Colorectal cancer represents the third most common cancer type worldwide and is a leading cause of cancer-related mortality in the United States and Western countries. Rodent models have been invaluable to study the etiology of colorectal cancer and to test novel chemoprevention avenues. In the past, the laboratory mouse has become one of the best preclinical models for these studies due to the availability of genetic information for commonly used mouse strains with well-established and precise gene targeting and transgenic techniques. Well-established chemical mutagenesis technologies are also being used to develop mouse and rat models of colorectal cancer for prevention and treatment studies. In addition, xenotransplantation of cancer cell lines and patient-derived xenografts has been useful for preclinical prevention studies and drug development. This review focuses on the recent use of rodent models to evaluate the utility of novel strategies in the prevention of colon cancers including immune prevention approaches and the manipulation of the intestinal microbiota.

Colorectal cancer represents the third most common cancer type worldwide and is a leading cause of cancer-related mortality in the United States and Western countries (1). Colorectal cancer is caused by the accumulation of genetic mutations and epigenetic alterations that drive the transformation of the colonic mucosa and the progression from early colonic lesions to adenomas and adenocarcinomas. Although the vast majority of colorectal cancers are considered sporadic, about 2% to 5% of colorectal cancers are caused by pathogenic variants in DNA mismatch-repair (MMR) genes or the adenomatous polyposis coli (APC) (2) gene resulting in the highly penetrant hereditary cancer syndromes Lynch syndrome (LS) and familial adenomatous polyposis (FAP), respectively (3, 4). Another 20% to 30% of colorectal cancer cases are considered to have a hereditary component due to an associated family history or germline mutations in other genes (5). In addition to genetic alterations, environmental and dietary factors significantly impact the development of colorectal cancer. For example, although red meat consumption and smoking increase the risk for colorectal cancer, vegetables and fruit with high fiber content decrease colorectal cancer risk (6–8). In addition, resistant starch, a component of dietary fiber in certain foods, significantly lowered the risk of extra colonic cancers in LS patients in the CAPP2 clinical trial study (9). Furthermore, chronic stress and the widespread use of antibiotics with alteration of the gut microbiota may be linked to early-onset colorectal cancer (10, 11). Although in recent years the overall incidence of colorectal cancer has declined in older individuals (>50 years), likely due to more extensive colorectal cancer population screening, the colorectal cancer incidence has significantly increased in younger adults (12) with the causes remaining largely unknown. Despite numerous advances in understanding the genetic and environmental factors affecting the etiology and progression of colorectal cancer, preclinical animal models continue to be indispensable not only for studying the mechanisms of pathogenesis but also for developing novel approaches for the prevention and treatment of colorectal cancer. In recent years, comprehensive reviews have been published that describe these models as well as their advantages and disadvantages in different uses (13–15). In prevention studies, mouse models have previously been used to study the impact of diets, such as so-called Western-style diets, on intestinal tumor development (16). In addition, nonsteroidal anti-inflammatory drugs (NSAID) such as aspirin, sulindac, and naproxen have been tested on the prevention of intestinal cancer in LS and FAP mouse models (17–19). Here we focus on the more recent use of rodent models testing novel interventions for the prevention of colorectal cancer, including immune preventive approaches and strategies involving the manipulation of intestinal microbiota.

Approaches that use immunization are currently a focus of intensive research efforts as they hold promise in the prevention and treatment of colorectal cancer. Vaccination with tumor antigens can enhance infiltration of cytotoxic T-lymphocytes (CTL) into tumors and stimulate their cytotoxic activities against tumor cells expressing specific antigens leading to their elimination and thus preventing tumor formation or recurrence (20, 21). Once the immune system is primed by vaccination, T-memory cells are present for years and can be boosted periodically by further vaccination. Tumor antigens (TAs) are presented by MHC I/II (also known as human leukocyte antigen I and II (HLA I and II)) molecules to T cells to initiate immune responses and act as crucial factors for vaccine formulation (20–22). TAs can be divided into two subtypes: tumor-associated antigens (TAAs), which are overexpressed in tumor cells compared with normal cells, and tumor-specific antigens (TSAs), which are exclusively expressed in tumor cells (20–22). Different approaches have been used to administer TA vaccines, including the use of synthetic peptides, recombinant proteins (23, 24), whole tumor cells (25, 26), tumor antigen expression from viral vectors or bacteria (27, 28), nucleic acids (21, 22), dendritic cells (29, 30), and exosomes (31). Here we will focus on studies using TA vaccines that include peptide or full-length protein vaccines and describe examples in mouse models that used colorectal cancer TAs with frameshift-derived peptides (FSP) as well as TAs specifically expressed in colorectal cancers including carcinoembryonic antigen (32), mucin 1 (MUC1), squamous cell carcinoma antigen recognized by T-cell 3 (SART3), and KRAS (Table 1).

