Association of the microbiome with colorectal cancer development (Review)

  • Authors:
    • Maha-Hamadien Abdulla
    • Disha Agarwal
    • Jaikee Kumar Singh
    • Thamer Bin Traiki
    • Manoj Kumar Pandey
    • Rehan Ahmad
    • Sandeep Kumar Srivastava
  • View Affiliations

  • Published online on: March 3, 2021     https://doi.org/10.3892/ijo.2021.5197
  • Article Number: 17
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) is the second most common malignancy causing cancer‑related mortality globally. It is the third most common type of cancer detected worldwide. The recent concept of the human body supporting a diverse community of microbes has revealed the important role these microbes play synergistically in maintaining normal homeostasis. The balance between the microbiomes and epithelial cells of the human body is essential for normal physiology. Evidence from meta‑genome analysis indicates that an imbalance in the microbiome is prominent in the guts of patients with CRC. Several studies have suggested that the gut microbiota can secrete metabolites [short‑chain fatty acids (SCFAs), vitamins, polyphenols and polyamines] that modulate the susceptibility of the colon and rectum by altering inflammation and DNA damage. The state of microbiome imbalance (dysbiosis) has been reported in patients with CRC, with an increasing population of ‘bad’ microbes and a decrease in ‘good’ microbes. The ‘good’ microbes, also known as commensal microbes, produce butyrate; however, ‘bad’ microbes cause a pro‑inflammatory state. The complex association between pathological microbial communities leading to cancer progression is not yet fully understood. An altered microbial metabolite profile plays a direct role in CRC metabolism. Furthermore, diet plays an essential role in the risk of gastrointestinal cancer development. High‑fiber diets regulate the gut microbiome and reduce the risk of CRC development, and may be fruitful in the better management of therapeutics. In the present review, the current status of the microbiome in CRC development is discussed.12

1. Introduction

Globally CRC is one of most common malignancies, ranking as the third most frequently diagnosed type of cancer and the second cause of cancer-related mortality worldwide (1). CRC stems from acquiring genetic and epigenetic alterations over the course of several years referred to as the adenoma-carcinoma sequence (2). CRC is a multifaceted and heterogenous disease and its etiology emerges from an interaction between the host and the environment. The role of microbes among environmental factors in carcinogenesis has been recognized. Infectious agents have been estimated to cause >15% of all cancers; for example, Helicobacter pylori causes gastric cancer, hepatitis B and C viruses cause hepatocellular cancer and papilloma virus causes cervical cancer. The scientific community has begun to study the role of the host-microbe interaction in cancer progression. Microbiota refers to the diverse community of microorganisms present in a specific environment. This has emerged as an important environmental factor for gastrointestinal cancer and CRC. The gut microbiome consists of a large population of bacteria (>100 billion) interacting with intestinal cells of the host, affecting immunity and the metabolome (3). Microbial composition varies along the gastrointestinal tract; 70% of microbes are located in the colorectum where interaction and crosstalk take place (4). The large intestine, particularly the colon, is more prone to developing cancer as compared to the small intestine, due to the heavy colonization of the microorganism. Cancer incidence is 12-fold more in the colon as compared to the remainder of the gastrointestinal tract (5).

The microbiota is important for normal physiological functions, such as energy harvest (6) and the immune maturity of the gut (7). Alterations in relative abundance can modulate the balance, leading to pathological conditions, such as obesity, metabolic disorders and autoimmune disease (8-10). Certain microbes and their metabolites may create a microenvironment that is more favorable to cancer growth (11). Emerging evidence suggests that the gut microbiota plays an important role in the initiation and progression of CRC (12). Previous research with germ-free animals have demonstrated a role for the microbiota in a number of models of carcinogenesis (13). The association between the microbiota, inflammation and CRC is well understood; patients with inflammatory bowel disease (IBD) are known to be more susceptible to CRC progression. The composition and diversity of microorganisms varies among different individuals, depending on diet, antibiotic/medicine consumption and chemical exposure (14). Dietary habits and lifestyle are well-established risk factors known to alter the gut microbiota. Alterations in diet have been found to affect the microbiota (15). The gut microbiota releases various metabolites that may have beneficial or damaging effects on the host. The production of metabolites, such as short-chain fatty acids (SCFAs), polyphenols, vitamins and polyamines lead to the pathogenesis of human disease. Recent findings have reported that SCFAs, specifically butyrate, play a critical role in immunomodulatory functions. Alterations in SCFA levels and other amino acid metabolites have been known to play an important role in cancer progression and metastasis (16). The biosynthesis of chemical carcinogens by microorganisms, such as N-nitroso compounds and acetaldehyde are among the potential mechanisms through which the microbiota may play a role in cancer progression. Resarch on the gut microbiota has provided a new direction and hope for the early detection of CRC, specifically during the early stages, increasing the 5-year survival rate, as compared to the late stages (17). The detection of alterations in certain microorganisms has provided a promising strategy for the early diagnosis of CRC (17). Some microbes have been shown to exert a protective effect against CRC by metabolite production, immune tolerance and outcompete with detrimental microbes (18). The better understanding of the microbiota and host interaction would provide novel opportunities for the early detection of CRC and therapeutics targeting the microbiota. The present review focuses on the role of the microbiota associated with CRC development.

2. Bacteria associated with CRC

It is known that an adult human houses approximately 1013 bacteria in the colon and other parts of the large intestine, which are chiefly responsible for maintaining gut homeostasis (19). It is the commensal relation with the gut microbiota that inhibits the invasion and colonization of pathogenic bacterial species in the gut and helps boost immunity. The gut microbiota is composed of several hundreds of bacterial species, which are predominantly anaerobic in nature, and include species such as Lactobacilli, Enterobacteriaceae, Streptococci, Bacteroides, Bifidobacterium, Fusobacterium, Enterococci, Peptostreptococcus and Atopobium (20). A few of these bacterial populations have been identified as contributors to CRC and tumorigenesis, exerting a pro-carcinogenic effect, whereas few of these are known to inhibit tumorigenesis in the colon, acting as protective species. Comparative analyses of the metagenomic and metataxonomic profiles of the gut microbiota found in the colon of patients with CRC and healthy individuals revealed that the gut microbiota composition of a patient with CRC differed significantly from that of a healthy individual, where pro-carcinogenic taxa such as Fusobacterium, Bacteroides, Escherichia and Streptococcus were found in abundance along with a reduction in the population of protective microbes, such as Bifidobacterium, Lactobacilli and Clostridium (21-24).

A pyrosequencing-based strategy adopted to study microbial dysbiosis in patients with CRC explained that the elevation in the population of Bacteroides fragilis (B. fragilis) can be linked to the development of cancer in the colon (25). B. fragilis is known to be an enterotoxigenic strain producing a bacterial toxin (metalloprotease) known as the B. fragilis toxin (BFT) responsible for the virulent properties of the strain (26). In the study by Ahn et al the taxonomic analyses of the gut microbiota was carried out by the amplification of 16srRNA sequences obtained from fecal bacterial DNA; it was observed that the increased abundance of the anaerobic Gram-negative genera, Fusobacterium and Porphyromonas, led to an increased risk of CRC development in the individuals (27). Fusobacterium nucleatum (F. nucleatum) was found in abundance in the fecal samples of patients with CRC and various clinical and metagenomic studies have highlighted a significant role of F. nucleatum in the inflammatory response of IBD and CRC (27,28). Along the similar lines of research i.e., based on the sequencing of 16srRNA, the clinical study performed by Wu et al revealed that Campylobacter and Fusobacterium species were relatively more abundant in CRC samples along with several other families, such as Enterococcaceae, Staphylococcaceae and Eubacteriaceae, exerting pathogenic effects in the colon (28). Another anaerobic commensal bacterium predominant in the gut microbiota is Escherichia coli (E. coli) and a previous study on mice inoculated with colon cancer associated E. coli strains reported an increase in the colonization of tumor-associated and mucosa-associated E. coli strains in patients with CRC (29). E. coli strains obtained from CRC samples were found to express certain toxin-producing genes, such as colibactin, which confer bacterial cells with properties to induce DNA damage and genomic instability, subsequently causing colorectal carcinogenesis (30). The statistical study by Zhang et al also revealed an elevation in the numbers of Devosia in the gut microbiota of patients with CRC (31).

Apart from elevated levels of several pathogenic microbes in the mucosal and fecal samples of patients with CRC, Ahn et al in their study, also highlighted a decrease in the population of Gram-positive strains of Clostridia, specifically Coprococcus, increasing the risk of inflammation in the colon followed by tumorigenesis (27). Some butyrate-producing genera, such as Roseburia, Faecalibacterium and Eubacterium were significantly reduced in the microbiota obtained from CRC samples, suggesting that dysbiosis in the fecal microbiota can serve as a marker for CRC detection (28,31). Another notable observation for the dysbiosis of the colon microbiota was made by Marchesi et al where upon investigating the tumor tissues of patients with CRC, a significant reduction in the population of Enterobacteriaceae was identified, namely in species such as Citrobacter, Shigella, Serratia, Salmonella and Kulyvera spp. (32). Lactic acid bacteria (LAB), such as Bifidobacterium and Lactobacilli have also been found to play a negative role in CRC (33). An imbalance in the microbial population can alter the micro-environment of the intestine, which cause an imbalance in crucial intrinsic and extrinsic factors, resulting in the initiation of cancerous growth. A summary of the bacteria playing a role in CRC development is provided in Table I.

