therapy against cancer. Cancers (Basel) 2022;14(15):3563. doi: 10.3390/cancers14153563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Yu A I, Zhao L, Eaton K A. Gut microbiota modulate CD8 T cell responses to influence colitis-associated tumorigenesis. Cell Rep. 2020;31(01):107471. doi: 10.1016/j.celrep.2020.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Ivanov I I, Atarashi K, Manel N. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(03):485–498. doi: 10.1016/j.cell.2009.09.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Overacre-Delgoffe A E, Bumgarner H J, Cillo A R. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity. 2021;54(12):2812–2.824E7. doi: 10.1016/j.immuni.2021.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wilson M R, Jiang Y, Villalta P W.The human gut bacterial genotoxin colibactin alkylates DNA Science 2019363(6428):eaar7785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Arthur J C, Perez-Chanona E, Mühlbauer M.Intestinal inflammation targets cancer-inducing activity of the microbiota Science 2012338(6103):120–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Tomkovich S, Yang Y, Winglee K. Locoregional effects of microbiota in a preclinical model of colon carcinogenesis. Cancer Res. 2017;77(10):2620–2632. doi: 10.1158/0008-5472.CAN-16-3472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Genomics England Research Consortium Pleguezuelos-Manzano C, Puschhof J, Rosendahl Huber A. Mutational signature in colorectal cancer caused by genotoxic pks +E. coli Nature 2020580(7802):269–273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hwang S, Lee C G, Jo M. Enterotoxigenic Bacteroides fragilis infection exacerbates tumorigenesis in AOM/DSS mouse model . Int J Med Sci. 2020;17(02):145–152. doi: 10.7150/ijms.38371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Boleij A, Hechenbleikner E M, Goodwin A C. The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients . Clin Infect Dis. 2015;60(02):208–215. doi: 10.1093/cid/ciu787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Clay S L, Fonseca-Pereira D, Garrett W S. Colorectal cancer: the facts in the case of the microbiota. J Clin Invest. 2022;132(04):e155101. doi: 10.1172/JCI155101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Gur C, Ibrahim Y, Isaacson B. Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 2015;42(02):344–355. doi: 10.1016/j.immuni.2015.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mitsuhashi K, Nosho K, Sukawa Y. Association of Fusobacterium species in pancreatic cancer tissues with molecular features and prognosis . Oncotarget. 2015;6(09):7209–7220. doi: 10.18632/oncotarget.3109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hieken T J, Chen J, Hoskin T L. The microbiome of aseptically collected human breast tissue in benign and malignant disease. Sci Rep. 2016;6:30751. doi: 10.1038/srep30751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Rubinstein M R, Baik J E, Lagana S M. Fusobacterium nucleatum promotes colorectal cancer by inducing Wnt/β-catenin modulator Annexin A1 . EMBO Rep. 2019;20(04):e47638. doi: 10.15252/embr.201847638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Abed J, Emgård J E, Zamir G. Fap2 Mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed Gal-GalNAc . Cell Host Microbe. 2016;20(02):215–225. doi: 10.1016/j.chom.2016.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Meng Q, Gao Q, Mehrazarin S. Fusobacterium nucleatum secretes amyloid-like FadA to enhance pathogenicity. EMBO Rep. 2021;22(07):e52891. doi: 10.15252/embr.202152891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Yu T, Guo F, Yu Y. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy . Cell. 2017;170(03):548–5.63E18. doi: 10.1016/j.cell.2017.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Vétizou M, Pitt J M, Daillère R.Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota Science 2015350(6264):1079–1084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Leach D R, Krummel M F, Allison J P.Enhancement of antitumor immunity by CTLA-4 blockade Science 1996271(5256):1734–1736. [DOI] [PubMed] [Google Scholar]
  • 66.He D, Li X, An R. Response to PD-1-based immunotherapy for non-small cell lung cancer altered by gut microbiota. Oncol Ther. 2021;9(02):647–657. doi: 10.1007/s40487-021-00171-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Le D T, Durham J N, Smith K N.Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade Science 2017357(6349):409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mager L F, Burkhard R, Pett N.Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy Science 2020369(6510):1481–1489. [DOI] [PubMed] [Google Scholar]
  • 69.Alexander J L, Wilson I D, Teare J, Marchesi J R, Nicholson J K, Kinross J M. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(06):356–365. doi: 10.1038/nrgastro.2017.20. [DOI] [PubMed] [Google Scholar]
  • 70.Takasuna K, Hagiwara T, Hirohashi M. 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):3752–3757. [PubMed] [Google Scholar]
  • 71.Bhatt A P, Pellock S J, Biernat K A. Targeted inhibition of gut bacterial β-glucuronidase activity enhances anticancer drug efficacy. Proc Natl Acad Sci U S A. 2020;117(13):7374–7381. doi: 10.1073/pnas.1918095117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jobin C.Precision medicine using microbiota Science 2018359(6371):32–34. [DOI] [PubMed] [Google Scholar]
  • 73.Thomas R M, Jobin C. The microbiome and cancer: is the “oncobiome” mirage real? Trends Cancer. 2015;1(01):24–35. doi: 10.1016/j.trecan.2015.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinics in Colon and Rectal Surgery are provided here courtesy of Thieme Medical Publishers

RESOURCES