Table 1.

Vaccination in mouse colorectal cancer (CRC) models.

Animal strainTumor modelVaccination strategyTreatment outcomeReference
C57BL/6J VCMsh2 (Villin-Cre/Msh2loxP/loxP) FSPs: Nacad, Maz, Senp6, and Xirp1, Reduced CRC and promoted the proliferation of cytotoxic T cells to mediate anticancer immune responses 17  
BALB/c CT26 tumor cell xenotransplant KRAS (G12D)-inhibitory bicyclic peptide KS-58 Reduced CRC 36,37  
C57BL/6J CEA-A2Kb transgene CEA peptides + 3H1 Reduced the growth of MC-38-CEA xenotransplants and induced the antitumor immunity by increasing the proliferation and secretion of Th1 cytokines by CD4+ cells 38,39  
C57BL/6J ApcMin/CEA CEA-based vaccine Reduced CRC tumor multiplicity 40  
C57BL/6J MUC1 transgene + MC38 tumor cells xenotransplant MUC1-specific vaccine Prevented the growth of MUC1 overexpressing MC38 tumor cell xenotransplants and stimulated interferon-g producing CD4+ Th1 cells and CD8+ CTLs 42,43  
C57BL/6J MUC1+IL10−/− MUC1-specific vaccine Prevented dysplasia, IBD, and development of IBD-associated colon cancer 44  
BALB/c CT26 tumor cell xenotransplant CD133+CCSCs expressing MUC1 Reduced growth of CT26 CRC xenotransplants, increased NK cell cytotoxicity, and promoted the release of IFNγ, Perforin, and Granzyme B 45  
BALB/c CT26 tumor cell xenotransplant Polyplex micelles containing SART3, CD40L, and GM-CSF Reduced growth of CT26 CRC xenotransplants and stimulated CTL and NK activities 53  
C57BL/6N GL261 glioma and B16f1 melanoma cells xenotransplant Long survivin peptide Increased CD8-mediated tumor cell death and increased overall survival 55  
BALB/c CT26 tumor cell xenotransplant Long survivin synthetic peptide (LSP) Inhibited the growth of CT26 CRC xenotransplants and upregulated tumor infiltration of CD4+ and CD8+ T cells 56  
Animal strainTumor modelVaccination strategyTreatment outcomeReference
C57BL/6J VCMsh2 (Villin-Cre/Msh2loxP/loxP) FSPs: Nacad, Maz, Senp6, and Xirp1, Reduced CRC and promoted the proliferation of cytotoxic T cells to mediate anticancer immune responses 17  
BALB/c CT26 tumor cell xenotransplant KRAS (G12D)-inhibitory bicyclic peptide KS-58 Reduced CRC 36,37  
C57BL/6J CEA-A2Kb transgene CEA peptides + 3H1 Reduced the growth of MC-38-CEA xenotransplants and induced the antitumor immunity by increasing the proliferation and secretion of Th1 cytokines by CD4+ cells 38,39  
C57BL/6J ApcMin/CEA CEA-based vaccine Reduced CRC tumor multiplicity 40  
C57BL/6J MUC1 transgene + MC38 tumor cells xenotransplant MUC1-specific vaccine Prevented the growth of MUC1 overexpressing MC38 tumor cell xenotransplants and stimulated interferon-g producing CD4+ Th1 cells and CD8+ CTLs 42,43  
C57BL/6J MUC1+IL10−/− MUC1-specific vaccine Prevented dysplasia, IBD, and development of IBD-associated colon cancer 44  
BALB/c CT26 tumor cell xenotransplant CD133+CCSCs expressing MUC1 Reduced growth of CT26 CRC xenotransplants, increased NK cell cytotoxicity, and promoted the release of IFNγ, Perforin, and Granzyme B 45  
BALB/c CT26 tumor cell xenotransplant Polyplex micelles containing SART3, CD40L, and GM-CSF Reduced growth of CT26 CRC xenotransplants and stimulated CTL and NK activities 53  
C57BL/6N GL261 glioma and B16f1 melanoma cells xenotransplant Long survivin peptide Increased CD8-mediated tumor cell death and increased overall survival 55  
BALB/c CT26 tumor cell xenotransplant Long survivin synthetic peptide (LSP) Inhibited the growth of CT26 CRC xenotransplants and upregulated tumor infiltration of CD4+ and CD8+ T cells 56  