Table I

Bacteria playing a key role in the development of colorectal cancer.

Table I

Bacteria playing a key role in the development of colorectal cancer.

BacteriaEffectorMechanisms of action
Escherichia coliGenotoxins (pks)DNA damage
Enterotoxigenic Bacteroides fragilisBacteroides fragilis toxinInflammation, immune surveillance, EMT
Fusobacterium nucleatumFadA, Fap2DNA damage, antitumor immune activity,
Streptococcus gallolyticusSGG specific bacteriocinCell proliferation
Peptostreptococcus anaerobiusUnknownROS accumulation, Cholesterol biosynthesis

3. Bacterial metabolites and CRC development

To date, various metagenomic studies performed on clinical samples obtained from individuals suffering from CRC have pointed towards the abundance of pathogenic bacterial strains of F. nucleatum, Bacteroides fragilis and E. coli. On the contrary, a significant reduction in the population of butyrate-producing strains belonging to the class Clostridia, are also considered to be a contributing factor to CRC. Thus, the understanding of the metabolites produced by these species and their mechanisms of action is critical in order to reach to better diagnostic and therapeutic conclusions and to assist in devising novel treatment strategies.

F. nucleatum

Several studies on F. nucleatum have suggested the involvement of this bacteria in triggering epithelial-mesenchymal transition (EMT) in colon tissues. F. nucleatum is known to adhere and invade epithelial cells by employing several virulence factors, such as Fusobacterium adhesin A (Fad A), Fusobacterium autotransporter protein 2 and Fusobacterium outer membrane protein A responsible for promoting pro-oncogenic signaling and CRC development (34-36). In order to provide further insight into the metabolite alteration leading to EMT induced by these virulence factors in colorectal carcinogenesis, Ma et al carried out a detailed analysis on the interactions of these virulence factors with E-cadherin/β-catenin resulting in an altered molecular mechanism upon the infection of epithelial cells with F. nucleatum (37). Upon infection, F. nucleatum was found to increase the phosphorylation of p65, a crucial component of NF-κB signaling and to enhance the production of interleukin (IL)-6, IL-1β and matrix metalloproteinase (MMP)-13. This activation of NF-κB signaling indicated that F. nucleatum induced inflammation in colon tissue, which subsequently led to tumorigenesis (37). From previous studies conducted on cell adhesion, it is a well-known fact that the loss of E-cadherin and β-catenin-mediated cell adhesion is a crucial factor for inflammation and the induction of tumorigenesis (38,39). Surprisingly, F. nucleatum did not alter the expression levels of E-cadherin and β-catenin in epithelial cells and was found to interact only with E-cadherin to induce inflammatory response and promote malignant phenotype of CRC (37). Another study by Abed et al revealed that Fap2, a Gal-GalNAc binding protein, is expressed on the surface of F. nucleatum (36). Fap 2 functions as Gal-GalNAc lectin in CRC cells, where it binds to Gal-GalNAc overexpressed in CRC cells and causes the expansion of CRC by inhibiting antitumor immune activity by targeting T cell immunoglobulin and ITIM domain (TIGIT) (35,36). This indicates that virulence factors, such as FadA along with surface proteins of F. nucleatum may be a potential target for the development of an immunotherapy against CRC.

The role of F. nucleatum in experimental colitis was recently confirmed and it was found that F. nucleatum along with dextran sodium sulfate (DSS) synergistically promoted EMT and the aggressiveness of CRC (40). F. nucleatum was found to play a pro-tumorigenic role by activating the EGFR, AKT and ERK signaling pathway in azoxymethane (AOM)/DSS-induced CRC. The F. nucleatum cell surface protein, FadA, induces DNA damage by upregulating chk2 in CRC progression. Treatment with F. nucleatum resulted in DNA damage in adenomatous polyposis coli (APC) (−/+) mice (41). The number, size and tumor burden were reduced following treatment with FadA−/− F. nucleatum as compared to WT, demonstrating a direct role of the cell surface protein of F. nucleatum in tumor progression (41). Recently, Okita et al described a heavy to moderate load of F. nucleatum (Fn) DNA associated with high microsatellite instability (MSI-H) and L/E [L: MSI-L/E: Elevated level of microsatellite alterations at selected tetra-nucleotide repeats (EMAST)] in two CRC cohorts; they further presented evidence that Fn activated factors that promote γ-H2AX, a marker for DNA damage (42). In another study, murine and human enteroid-derived monolayers (EDMs) co-cultured with Fn exhibited a downregulation of a key DNA repair protein, NEIL2 (DNA glycosylase) (43). NEIL−/− EDMs exhibited increased DNA damage and elevated cytokine levels. Furthermore, NEIL2 downregulation is mainly observed in microsatellite stable (MSS) CRC, as compared to MSI CRC, indicating that Fn accumulation induces NEIL2 downregulation, resulting in DNA damage and thus leading to CRC progression. A recent study on microRNA (miRNA/miR) profiles and proteomic analysis from Fn-infected and non-infected cells found that the generation of miR1246/92b-3p/27a-3p-rich and CXCL16/RhoA/IL-8 exosomes promoted pro-metastasis (44). Bone-marrow derived mesenchymal stem cells (BMSCs) infected with Fn have also exhibited increased tumor growth and an enhanced susceptibility to tumors an APC (−/+) mouse transplantation model. Fn-infected BMSCs also displayed accelerated cancerinitiating potential and invasiveness in a nude murine model by activating Wnt-β-catenin-transforming growth-interacting factor (TGIF) signaling pathways (45). The co-culture of human CRC cell lines (SW480 and HCT116) with Fn was also shown to result in an increased expression of mesenchymal markers, such as Vimentin, Snail and Zeb1. Furthermore, Fn infection increased the capability of intrusion, migration and tumor-sphere formation along with a high expression of CD44, indicating that Fn plays a critical role in EMT and the cancer stem cell phenotype (46). These finding confirmed that the Fn play important role in the progression of CRC.

Bacteroides fragilis

Enterotoxigenic Bacteroides fragilis (ETBF) is an enterotoxin producing bacterium found in an abundance in fecal samples obtained from patients with CRC is known to cause virulence with the aid of a metalloprotease holotoxin, BFT, also known as fragylisin (47). BFT is associated with acute diarrhea, IBD and CRC. This BFT binds to the colonic epithelial cell (CEC) surface protein receptor, initiating E-cadherin cleavage (47). E-cadherin in its intracellular domain is bound to β-catenin, which is a very crucial component of the Wnt signaling pathway. The degradation of E-cadherin releases β-catenin and facilitates EMT, and increases cell permeability by decreasing cell adhesion (48). The degradation of E-cadherin also facilitates the production of chemokines, such as IL-6, IL-8 and IL-1β, which activate NF-κB and MAPK signaling in CEC, generating inflammatory response in cells followed by carcinogenesis (49). The binding of BFT to CEC receptor also leads to the activation of key cellular pathways, such as signal transducer and activator of transcription (STAT)3 along with the formation of reactive oxygen species and nitrogen species which are known to cause DNA damage in epithelial cells and this genomic instability caused due to damaged DNA enhances expression of several oncogenes such as c-Myc by multi-folds, leading to the development of adenomas and carcinomas in the colon and rectum (50). Zamani et al investigated the mucosal colonization of ETBF to find the potential association of ETBF in benign and malignant lesions. B. fragilis was abundantly associated with patient samples as compared to healthy control (51). ETBF was increasingly associated with serrated lesions and with low-grade dysplasia adenoma. Liu et al found that ETBF increased the stemness of CRC by upregulating Nanog and Sox2 expression. They found that mechanistically, ETBF significantly elevated JMJD2B by activating TLR4 pathway (52). In a colitis CRC model, with ETBF colonization in wild-type BALB/c mice, the administration of AOM/DSS increased the rapid development of large number of polyps mainly in the colon and rectum (53). Roberti et al studied the contribution of gut microbes to the elicitation of the follicular helper T (TFH) response (54). Ileal microbiota (Bacteroides fragilis and Erysipelotrichaceae) was involved in the protective immune response against colon cancer. The ileal microbiome plays an important role in immune surveillance and in the prognosis of proximal colon cancer and chemotherapy-mediated ileal crypt apoptosis (54). Bacteroides fragilis-associated long non-coding RNA (lncRNA1: BFAL1) mediates ETBF cancer progression in CRC; BFAL1 expression is increased in CRC compared with adjacent normal tissues. ETBF promotes CRC tumor growth through BFAL1 by activating RHEB (Ras homolog)/mTOR signaling. BFAL1 and ETBF are highly expressed in tumor tissues and predict poor outcomes in CRC (55).

E. coli

E. coli is a Gram-negative commensal bacterium of the human microbiota and represents the most common cultivable and aero-anaerobic bacteria. A number of studies have demonstrated a clear link with CRC (56-60). According to the acquisition of factors of virulence, there are four E. coli phylogenic groups (A, B1, B2 and D). Groups A and B1 are generally not pathogenic; however, groups B2 and D are involved in intestinal and extra-intestinal pathogenesis. Crohn's disease, a chronic IBD known to be a risk factor for CRC, is caused by some strains of phylogroup B2 that are associated with it (60,61). Swidsinski et al (58) and Martin et al (59) demonstrated that mucosa-associated E. coli resided in higher numbers in patients with CRC than in the controls, supporting the central role of these bacteria in CRC development. Pathogenic E. coli strains synthesize various virulence factors (62). These factors consist of several toxins known as cyclomodulins, such as cytolethal distending toxins (CDT), cytotoxic necrotizing factor (CNF), cycle inhibiting factor and colibactin. Cyclomodulins are genotoxic and known to alter cell cycle progression, proliferation, cell differentiation and apoptosis (63-68). Cuevas-Ramos et al stated that a specific type of bacterial strain belonging to the B2 phylogenetic group possessed a unique gene island termed 'pks', which translates into the genotoxic compound, colibactin, possessing genotoxic properties causing DNA double-strand breaks and chromosomal instability (CSI) in human cells (68). As regards cyclomodulin-producing E. coli, a previous study revealed an increased prevalence of cyclomodulin-producing B2 E. coli in colon tumor biopsies, suggesting a possible role of such pathogenic E. coli in colon carcinogenesis (30).

This genotoxic compound generates double-strand breaks in DNA causing damage to several portions of DNA, rendering it unstable and this instability of the genome paves the way for the upregulation of expression of oncogenes, such as c-Myc, which results in the formation of adenomas and the development of CRC (56,67).

Peptostreptococcus ssp

Recent studies have found that patients with an enrichment of Peptostreptococcus stomatis (P. stomatis) and Peptostreptococcus anaerobius (P. anaerobius) have a higher risk of CRC development (69-71). Patient stool and tissue are enriched with P. anaerobius in CRC. Tsoi et al indicated the pro-tumorigenic role of P. anaerobius modulating TLR2 and 4 leading to reactive oxygen species (ROS) accumulation, resulting in cholesterol biosynthesis and proliferation (70). Purcell et al found the enrichment of P. stomatis in consensus molecular subtype 1 (CMS1) of CRC tumor tissue (71). P. stomatis supports bacteria colonization by promoting acidic and hypoxia around tumor microenvironment (71). Long et al found that P. anaerobius promoted CRC in APC (−/+) mice and altered tumor immunity by the PCWBR2-integrin α21 -PI3K-AKT-NFkB signaling pathways, identifying the PCWBR2-integrin α21 axis as a potential therapeutic target in CRC (72).

Streptococcus gallolyticus

S. gallolyticus has long been associated with CRC. Kumar et al reported significant role of few selected strains of Streptococcus gallolyticus subsp. gallolyticus (SGG) in CRC and upon a detailed analysis of its metabolite profile and mechanisms of pathogenesis, S. gallolyticus was found to enhance the proliferation of CRC cells by targeting Wnt/β-catenin signaling and upregulating β-catenin in cells, thereby elevating the expression of target oncogenes c-Myc and cyclin-D, resulting in the adenocarcinoma of CRC tissue (73). Kwong et al found that CRC diagnosis was associated with S. gallolyticus by perturbing barrier function (69). Another study found that tumorigenic conditions promoted the S. gallolyticus colonization of gut by activation of SGG-specific bacteriocin and replacing commensal enterococci (74). Some strains of S. gallolyticus have been found to induce cell proliferation designated as proliferation-promoting Sg: PP-Sg and others were not found to stimulate cell proliferation and therefore classified as non-proliferation-promoting: NP-Sg. PP-Sg found to higher colonization in mice as compared to NP-Sg, owing to the better interaction of PP-Sg with host epithelial cells. Furthermore, PP-Sg promoted CRC development in an AOM mouse model (73) (Table I).

Apart from these bacterial species mentioned above, there are several other microbes predominantly found in the fecal and mucosal samples of patients with CRC, and play a key role in pathogenesis and carcinogenesis. Microbiota analysis of healthy individuals indicated that several species of LAB namely, Lactobacillus acidophilus, L. casei, L. rhamnosus exert anti-inflammatory effects in maintaining gut homeostasis (75,76). L. rahmnosus was found to reduce the levels of β-catenin and NF-κB p65 proteins, and to induce the expression of tumor suppressor gene p53 and anti-apoptotic factor BAX in colon epithelial cell, thus preventing CRC (76). Bifidobacterium, another LAB, exerts a negative effect on CRC proliferation due to its property to reduce β-glucuronidase activity in the gut which enhances the chemotherapeutic efficacy of CPT-11, hence providing a beneficiary role in the treatment of CRC (77). The detailed role of these commensal LABs in the prevention of CRC has yet to be identified and can be explored in the near future in order to gain better insight into potential treatment strategies for CRC. The abundance of Parvimonas micra has been found in the stool of patients with CRC. P. micra was found to inhibit the NOD2 signaling, giving rise to an inflammatory and pro-tumorigenic microenvironment (78). Xu et al found that P. micra abundance was elevated in patients with CRC and was low in healthy individuals and patients with colorectal adenoma (79). The overabundance of Porphyromonas gingivalis has been found in patients with CRC (80). Yang et al reported an association between Prevotella intermedia with a higher risk of CRC development (81). Another study identified P. intermedia in a multinational cohort of fecal samples of CRC (82). Gemella morbillorum has been found to regulate IL-12 production and thereby, immunoregulation in CRC. Other species of Gemella regulate the protective function of the adaptive immune response at the mucosal surface by cleaving IgA1 (83,84). Still, however, no direct role of this bacteria has been reported in CRC development. A schematic diagram of the mechanisms of action of major species of bacteria involved in the development of CRC in humans is presented in Fig. 1.

4. Host-microbe interaction

CRC can be caused due to the cumulative effects of several genetic, epigenetic and environmental factors, which modulate microbial composition in the gut, altering the metabolite profile and the immune response of the body accordingly. CRC is not a single-step process, but rather a culmination of several steps which involve a number of changes in the genetic and epigenetic machinery of the host. The development of CRC begins with the transition of a normal epithelium into the hyperproliferative epithelium, which eventually leads to the loss of its structure and function, leading to a condition known as hyperplasia followed by dysplasia, leading to the formation of adenomas. These adenomas are non-malignant in nature and are known as polyps (85). In this section, the host-microbe interaction in the context of epigenomic modifications and immune system alterations is discussed.

Microbiota-genome/epigenome interactions

Allen and Sears highlighted the impact of the dysbiosis of the gut microbiome on genome and epigenomics of the colon epithelial cells which increased cell proliferation and tumor formation, growth and metastasis (86). DNA methylation patterns and histone marks on several promoters and enhancers were found to be dysregulated, leading to the downregulation of tumor suppressor genes (TSGs) and the upregulation of oncogenes. Several of the miRNAs (oncomiRs and anti-oncomiRs) and long non-coding RNAs have also been found to be dysregulated and associated with CRC (86). B. fragilis has been found to induce the hypermethylation of several TSGs, such as Hoxa5, Polg, Runx1, Runx3, CD37, Stx11, Tceb2, Lgr6, Cdx1 and Fut4, causing carcinoma development in the colon and rectum (86). Xia et al identified F. nucleatum and Hungatella hathewayi to be highly associated with the upregulation of DNA methyltransferases which cause the hypermethylation of promoter of TSG CDX2 and MLH1 (87). In a previous study, the APC gene and DNA mismatch repair (MMR) system were the prime genetic factors identified, which if altered or silenced, led to the development of CRC (88). In an extensive clinical analysis performed by Gagnière et al the MMR pathway was shown to be most affected by entero-pathogenic E. coli. They reported that pks+ E. coli strains downregulated the expression of MLH1 genes and inhibited the formation of MLH1 MMR protein in T-84 cells, which led to MSI and tumorigenesis (89). In addition, methylation in APC and INK4a TSG also promoted tumor development in the case of colitis-associated CRC (90-92). Streptococcus species have also been identified to be associated with APC gene hypermethylation (87).

Apart from methylation, dysregulation in the expression of miRNAs serves as a major epigenetic marker responsible for CRC. F. nucleatum is often found to increase RASA1 expression by inhibiting miR-21 expression, thus leading to chronic inflammation in the intestine, initiating carcinogenesis (93). F. nucleatum has also been found to downregulate the expression of miR-4802 and miR-18a, which lead to resistance against chemotherapeutic drugs administered to patients suffering from CRC (94). Several other oncomiRs and anti-oncomiRs were analyzed from fecal samples and gut mucosa, which revealed differential miRNA profiles in the presence of different gut microbiome, and this variation in miRNA profiles can also serve as a fingerprint for the detection and diagnosis of CRC (95,96).