TAA-based vaccination: The use of FSP antigens (or neoantigens) is a new promising immune-preventive approach against cancers with microsatellite instability (MSI; refs. 17, 33, 34). FSPs are TSAs that result from frameshift mutations that occur at an increased frequency at coding microsatellites associated with MMR-deficient (MMR-d) tumors including the hereditary form Lynch syndrome (LS). Recent studies identified immunogenic MSI-associated neoantigens in MMR-d colorectal cancers (23, 33) that promoted the proliferation of cytotoxic T cells to mediate anticancer immune responses (35). A recent study by Gebert and colleagues provided evidence of the tumor-preventive potential of FSP-based vaccines in the VCMsh2 mouse model of LS (17). The authors tested and validated the immunogenicity of four candidate FSPs, Nacad, Maz, Senp6, and Xirp1, that are shared by the majority of MSI-positive intestinal tumors in VCMsh2 mice. Prevaccination with these FSPs reduced the tumor load and increased survival of the mice, suggesting that vaccination with shared FSP neoantigens may be a suitable approach for cancer prevention in LS (17). These studies represent important steps toward the development of neoantigen-based vaccines for the prevention of MSI-positive cancers in humans.

KRAS oncogene mutations, and specifically KRAS (G12D) and KRAS (G12V), are found in a significant proportion of colorectal cancers (20). Recent studies showed that KRAS mutant peptides stimulate CTLs in colorectal cancers. Sakamoto and colleagues showed that administration of the KRAS (G12D)-inhibitory bicyclic peptide KS-58 in mice with CT26 colorectal cancer tumor cell transplants expressing KRASG12D inhibited tumor cell proliferation and suppressed tumor growth (36, 37).

TAA-based vaccination: In addition, TAAs overexpressed in human colorectal cancer have been tested in mouse models. One example is CEA (carcinoembryonic antigen), a nonspecific serum biomarker that is elevated in various malignancies including colorectal cancers and other cancers. CEA is used to monitor colorectal cancer metastasis in human patients and transgenic mouse models. Vaccination of C57BL/6J-CEA-A2Kb transgenic mice (expressing both CEA and HLA-A2) with a combination of CEA peptides and the anti-idiotype antibody 3H1 (mimicking a specific epitope on CEA) increased the proliferation of splenocytes and secretion of Th1 cytokines by CD4+ T cells and induced antitumor immunity against MC-38-CEA-A2Kb colon carcinoma cell transplants (38, 39). A CEA-based vaccine also significantly reduced tumor multiplicity in ApcMin/CEA transgenic mice and significantly improved overall survival (OS) compared with ApcMin mice without causing autoimmune complications (40). These data in mice suggest that immunization with CEA-based vaccines could be useful for colorectal cancer prevention in humans.