Microbiota-immune system interaction

The gut microbiota is known to contribute immensely towards the maintenance of the immune system. Fluctuations in the dynamic equilibrium of this microbiota composition leads to defects in the immune system, resulting in inflammation and in tumor initiation. Genotoxins from B. fragilis and pks+ E. coli are known to induce inflammation in colon epithelial cells and this inflammatory response by the host induces genetic and epigenetic alterations, which contribute to CRC development (97). Mutations in the tumor suppressor p53 gene are commonly associated with cancer initiation, as well as progression, and they have been found to prolong the effects of NF-κB signaling, which generates the inflammatory response in cells (98). Inflammation triggers oxidative stress, increasing DNA damage and causing mutations in genes, such as APC, directing the Wnt signaling pathway and KRAS, which initiates adenoma formation and is followed by the loss of chromosome 18q and mutations in TP53, resulting in CSI and in the formation of carcinomas (99,100). Inflammation in cells often regulates the production of chemokines and cytokine-driven signaling pathways, such as the NF-κB, PI3K, Akt and ERK pathways. These signaling pathways are responsible for the initiation of tumorigenesis by either upregulating Wnt signaling, which promotes cell proliferation or by inhibiting apoptosis (101). F. nucleatum has also been found to enhance pro-inflammatory markers and the infiltration of CD11b+ myeloid immune cells and few macrophages, activating Th17 cells and TGFβ signaling, which promotes tumor initiation and angiogenesis (102).

This interaction of F. nucleatum with the host immune system suggests its prime role in CRC. F. nucleatum induces cancer development by altering the host immune response in the tumor microenvironment. F. nucleatum expresses various cell surface proteins (FadA, Fap2 and RadD) which can activate inflammatory factors and favor an environment for tumor growth by recruiting inflammatory cells. F. nucleatum can promote the immune suppression of the intestinal mucosa by inhibiting the function of T cells, natural killer (NK) cells and macrophages, resulting in CRC progression (103). Cancer-targeting NK cells are also inhibited by F. nucleatum, due to the binding of Fap2 protein on T-cells (35). It is the combination of CSI and MSI mechanisms that govern the development of adenoma-carcinoma in colorectal tissues. CSI is under the influence of immune factors, which is in a synergy with the MSI pathway characterized by changes in the epigenetics of colon epithelial cells (100); the three commonly found pro-carcinogenic microbial strains, F. nucleatum, B. fragilis and E. coli, have been observed to exert an equal influence on both the pathways driving towards CRC. By contrast, several protective microbial species known to produce butyrate and propionate exert an inhibitory effect on CRC by producing SCFAs and activating cancer-preventing phytochemicals (100). A schematic illustration of the crosstalk between several factors which play a key role in the development of CRC is presented in Fig. 2.

The composition of microbes in the host are a continuous influence on the environment and in order to protect themselves, microbes belonging to specific species tend to form mucosal biofilms. These microbial biofilms exert a protective function for the microbiota from immune factors present in the host (104). The study by Dejea et al demonstrated that biofilms were characteristically found in almost all the patients with right-sided CRC (105). The microbial composition of these biofilms was then further studied and it was predominantly found that microbes belonged to Bacteroides, Fusobacteria, Clostridia, Bifidobacterium and E. coli (104,105). These biofilms in the colon have been found to exert pro-carcinogenic effects by disrupting E-cadherin, and enhancing IL-6, Ki63 and pSTAT3 expression in the colon epithelium (105), more specifically on the right side. The presence of these biofilms often results in a poor prognosis (100). Thus, it can be conveniently concluded that biofilms can serve as a signature biomarker for CRC.

5. Microbiome-associated early diagnosis

The present review thus far discussed several microbial species which act as a driving force behind the occurrence of CRC. This dysbiosis in the microbiota is frequently studied by obtaining fecal samples. The metabolomic study of fecal dysbiosis has pointed towards the utility of microbial dysbiosis as a signature biomarker for the early prognosis and diagnosis of CRC (106). The most pathogenic bacterial strains identified as driving microbes for CRC, F. nucleatum and B. fragilis, can serve as fecal biomarkers for the early diagnosis of CRC (107,108), since elevated levels of these bacterial species have been found to be associated with an elevation in the levels of major inflammatory mediators (109). F. nucleatum has been found to increase the levels of β-catenin and TGF-β, whereas B. fragilis upregulates the expression of NF-κB, COX-2 and MMP-9, which can be indicative of early signs of CRC. Faecalibacterium prausnitzii (F. prausnitzii) was found in low levels in the fecal microbiota of patients with CRC and was responsible for low levels of β-catenin. Hence, the detection of F. nucleatum, B. fragilis and F. prausnitzii in fecal samples of individuals can help detect signs of CRC at an early stage. In addition, the analysis of these inflammatory mediators along with immunohistochemical markers, such as enhanced KRAS expression and decreased MLH1 expression can serve as effective diagnostic markers for early prognosis of CRC (109). Wu et al carried out a 16srRNA based meta-analysis on fecal biomarkers for CRC and adenoma and identified 24 biomarkers sorted into three clusters, out of which first and third cluster were found to have heterogenous population of bacteria and second cluster had relatively homogenous population comprising mainly of members of Clostridiales order (110). These clusters were claimed to be distinguishing biomarkers between adenoma and cancer in the colon and rectum (110). Microbes belonging to genera Porphyromonas, Parvimonas, Hungatella and Bacteroides were CRC-associated biomarkers, whereas Streptococcus thermophilus TH1435, Roseburia intestinalis, Blautia faecis and Eubacterium ruminantium were found to be adenoma-associated biomarkers (110). miRNAs are often found to mediate the crosstalk between microbes and the immune system. Hence, detecting fecal miRNAs and their analysis can prove to be beneficial in the prognosis of CRC and may also provide insight into the early diagnosis, as these fecal miRNAs play a significant role in the fecal dysbiosis of the microbial population (111-113). These fecal miRNAs interact with the microbiota and have been found to help F. nucleatum and E. coli invade host intestinal cells, disrupting the intestinal homeostasis (114). A preliminary investigation in this context was performed by Li et al to find a conclusive association between the differential expression of several oncomiRs and anti-oncomiRs, and distinctive microbiome profiles in CRC specimens (115). Fecal miRNAs can be upregulated, as well as downregulated under the influence of bacterial metabolites and virulence factors, exerting a carcinogenic effect (116). Among several dysregulated miRNAs, the ones which are upregulated were miR-17-92 cluster, miR-20a, miR-135, miR-144, miR-221 and miR-92a, and those found to be downregulated were miR-29a, miR-224, miR-143, miR-145 and miR-4478, significantly contributing to CRC and serving as non-invasive fecal biomarkers for the early diagnostics and therapeutic implication in CRC.

6. Exploiting the microbiome for CRC therapeutics

CRC is not a single-step process occurring due to one particular pathway or one individual bacterial strain, but rather an amalgamation of several epigenetic and immunomodulated cascades, which are initiated due to an increased abundance and synergistic effect of characteristic 'Driver and Passenger' bacteria i.e., F. nucleatum, B. fragilis, E. coli, E. faecalis and Streptococcus spp. Therefore, there are several treatment strategies devised and several of these are on their way from 'bench to bedside' in order to combat CRC. These therapeutic strategies can be broadly classified into chemotherapy and immunotherapy, following two different approaches but reaching to a similar conclusion i.e., a cure for CRC. Chemotherapy involves the administration of potent anticancer drugs (5-fluorouracil, oxaliplatin, irinotecan, etc.) in combination, to eliminate or inhibit tumor cells. Targeted therapeutics have been approved by the FDA for the treatment of CRC, such as cetuximab, a monoclonal antibody against EGFR, bevacizumab, inhibitor of angiogenesis and capecitabine, inhibitor of DNA synthesis (94). The efficacy of these chemotherapies are often found to be affected by gut microbiota-induced toxicity and chemoresistance (117-119); for instance, F. nucleatum renders CRC resistant to the chemotherapeutic drugs, oxaliplatin and 5-FU. This chemoresistance by F. nucleatum was observed on colorectal cell lines in vivo and the induction of autophagy by F. nucleatum has been found to be the prime reason behind it (119). A previous study found that F. nucleatum was also involved in the risk of recurrence following neoadjuvant chemoradiotherapy in locally advanced rectal cancer (120). Within the tumor microenvironment, the intestinal microbiota was found to regulate the functions of myeloid derived cells, thereby affecting the response to chemotherapy against cancer (85). Since the gut microbiota was often observed to interfere and affect the efficacy of anticancer drugs through its interaction with immune cells (121-123), there is an urgent need to identify alternatives to chemotherapy.

Immunotherapy is a biological method of motivating the immune system to fight by stimulating or suppressing body's own immune cells as per requirement in order to elicit an immune response. Now that microbes have been found to play a key role in development, as well as in the diagnosis of CRC, it was hypothesized by several groups that protective bacterial species if restored/maintained in the intestine, can serve as bio-therapeutic in triggering the host immune response against virulence factors generated by drivers of CRC. Several clinical trials have highlighted the antitumor potential of L. acidophilus and Bifidobacterium, and suggested their utility as a probiotic in the treatment of CRC, where it can be administered orally to patients. This will help restore the balance of commensal microbial genera in the gut and preventing intestinal toxicity (79). The study by Sivan et al detailed the potency of Bifidobacterium as a probiotic and its efficacy as a biotherapeutic agent (121). Bifidobacterium in the gut, on its oral administration to patients with CRC, reduced tumor growth to an extent similar to that obtained by treatment with anti-PD-L1 therapy, and when the probiotic in combination with therapy was used for treatment, Bifidobacterium enhanced the response of anti-PD-L1 antibody therapy and abolished tumor growth (121). Another immune checkpoint therapy involves targeting anti-CTLA4 in the intestine to induce the maturation and activation of dendritic cells and exert antitumor effects; several gut microbiota species are known to have a positive impact on this immunotherapy (122). A positive impact of the gut microbiota in several other immune therapies have been deduced which target induction of CD8+ T-lymphocytes, macrophages and the activation of dendritic cell, and major histocompatibility complex (MHC) driven pathways in order to exert antitumor effects (123). The restoration of the commensal gut microbiota population beneficial for intestinal epithelia, can trigger several immunoregulatory pathways and minimize adverse effects of immunotherapy. Thus, it may prove to be crucial in treatment of CRC and can pave the way for the development of novel therapeutic approaches (22). Another therapeutic approach involves the bioengineering of bacteria to trigger the immune response in the host. Cancer-invading bacterial cells were engineered, such that they produce a short hairpin segment of RNA which can effectively interact with β-catenin to suppress Wnt signaling and inhibit tumor formation in colorectal tissue (124). Thus, the human gut microbiota plays a crucial role in determining the efficacy and toxicity potential of a treatment, and also provides a great future prospect for developing novel biotherapeutics using gut microbiota and its metabolites for the treatment of CRC. A summary of novel therapeutic strategies for the treatment of CRC and their possible mechanisms of action is presented in Table II.

Table II

Novel therapeutic strategies for the treatment of colorectal cancer and their possible mechanisms of action.

Table II

Novel therapeutic strategies for the treatment of colorectal cancer and their possible mechanisms of action.

Therapeutic strategyMechanisms of action
L. acidophilus and Bifidobacterium as ProbioticsReduces tumor growth by enhancing anti-PD-L1 antibody response.
Probiotics in combination with immunotherapyTargeting anti-CTLA4 response and activation of dendritic cells
Bioengineering of bacteria as novel therapeuticsProduces shRNA which downregulates Wnt signaling on interaction with β-catenin

7. Conclusion

Several studies have revealed that the microbiota composition has been altered in benign lesions and in malignant tumors of the colon and rectum. Moreover, in patients with CRC, a dysbiosis in the gut microbiota has been found as compared to healthy controls. An enrichment in pro-inflammatory microbiota and the depletion in butyrate-producing bacteria has been noted. The dysbiosis of the gut microbiota in CRC results in the impairment of the intestinal epithelial barrier function, the activation of pro-inflammatory responses, genotoxin synthesis and toxic metabolite generation. The gut microbiota should be considered as a prime factor that can contribute to both CRC initiation and development. Dysbiosis can be avoided by the intake of dietary components that can alter the cancer-associated microbiome and suppress intestinal inflammation. This strategy can improve the cancer therapeutic response and prevent the progression of CRC. Furthermore, once the microbiome composition of a given patient is characterized, using a personalized medical approach, a desired bacterial equilibrium could be restored using pre and probiotics and a tailored phage therapeutics. Identifying the mechanistic pathways through which the gut microbiota influences CRC would help in devising more effective strategies for the treatment of CRC. Therefore, targeting metabolome by drugs or diet modulation and developing immunogenic peptides against cell surface proteins would improve the therapeutic efficacy for CRC and overall survival.

Availability of data and materials

Not applicable.

Authors' contributions

DA, MA, RA and SKS conceived the study, and drafted and wrote the manuscript. MKP, JKS and TBT were involved in the critical review of the manuscript. SKS, RA and MKP revised and edited the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors state that they have no competing interests.

Acknowledgments

The present study was supported by the Deanship of Scientific Research, King Saud University, for funding through the Vice Deanship of Scientific Research Chairs.

Funding

No funding was received.

References

1 

Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, Cercek A, Smith RA and Jemal A: Colorectal cancer statistics, 2020. CA Cancer J Clin. 70:145–164. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Keum N and Giovannucci E: Global burden of colorectal cancer: Emerging trends, risk factors and prevention strategies. Nat Rev Gastroenterol Hepatol. 16:713–732. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Helmink BA, Khan MAW, Hermann A, Gopalakrishnan V and Wargo JA: The microbiome, cancer, and cancer therapy. Nat Med. 25:377–388. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Dahmus JD, Kotler DL, Kastenberg DM and Kistler CA: The gut microbiome and colorectal cancer: A review of bacterial pathogenesis. J Gastrointest Oncol. 9:769–777. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Gagnière J, Raisch J, Veziant J, Barnich N, Bonnet R, Buc E, Bringer MA, Pezet D and Bonnet M: Gut microbiota imbalance and colorectal cancer. World J Gastroenterol. 22:501–518. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER and Gordon JI: An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 444:1027–1031. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Chung H, Pamp SJ, Hill JA, Surana NK, Edelman SM, Troy EB, Reading NC, Villablanca EJ, Wang S, Mora JR, et al: Gut immune maturation depends on colonization with a host-specific microbiota. Cell. 149:1578–1593. 2012. View Article : Google Scholar : PubMed/NCBI

8 

John GK and Mullin GE: The gut microbiome and obesity. Corr Oncol Rep. 18:452016. View Article : Google Scholar

9 

Dabke K, Hendrick G and Devkota S: The gut microbiome and metabolic syndrome. J Clin Invest. 129:4050–4057. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Xu H, Liu M, Cao J, Li X, Fan D, Xia Y, Lu X, Li J, Ju D and Zhao H: The dynamic interplay between the gut microbiota and autoimmune diseases. J Immunol Res. 2019:75460472019. View Article : Google Scholar : PubMed/NCBI

11 

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. 14:207–215. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Ternes D, Karta J, Tsenkova M, Wilmes P, Haan S and Letellier E: Microbiome in colorectal cancer: How to get from Meta-omics to Mechanism. Trends Microbiol. 28:401–423. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Schwabe RF and Jobin C: The microbiome and cancer. Nat Rev Cancer. 13:800–812. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI and Knight R: Bacterial community variation in human body habitats across space and time. Science. 326:1694–1697. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, et al: Linking long-term dietary patterns with gut microbial enterotypes. Science. 334:105–108. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Bhat MI and Kapila R: Dietary metabolites derived from gut microbiota: Critical modulators of epigenetic changes in mammals. Nutr Rev. 75:374–389. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Wong SH, Kwong TNY, Chow TC, Luk AKC, Dai RZW, Nakatsu G, Lam TYT, Zhang L, Wu JCY, Chan FKL, et al: Quantitation of faecal Fusobacterium improves faecal immunochemical test in detecting advanced colorectal neoplasia. Gut. 66:1441–1448. 2017. View Article : Google Scholar :

18 

Coker OO, Nakatsu G, Dai RZ, Wu WKK, Wong SH, Ng SC, Chan FKL, Sung JJY and Yu J: Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 68:654–662. 2019. View Article : Google Scholar :

19 

Ley RE, Peterson DA and Gordon JI: Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell. 124:837–848. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Tlaskalová-Hogenová H, Stepánková R, Hudcovic T, Tucková L, Cukrowska B, Lodinová-Zádníková R, Kozáková H, Rossmann P, Bártová J, Sokol D, et al: Commensal bacteria (normal microflora), mucosal immunity and chronic inflammatory and autoimmune diseases. Immunol Lett. 93:97–108. 2004. View Article : Google Scholar : PubMed/NCBI

21 

Wang T, Cai G, Qiu Y, Fei N, Zhang M, Pang X, Jia W, Cai S and Zhao L: Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 6:320–329. 2012. View Article : Google Scholar :

22 

Zackular JP, Rogers MA, Ruffin MT IV and Schloss PD: The human gut microbiome as a screening tool for colorectal cancer. Cancer Prev Res (Phila). 7:1112–1121. 2014. View Article : Google Scholar

23 

Yu YN and Fang JY: Gut microbiota and colorectal cancer. Gastrointest Tumors. 2:26–32. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Wong SH and Yu J: Gut microbiota in colorectal cancer: Mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 16:690–704. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Sobhani I, Tap J, Roudot-Thoraval F, Roperch JP, Letulle S, Langella P, Corthier G, Tran Van Nhieu J and Furet JP: Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One. 6:e163932011. View Article : Google Scholar : PubMed/NCBI