MUC-1 (CD-227) is an epithelial membrane glycoprotein that is expressed in ∼70% of colorectal cancers and correlates with poor prognosis (41). Immunization with a MUC1-specific TAA vaccine prevented the growth of MUC1 overexpressing MC38 tumor cell xenografts in MUC1 transgenic mice. In these studies, MUC1-specific vaccination stimulated interferon-γ producing CD4+ Th1 cells and CD8+ CTLs and generated strong immune responses against MUC1-expressing cancer cells without damaging normal tissues. It also induced a strong humoral memory response (42, 43). Another study in the MUC1+IL10−/− mouse model of inflammatory bowel disease-associated colorectal cancer (IBD-colorectal cancer) that overexpresses abnormal hypoglycosylated MUC1 showed that vaccination against MUC1 prevented dysplasia, IBD, and development of colitis-associated colon cancer by increasing mucosal immunity (44). Increasing evidence further suggests that MUC1 participates in the maintenance, tumorigenicity, and metastasis of colorectal cancer stem cells (CCSC). The vaccination of BALB/c mice with CD133+ CCSCs expressing MUC1 significantly reduced the growth of CT26 colorectal cancer transplants by the targeted killing of CCSCs indicated by the decrease in cells expressing CD133 and ALDH stem cell markers in these tumors. It also increased NK cell cytotoxicity and promoted the release of IFNγ, Perforin, and Granzyme B (45). Importantly, MUC1 peptide vaccination significantly boosted the T-cell immune response in human patients with a history of advanced colon adenoma and elicited long-term memory important for cancer prevention (46).

Another example of a TAA that has been tested in mouse models is squamous cell carcinoma recognized by T-cell-3 (SART3). SART3 is overexpressed in ∼70% of colorectal cancers and has multiple roles in gene regulation (47), cancer immunology (48, 49), and the maintenance of stem cell pluripotency (50). Vaccination with SART3-derived peptides was shown to induce cytotoxic T-cell antitumor immune responses (51) in human colorectal cancer patients by binding to various MHC haplotypes (20, 49, 52). Vaccination of BALB/c mice with a SART3 DNA expression vector in combination with CD40 L and GM-CSF expression vectors (to stimulate DC responses and enhance vaccination) on a polyplex micelle platform stimulated CTL and NK activities and inhibited the growth and metastasis of CT26 colorectal cancer xenotransplants (53).

Survivin, an inhibitor of apoptosis protein (IAP), is overexpressed in many cancers, including colorectal cancers, and its expression shows a strong correlation with TP53 mutation (54). Vaccination with a long peptide vaccine (SurVaxM) that spans amino acids 53 through 67 of the human survivin protein sequence led to survivin-specific CD8-mediated tumor cell death and increased the OS of C57BL6 mice implanted with murine GL261 glioma cells or B16f1 melanoma cells (55). Vaccination with a long synthetic peptide (LSP)-based cancer vaccine targeting amino acids 17–34, 84–110, and 122–142 of the survivin coding region also significantly upregulated tumor infiltration of CD4+ and CD8+ T cells and inhibited the growth of CT26 colorectal cancer xenotransplants in BALB/c mice and prevented tumor relapse (56). These results suggest that targeting survivin could be another approach for immunization against colorectal cancer.

Recent technological advances have made it possible to define the taxonomic profiles of the human gut bacteria in great detail and perform investigations into their roles in the etiology and development of colorectal cancer. Large-scale population studies have defined “healthy microbiota” and unhealthy colorectal cancer–associated microbiota profiles and manipulation of the gut microbiota has been evaluated in cancer prevention and treatment (57, 58). Colorectal cancer was shown to be associated with alterations in microbiota composition (dysbiosis) and specific bacteria species were found to play causal roles in the pathogenesis of colorectal cancer (59, 60). Bacteria can directly promote colorectal cancer via toxin-mediated mutagenesis, or indirectly through the opportunistic outgrowth of bacteria in the tumor microenvironment affecting the integrity of intestinal epithelia and the replication of intestinal cells. For example, Escherichia coli can secrete genotoxic DNA-damaging agents (61), whereas Fusobacterium nucleatum alters Toll-like receptor signaling and thus inhibit apoptosis (62). Other procarcinogenic effects are exerted by bacteria secreting metabolites that support the growth of cancer cells or interfere with the capability of the host immune system to attack cancer cells (63–65). In addition, recent studies have shown that the efficacy of both chemotherapy and immunotherapy depends on the gut microbiota, and approaches were developed and tested to achieve optimal treatment responses in patients with compromised gut microbiota (66).

Current interventional strategies explore the modulation of the gut bacteria to restore the altered microbiota equilibria associated with higher colorectal cancer risk. These include the use of probiotics, prebiotics, or postbiotics, the elimination of specific pathogens with antibiotics and altering pathogenic microbiota via fecal microbiota transplantation (FMT; Table 2). Each of these approaches showed encouraging results in preventing colorectal cancer in animal models, some of which are outlined below.