26 

Sears CL: Enterotoxigenic bacteroides fragilis: A rogue among symbiotes. Clin Micobiol Rev. 22:349–369. 2009. View Article : Google Scholar

27 

Ahn J, Sinha R, Pei Z, Dominianni C, Wu J, Shi J, Goedert JJ, Hayes RB and Yang L: Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst. 105:1907–1911. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Wu N, Yang X, Zhang R, Li J, Xiao X, Hu Y, Chen Y, Yang F, Lu N, Wang Z, et al: Dysbiosis signature of fecal microbiota in colorectal cancer patients. Microb Ecol. 66:462–470. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Bonnet M, Buc E, Sauvanet P, Darcha C, Dubois D, Pereira B, Déchelotte P, Bonnet R, Pezet D and Darfeuille-Michaud A: Colonization of the human gut by E coli and colorectal cancer risk. Clin Cancer Res. 20:859–867. 2014. View Article : Google Scholar

30 

Buc E, Dubois D, Sauvanet P, Raisch J, Delmas J, Darfeuille-Michaud A, Pezet D and Bonnet R: High prevalence of mucosa-associated E coli producing cyclomudulin and genotoxin in colon cancer. PLoS One. 8:e569642013. View Article : Google Scholar

31 

Zhang H, Chang Y, Zheng Q, Zhang R, Hu C and Jia W: Altered intestinal microbiota associated with colorectal cancer. Front Med. 13:461–470. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Marchesi JR, Dutilh BE, Hall N, Peters WHM, Roelofs R, Boleji A and Tjalsma H: Towards the human colorectal cancer microbiome. PLoS One. 6:e204472011. View Article : Google Scholar : PubMed/NCBI

33 

Zou S, Fang L and Lee MH: Dysbiosis of Gut microbiota in promoting the development of colorectal cancer. Gastroenterol Rep (Oxf). 6:1–12. 2018. View Article : Google Scholar

34 

Rubinstein MR, Wang X, Liu W, Hao Y, Cai G and Han YW: Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. 14:195–206. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Gur C, Ibrahim Y, Isaacson B, Yamin R, Abed J, Gamliel M, Enk J, Bar-On Y, Stanietsky-Kaynan N, Coppenhagen-Glazer S, et al: Binding of the Fap2 protein of Fusobacterium nucleatum to human inhibitory receptor TIGIT protects tumors from immune cell attack. Immunity. 42:344–355. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Abed J, Emgard JE, Zamir G, Faroja M, Almogy G, Grenov A, Sol A, Naor R, Pikarsky E, Atlan KA, et al: Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed gal-GalNAc. Cell Host Microbe. 20:215–225. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Ma CT, Luo HS, Gao F, Tang QC and Chen W: Fusobacterium nucleatum promotes the progression of colorectal cancer by interacting with E-cadherin. Oncol Lett. 16:2606–2612. 2018.PubMed/NCBI

38 

Kourtidis A, Lu R, Pence LJ and Anastasiadis PZ: A central role for cadherin signaling in cancer. Exp Cell Res. 358:78–85. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Kim WK, Kwon Y, Jang M, Park M, Kim J, Cho S, Jang DG, Lee WB, Jung SH, Choi HJ, et al: β-catenin activation down-regulates cell-cell junction-related genes and induces epithelial-to-mesenchymal transition in colorectal cancer. Sci Re. 9:184402019.

40 

Yu MR, Kim HJ and Park HRF: Fusobacterium nucleatum accelerates the progression of colitis-associated colorectal cancer by promoting EMT. Cancers (Basel). 12:27282020. View Article : Google Scholar

41 

Guo P, Tian Z, Kong X, Yang L, Shan X, Dong B, Ding X, Jing X, Jiang C, Jiang N and Yu Y: FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J Exp Clin Cancer Res. 39:2022020. View Article : Google Scholar : PubMed/NCBI

42 

Okita Y, Koi M, Takeda K, Ross R, Mukherjee B, Koeppe E, Stoffel EM, Galanko JA, McCoy AN, Keku TO, et al: Fusobacterium nucleatum infection correlates with two types of microsatellite alterations in colorectal cancer and triggers DNA damage. Gut Pathol. 12:462020. View Article : Google Scholar

43 

Sayed IM, Chakraborty A, Abd El-Hafeez AA, Sharma A, Sahan AZ, Huang WJM, Sahoo D, Ghosh P, Hazra TK and Das S: The DNA Glycosylase NEIL2 suppresses Fusobacterium-infection-induced inflammation and DNA damage in colonic epithelial cells. Cells. 9:19802020. View Article : Google Scholar :

44 

Guo S, Chen J, Chen F, Zeng Q, Liu WL and Zhang G: Exosomes derived from Fusobacterium nucleatum-infected colorectal cancer cells facilitate tumour metastasis by selectively carrying miR-1246/92b-3p/27a-3p and CXCL16. Gut. Nov 10–2020.Epub ahead of print. View Article : Google Scholar

45 

Lin R, Han C, Ding Z, Shi H, He R, Liu J, Qian W, Zhang Q, Fu X, Deng X, et al: Knock down of BMSC-derived Wnt3a or its antagonist analogs attenuate colorectal carcinogenesis induced by chronic Fusobacterium nucleatum infection. Cancer Lett. 495:165–179. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Wang Q, Yu C, Yue C and Liu X and Liu X: Fusobacterium nucleatum produces cancer stem cell characteristics via EMT-resembling variations. Int J Clin Exp Pathol. 13:1819–1828. 2020.PubMed/NCBI

47 

Wu S, Rhee KJ, Zhang M, Franco A and Sears CL: Bacteroides fragilis toxin stimulates intestinal epithelial cell shedding and gamma-secretase dependent E-cadherin cleavage. J Cell Sci. 120:1944–1952. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Tian X, Liu Z, Niu B, Zhang J, Lee SR, Zhao Y, Harris DC and Zheng G: E-cadherin/beta-catenin complex and the epithelial barrier. J Biomed Biotechnol. 2011:5673052011. View Article : Google Scholar

49 

Sears CL, Geis AL and Housseau F: Bacteroides fragilis subverts mucosal biology: From symbiont to colon carcinogenesis. J Clin Invest. 124:4166–4172. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Nistal E, Fernández-Fernández N, Vivas S and Olcoz JL: Factors determining colorectal cancer: The role of the intestinal microbiota. Front Oncol. 5:2202015. View Article : Google Scholar : PubMed/NCBI

51 

Zamani S, Taslimi R, Sarabi A, Jasemi S, Sechi LA and Feizabadi MM: Enterotoxigenic Bacteroides fragilis: A possible etiological candidate for bacterially-induced colorectal precancerous and cancerous lesions. Frontier Cell Infect Microbiol. 9:4492020. View Article : Google Scholar

52 

Liu QQ, Li CM, Fu LN, Wang HL, Tan J, Wang YQ, Sun DF, Gao QY, Chen YX and Fang JY: Enterotoxigenic Bacteroides fragilis induces the stemness in colorectal cancer via upregulating histone demethylase JMJD2B. Gut Microbes. 12:17889002020. View Article : Google Scholar :

53 

Hwang S, Lee CG, Jo M, Park CO, Gwon SY, Hwang S, Yi HC, Lee SY, Eom YB, Karim B and Rhee KJ: Enterotoxigenic Bacteroides fragilis infection exacerbates tumorigenesis in AOM/DSS mouse model. Int J Med Sci. 17:145–152. 2020. View Article : Google Scholar :

54 

Roberti MP, Yonekura S, Duong CPM, Picard M, Ferrere G, Tidjani Alou M, Rauber C, Iebba V, Lehmann CHK, Amon L, et al: Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer. Nat Med. 26:919–931. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Bao Y, Tang J, Qian Y, Sun T, Chen H, Chen Z, Sun D, Zhong M, Chen H, Hong J, et al: Long noncoding RNA BFAL1 mediates enterotoxigenic Bacteroides fragilis-related carcinogenesis in colorectal cancer via the RHEB/mTOR pathway. Cell Death Dis. 10:6752019. View Article : Google Scholar : PubMed/NCBI

56 

Arthur JC, Perez-Chanona E, Muhlbauer M, Tomkovich S, Uronis JM, Fan TJ, Campbell BJ, Abujamel T, Dogan B, Rogers AB, et al: Intestinal inflammation targets cancer-inducing activity of the microbiota. Science. 338:120–123. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Tjalsma H, Boleij A, Marchesi JR and Dutilh BE: A bacterial driver-passenger model for colorectal cancer: Beyond the usual suspects. Nat Rev Microbiol. 10:575–582. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Swidsinski A, Khilkin M, Kerjaschki D, Schreiber S, Ortner M, Weber J and Lochs H: Association between intraepithelial Escherichia coli and colorectal cancer. Gastroenterology. 115:281–286. 1998. View Article : Google Scholar : PubMed/NCBI

59 

Martin HM, Campbell BJ, Hart CA, Mpofu C, Nayar M, Singh R, Englyst H, Williams HF and Rhodes JM: Enhanced Escherichia coli adherence and invasion in Crohn's disease and colon cancer. Gastroenterology. 127:80–93. 2004. View Article : Google Scholar : PubMed/NCBI

60 

Darfeuille-Michaud A, Neut C, Barnich N, Lederman E and Di Martino P: Presence of adherent Escherichia coli strains in ileal mucosa of patients with Crohn's disease. Gastroenterology. 115:1405–1413. 1998. View Article : Google Scholar : PubMed/NCBI

61 

Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser AL, Barnich N, Bringer MA, Swidsinski A, Beaugerie L and Colombel JF: High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn's disease. Gastroenterology. 127:412–421. 2004. View Article : Google Scholar : PubMed/NCBI

62 

Lax AJ: Opinion: Bacterial toxins and cancer-a case to answer? Nat Rev Microbiol. 3:343–349. 2005. View Article : Google Scholar : PubMed/NCBI

63 

Thelestam M and Frisan T: Cytolethal distending toxins. Rev Physiol Biochem Pharmacol. 152:111–133. 2004. View Article : Google Scholar : PubMed/NCBI

64 

Falzano L, Filippini P, Travaglione S, Miraglia AG, Fabbri A and Fiorentini C: Escherichia coli cytotoxic necrotizing factor 1 blocks cell cycle G2/M transition in uroepithelial cells. Infect Immun. 74:3765–3772. 2006. View Article : Google Scholar : PubMed/NCBI

65 

Malorni W and Fiorentini C: Is the Rac GTPase-activating toxin CNF1 a smart hijacker of host cell fate? FASEB J. 20:606–609. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Jubelin G, Chavez CV, Taieb F, Banfield MJ, Samba-Louaka A, Nobe R, Nougayrède JP, Zumbihl R, Givaudan A, Escoubas JM and Oswald E: Cycle inhibiting factors (CIFs) are a growing family of functional cyclomodulins present in invertebrate and mammal bacterial pathogens. PLoS One. 4:e48552009. View Article : Google Scholar : PubMed/NCBI

67 

Nougayrede JP, Homburg S, Taieb F, Boury M, Brzuszkiewicz E, Gottschalk G, Buchrieser C, Hacker J, Dobrindt U and Oswald E: Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 313:848–851. 2006. View Article : Google Scholar : PubMed/NCBI

68 

Cuevas-Ramosa G, Petita CR, Marcqa I, Bourya M, Oswalda E and Nougayrède JP: Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc Natl Acad Sci USA. 107:11357–11542. 2010.

69 

Kwong TNY, Wang X, Nakatsu G, Chow TC, Tipoe T, Dai RZW, Tsoi KKK, Wong MCS, Tse G, Chan MTV, et al: Association between bactereia from specific microbes and subsequent diagnosis of colorectal cancer. Gastroenterology. 155:383–390.e8. 2018. View Article : Google Scholar

70 

Tsoi H, Chu ESH, Zhang X, Sheng J, Nakatsu G, Ng SC, Chan AWH, Chan FKL, Sung JJY and Yu J: Peptostreptococcus anaerobius induces intracellular cholesterol biosynthesis in colon cells to induce proliferation and causes dysplasia in mice. Gastroenterology. 152:1419–1433.e5. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Purcell RV, Visnovska M, Biggs PJ, Schmeier S and Frizelle FA: Distinct gut microbiome patterns associate with consensus molecular subtypes of colorectal cancer. Sci Rep. 7:115902017. View Article : Google Scholar : PubMed/NCBI

72 

Long X, Wong CC, Tong L, Chu ESH, Ho Szeto C, Go MYY, Coker OO, Chan AWH, Chan FKL, Sung JJY and Yu J: Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat Microbiol. 4:2319–2330. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Kumar R, Herold JL, Schady D, Davis J, Kopetz S, Martinez-Moczygemba M, Murray BE, Han F, Li Y, Callaway E, et al: Streptococcus gallolyticus Subsp gallolyticus promotes colorectal tumor development. PLoS Pathog. 13:e10064402017. View Article : Google Scholar

74 

Aymeric L, Donnadieu F, Mulet C, du Merle L, Nigro G, Saffarian A, Bérard M, Poyart C, Robine S, Regnault B, et al: Colorect a l ca ncer sp eci f ic cond itions promote Streptococcus gallolyticus gut colonization. Proc Natl Acad Sci USA. 115:E283–E291. 2018. View Article : Google Scholar

75 

Konstantinov SR, Kuipers EJ and Peppelenbosch MP: Functional genomic analysis of Gut microbiota for CRC screening. Nat Rev Gastroenterol Hepatol. 10:741–745. 2013. View Article : Google Scholar : PubMed/NCBI

76 

Gamallat Y, Meyiah A, Kuugbee ED, Hago AM, Chiwala G, Awadasseid A, Bamba D, Zhang X, Shang X, Luo F and Xin Y: Lactobacillus rhamnosus induced epithelial cell apoptosis, ameliorates inflammation and prevents colon cancer development in an animal model. Biomed Pharmacother. 83:536–541. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Wallace BD, Wang H, Lane KT, Scott JE, Orans J, Koo JS, Venkatesh M, Jobin C, Yeh LA, Mani S and Redinbo MR: Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science. 330:831–835. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Marchesan J, Jiao YZ, Schaff RA, Hao J, Morelli T, Kinney JS, Gerow E, Sheridan R, Rodrigues V, Paster BJ, et al: TLR4, NOD1 and NOD2 mediate immune recognition of putative newly identified periodontal pathogens. Mol Oral Microbiol. 31:243–258. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Xu J, Yang M, Wang D, Zhang S, Yan S, Zhu Y and Chen W: Alteration of the abundance of Parvimonas micra in the gut along the adenoma-carcinoma sequence. Oncol Lett. 20:1062020. View Article : Google Scholar :

80 

Allali I, Boukhatem N, Bouguenouch L, Hardi H, Boudouaya HA, Cadenas MB, Ouldim K, Amzazi S, Azcarate-Peril MA and Ghazal H: Gut microbiome of Moroccan colorectal cancer patients. Med Microbiol Immunol (Berl). 207:211–225. 2018. View Article : Google Scholar

81 

Yang Y, Cai Q, Shu XO, Steinwandel MD, Blot WJ, Zheng W and Long J: Prospective study of oral microbiome and colorectal cancer risk in low-income and African American populations. Int J Cancer. 144:2381–2389. 2019. View Article : Google Scholar :

82 

Dai Z, Coker OO, Nakatsu G, Wu WKK, Zhao L, Chen Z, Chan FKL, Kristiansen K, Sung JJY, Wong SH and Yu J: Multi-cohort analysis of colorectal cancer metagenome identified altered bacteria across populations and universal bacterial markers. Microbiome. 6:702018. View Article : Google Scholar : PubMed/NCBI

83 

Sobrinho AR: Cytokine production in response to endodontic infection in germ-free mice. Oral Microbiol Immunol. 17:344–353. 2002. View Article : Google Scholar

84 

Lomholt JA and Kilian M: Immunoglobulin A1 protease activity in Gemella haemolysans. J Clin Microbiol. 38:2760–2762. 2000. View Article : Google Scholar : PubMed/NCBI

85 

Montalban-Arques A and Scharl M: Intestinal microbiota and colorectal carcinoma: Implications for pathogenesis, diagnosis, and therapy. EBioMedicine. 48:648–655. 2019. View Article : Google Scholar : PubMed/NCBI

86 

Allen J and Sears CL: Impact of the gut microbiome on the genome and epigenome of colon epithelial cells: Contributions to colorectal cancer development. Genome Med. 11:112019. View Article : Google Scholar : PubMed/NCBI

87 

Xia X, Wu WKK, Wong SH, Liu D, Kwong TNY, Nakatsu G, Yan PS, Chuang YM, Chan MW, Coker OO, et al: Bacteria pathogens drive host colonic epithelial cell promoter hypermethylation of tumor suppressor genes in colorectal cancer. Microbiome. 8:1082020. View Article : Google Scholar : PubMed/NCBI

88 

Sobhani I, Rotkopf H and Khazaie K: Bacteria-related changes in host DNA methylation and risk for CRC. Gut Microbes. 12:e18008982020. View Article : Google Scholar

89 

Gagnière J, Bonnin V, Jarrousse AS, Cardamone E, Agus A, Uhrhammer N, Sauvanet P, Déchelotte P, Barnich N, Bonnet R, et al: Interactions between microsatellite instability and human gut colonization by Escherichia coli in colorectal cancer. Clin Sci (Lond). 131:471–485. 2017. View Article : Google Scholar

90 

Foran E, Garrity-Park MM, Mureau C, Newell J, Smyrk TC, Limburg PJ and Egan LJ: Upregulation of DNA methyltransferase-mediated gene silencing, anchorage-independent growth, and migration of colon cancer cells by interleukin-6. Mol Cancer Res. 8:471–481. 2010. View Article : Google Scholar : PubMed/NCBI