Table 2.

Manipulation of microbiota in rodent colorectal cancer (CRC) models.

Animal strainTumor modelProbioticsTreatment outcomeReference
C57BL/10J IL10 knockout Lactobacillus salivarius Reduced prevalence of colon cancer and mucosal inflammatory activity 68  
68C57BL/6 Clostridioides difficile infection Akkermansia muciniphila Amelioration of CDI 70  
C57BL/6 Clostridioides difficile infection Phascolarctobacterium Prevention of Clostridioides difficile outgrowth 71  
C57BL/6 ApcMin Streptococcus thermophilus Reduced CRC 72  
C57BL/6 DMH Streptococcus thermophilus Reduced CRC 72  
Sprague–Dawley rats DMH Lactobacillus and Bifidobacteria Reduced inflammation, CRC 73  
C57BL/6 DMH Clostridium butyricum and Bacillus subtilis Reduced CRC 74  
C57BL/6 ApcMin Lactobacillus fermentum and Lactobacillus acidophilus Reduced CRC multiplicity 75  
C57BL/6J TS4-Cre/Apclox468 Lactobacillus acidophilus Reduced polyp number 76  
Animal strain Tumor model Prebiotics Treatment outcome Reference 
Sprague–Dawley rats AOM Xylooligosaccharides and fructooligosaccharides Reduced CRC 80  
C57BL/6 ApcMin Triterpenoid saponins and Bifidobacterium Reduced polyp number 81  
Fisher 344 rats DMH Galacto-oligosaccharides Reduced aberrant crypt foci 82  
Sprague–Dawley rats DMH Resistant starch Reduced aberrant crypt foci 83  
Fisher 344 rats AOM Inulin Reduced aberrant crypt foci/CRC 84  
Animal strain Tumor model Postbiotics Treatment outcome Reference 
C57BL/6 ApcMin/high-fat diet Butyrate produced by Clostridium butyricum Reduced CRC 91  
C57BL/6J TS4-Cre/Apclox468 Dietary fiber promoting SCFA production Reduced polyp number 92  
C57BL/6J Msh2−/−/ApcMin Microbial-derived butyrate Support CRC 97  
Animal strain Tumor model Antibiotics Treatment outcome Reference 
Nu/Nu; Taconic Fusobacterium nucleatum-positive CRC xenografts Metronidazole Reduced CRC-Fusobacterium nucleatum load 100  
C57BL/6J ApcΔ716 enterotoxigenic Bacteroides fragilis induced CRC Cefoxitin Reduced microadenomas 102  
BALB/c AOM/DSS Metronidazole/neomycin/ciprofloxacin /vancomycin Reduced CRC 103  
C57BL/6 AOM/DSS Streptomycin/metronidazole/vancomycin Reduced CRC 104  
C57BL/6 AOM/DSS Ampicillin/neomycin/metronidazole/vancomycin Reduced CRC 105  
Animal strain Tumor model FMT Treatment outcome Reference 
C57BL/6 AOM/DSS Human stool from CRC survivors that consumed rice bran daily Reduced CRC 107  
C57BL/6J ApcMin Human stool from YYFZBJS herbal medicine consumers Reduced CRC 108  
C57BL/6 AOM/DSS Stool from Mus musculus domesticus Reduced CRC 109  
Animal strainTumor modelProbioticsTreatment outcomeReference
C57BL/10J IL10 knockout Lactobacillus salivarius Reduced prevalence of colon cancer and mucosal inflammatory activity 68  
68C57BL/6 Clostridioides difficile infection Akkermansia muciniphila Amelioration of CDI 70  
C57BL/6 Clostridioides difficile infection Phascolarctobacterium Prevention of Clostridioides difficile outgrowth 71  
C57BL/6 ApcMin Streptococcus thermophilus Reduced CRC 72  
C57BL/6 DMH Streptococcus thermophilus Reduced CRC 72  
Sprague–Dawley rats DMH Lactobacillus and Bifidobacteria Reduced inflammation, CRC 73  
C57BL/6 DMH Clostridium butyricum and Bacillus subtilis Reduced CRC 74  
C57BL/6 ApcMin Lactobacillus fermentum and Lactobacillus acidophilus Reduced CRC multiplicity 75  
C57BL/6J TS4-Cre/Apclox468 Lactobacillus acidophilus Reduced polyp number 76  
Animal strain Tumor model Prebiotics Treatment outcome Reference 
Sprague–Dawley rats AOM Xylooligosaccharides and fructooligosaccharides Reduced CRC 80  
C57BL/6 ApcMin Triterpenoid saponins and Bifidobacterium Reduced polyp number 81  
Fisher 344 rats DMH Galacto-oligosaccharides Reduced aberrant crypt foci 82  
Sprague–Dawley rats DMH Resistant starch Reduced aberrant crypt foci 83  
Fisher 344 rats AOM Inulin Reduced aberrant crypt foci/CRC 84  
Animal strain Tumor model Postbiotics Treatment outcome Reference 
C57BL/6 ApcMin/high-fat diet Butyrate produced by Clostridium butyricum Reduced CRC 91  
C57BL/6J TS4-Cre/Apclox468 Dietary fiber promoting SCFA production Reduced polyp number 92  
C57BL/6J Msh2−/−/ApcMin Microbial-derived butyrate Support CRC 97  
Animal strain Tumor model Antibiotics Treatment outcome Reference 
Nu/Nu; Taconic Fusobacterium nucleatum-positive CRC xenografts Metronidazole Reduced CRC-Fusobacterium nucleatum load 100  
C57BL/6J ApcΔ716 enterotoxigenic Bacteroides fragilis induced CRC Cefoxitin Reduced microadenomas 102  
BALB/c AOM/DSS Metronidazole/neomycin/ciprofloxacin /vancomycin Reduced CRC 103  
C57BL/6 AOM/DSS Streptomycin/metronidazole/vancomycin Reduced CRC 104  
C57BL/6 AOM/DSS Ampicillin/neomycin/metronidazole/vancomycin Reduced CRC 105  
Animal strain Tumor model FMT Treatment outcome Reference 
C57BL/6 AOM/DSS Human stool from CRC survivors that consumed rice bran daily Reduced CRC 107  
C57BL/6J ApcMin Human stool from YYFZBJS herbal medicine consumers Reduced CRC 108  
C57BL/6 AOM/DSS Stool from Mus musculus domesticus Reduced CRC 109  