91 

Li Y, Deuring J, Peppelenbosch MP, Kuipers EJ, de Haar C and van der Woude CJ: IL-6-induced DNMT1 activity mediates SOCS3 promoter hypermethylation in ulcerative colitis-related colorectal cancer. Carcinogenesis. 3:1889–1896. 2012. View Article : Google Scholar

92 

Hartnett L and Egan LJ: Inflammation, DNA methylation and colitis-associated cancer. Carcinogenesis. 33:723–731. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Hashemi Goradel N, Heidarzadeh S, Jahangiri S, Farhood B, Mortezaee K, Khanlarkhani N and Neghadari B: Fusobacterium nucleatum and colorectal cancer: A mechanistic overview. J Cell Physiol. 234:2337–2344. 2018. View Article : Google Scholar : PubMed/NCBI

94 

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. 170:548–563.e16. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Tarallo S, Ferrero G, Gallo G, Francavilla A, Clerico G, Realis Luc A, Manghi P, Thomas AM, Vineis P, Segata N, et al: Altered fecal small RNA profiles in colorectal cancer reflected gut microbiome composition in stool samples. mSystems. 4:200289–19. 2019. View Article : Google Scholar

96 

Yuan C, Steer CJ and Subramanian S: Host-microRNA-Microbiota interaction in colorectal cancer. Genes (Basel). 10:2702019. View Article : Google Scholar

97 

Kang M and Martin A: Microbiome and colorectal cancer: Unraveling host-microbiota interactions in colitis-associated colorectal cancer development. Semin Immunol. 13:3–13. 2017. View Article : Google Scholar

98 

Cooks T, Pateras IS, Tarcic O, Solomon H, Schetter AJ, Wilder S, Lozano G, Pikarsky E, Forshew T, Rosenfeld N, et al: Mutant p53 prolongs NF-κB activation and promotes chronic inflammation and inflammation-associated colorectal cancer. Cancer Cell. 23:634–646. 2013. View Article : Google Scholar : PubMed/NCBI

99 

Lasry A, Zinger A and Ben-Neriah Y: Inflammatory networks underlying colorectal cancer. Nat Immunol. 17:230–240. 2016. View Article : Google Scholar : PubMed/NCBI

100 

Ruskov H, Burcharth J and Pommergaard HC: Linking gut microbiota to colorectal cancer. J Cancer. 8:3378–3395. 2017. View Article : Google Scholar

101 

Grivennikov SI: Inflammation and colorectal cancer: Colitisassociated neoplasia. Semin Immunopathol. 35:229–244. 2013. View Article : Google Scholar

102 

Coussens LM and Pollard JW: Leukocytes in mammary development and cancer. Cold Spring Harb Perspect Biol. 3:a0032852011. View Article : Google Scholar

103 

Chen T, Li Q, Wu J, Wu Y, Peng W, Li H, Wang J, Tang X, Peng Y and Fu X: Fusobacterium nucleatum promotes M2 polarization of macrophages in the microenvironment of colorectal tumours via a TLR4-dependent mechanism. Cancer Immunol Immunother. 67:1635–1646. 2018. View Article : Google Scholar : PubMed/NCBI

104 

Drewes JL, Housseau F and Sears CL: Sporadic colorectal cancer: Microbial contributors to disease prevention, development and therapy. Br J Cancer. 115:273–280. 2016. View Article : Google Scholar : PubMed/NCBI

105 

Dejea CM, Wick EC, Hechenbleikner EM, White JR, Mark Welch JL, Rossetti BJ, Peterson SN, Snesrud EC, Borisy GG, Lazarev M, et al: Microbiota organization is a distinct feature of proximal colorectal cancers. Proc Natl Acad Sci USA. 111:18321–18326. 2014. View Article : Google Scholar : PubMed/NCBI

106 

Villéger R, Lopès A, Veziant J, Gagnière J, Barnich N, Billard E, Boucher D and Bonnet M: Microbial markers in colorectal cancer detection and/or prognosis. World J Gastroenterol. 24:2327–2347. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Kostic AD, Gevers D, Pedamallu CS, Michaud M, Duke F, Earl AM, Ojesina AI, Jung J, Bass AJ, Tabernero J, et al: Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 22:292–298. 2012. View Article : Google Scholar :

108 

Yu J, Feng Q, Wong SH, Zhang D, Liang QY, Qin Y, Tang L, Zhao H, Stenvang J, Li Y, et al: Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut. 66:70–78. 2017. View Article : Google Scholar

109 

Wei Z, Cao S, Liu S, Yao Z, Sun T, Li Y, Li J, Zhang D and Zhou Y: Could gut microbiota serve as prognostic biomarker associated with colorectal cancer patients' survival? A pilot study on relevant mechanism. Oncotarget. 7:46158–46170. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Wu Y, Ziao N, Zhu R, Zhang Y, Wu D, Wang AJ, Fang S, Tao L, Li Y, Cheng S, et al: Identification of microbial markers across populations in early detection of colorectal cancer. bioRixv. https://doi.org/10.1101/2020.08.16.253344.

111 

Liu S, da Cunha AP, Rezende RM, Cialic R, Wei Z, Bry L, Comstock LE, Gandhi R and Weiner HL: The host shapes the gut Microbiota via fecal MicroRNA. Cell Host Microbe. 19:32–43. 2016. View Article : Google Scholar : PubMed/NCBI

112 

Yuan C, Burns MB, Subramanian S and Blekhman R: Interaction between Host MicroRNAs and the Gut Microbiota in Colorectal Cancer. mSystems. 3:e00205–17. 2018. View Article : Google Scholar : PubMed/NCBI

113 

Sarshar M, Scribano D, Ambrosi C, Palamara AT and Masotti A: Fecal microRNAs as innovative biomarkers of intestinal diseases and effective players in Host-Microbiome interactions. Cancers (Basel). 12:21742020. View Article : Google Scholar

114 

Yang T, Owen JL, Lightfoot YL, Kladde MP and Mohamadzadeh M: Microbiota impact on the epigenetic regulation of colorectal cancer. Trends Mol Med. 19:714–725. 2013. View Article : Google Scholar : PubMed/NCBI

115 

Li M, Chen WD and Wang YD: The roles of the gut microbiotamiRNA interaction in the host pathophysiology. Mol Med. 26:1012020. View Article : Google Scholar

116 

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. 357:1156–1160. 2017. View Article : Google Scholar : PubMed/NCBI

117 

Serna G, Ruiz-Pace F, Hernando J, Alonso L, Fasani R, Landolfi S, Comas R, Jimenez J, Elez E, Bullman S, et al: Fusobacterium nucleatum persistance and risk of recurrence after preoperative treatment in locally advanced rectal cancer. Ann Oncol. 31:1366–1375. 2020. View Article : Google Scholar : PubMed/NCBI

118 

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. 342:967–970. 2013. View Article : Google Scholar : PubMed/NCBI

119 

Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, et al: The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 342:971–976. 2013. View Article : Google Scholar : PubMed/NCBI

120 

Xu X and Zhang X: Effects of cyclophosphamide on immune system and gut microbiota in mice. Microbiol Res. 171:97–106. 2015. View Article : Google Scholar : PubMed/NCBI

121 

Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley JM, Benyamin FW, Lei YM, Jabri B, Alegre ML, et al: Commensal Bifidobacterium promotes antitumor activity and facilitates anti-PD-L1 efficacy. Science. 350:1084–1089. 2015. View Article : Google Scholar : PubMed/NCBI

122 

Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, et al: Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 350:1079–1084. 2015. View Article : Google Scholar : PubMed/NCBI

123 

Uribe-Herranz M, Bittinger K, Rafail S, Guedan S, Pierini S, Tanes C, Ganetsky A, Morgan MA, Gill S, Tanyi JL, et al: Gut microbiota modulates adoptive cell therapy via CD8α dendritic cells and IL-12. JCI Insight. 3:e949522018. View Article : Google Scholar

124 

Hold GL: Gastrointestinal microbiota and colon cancer. Dig Dis. 34:244–250. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 58 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Abdulla M, Agarwal D, Singh JK, Traiki TB, Pandey MK, Ahmad R and Srivastava SK: Association of the microbiome with colorectal cancer development (Review). Int J Oncol 58: 17, 2021
APA
Abdulla, M., Agarwal, D., Singh, J.K., Traiki, T.B., Pandey, M.K., Ahmad, R., & Srivastava, S.K. (2021). Association of the microbiome with colorectal cancer development (Review). International Journal of Oncology, 58, 17. https://doi.org/10.3892/ijo.2021.5197
MLA
Abdulla, M., Agarwal, D., Singh, J. K., Traiki, T. B., Pandey, M. K., Ahmad, R., Srivastava, S. K."Association of the microbiome with colorectal cancer development (Review)". International Journal of Oncology 58.5 (2021): 17.
Chicago
Abdulla, M., Agarwal, D., Singh, J. K., Traiki, T. B., Pandey, M. K., Ahmad, R., Srivastava, S. K."Association of the microbiome with colorectal cancer development (Review)". International Journal of Oncology 58, no. 5 (2021): 17. https://doi.org/10.3892/ijo.2021.5197