Probiotics are live microorganisms that can be administered in specific amounts to provide a health benefit (67). They are widely used as food supplements, and their functions span from directly affecting the microbiota to modulating inflammation and thus reducing colorectal cancer risk. Indeed, feeding the probiotic Lactobacillus salivarius reduced enterocolitis and intestinal tumor occurrence in the C57BL/10J-IL10 mouse model of colitis (68). Importantly, probiotics can help eliminate Clostridium difficile infections (CDI) that are frequently enriched after the use of antibiotics and are associated with inflammation and colon tumorigenesis (69). For example, in C57BL/6 mice with CDI, the administration of Akkermansia muciniphilia had a protective effect by limiting CDI-associated intestinal barrier damage and inflammation (70). The outgrowth of Clostridium difficile could also be prevented in C57BL/6 mice with CDI by reducing feed succinate, a metabolite that fuels the growth of Clostridium difficile. This was achieved by the colonization of Phascolarctobacterium, a probiotic bacteria consuming succinate (71). Recently, it was shown that Streptococcus thermophilus reduces intestinal cancer development by inducing β-galactosidase and by inactivating the oncogenic Hippo pathway in both ApcMin and azoxymethane (AOM)-injected mice (72). Interestingly, a cocktail of Lactobacillus and Bifidobacteria was shown to reduce colorectal cancer incidence in dimethylhydrazine (DMH)-treated Sprague–Dawley rats by enhancing Toll-like receptor 2 signaling, which improved epithelial barrier integrity in the colonic mucosa and reduced the incidence of aberrant crypt foci (73). Other probiotic mixtures of bacteria have inhibited intestinal tumorigenesis in chemically induced or in ApcMin mice (74, 75). In addition, genetic modification of Lactobacillus acidophilus that eliminated its ability to secrete lipoteichoic acid reduced the expression of proinflammatory molecules and the incidence of colon adenomas in TS4-Cre/Apclox468 mice with colon-specific Apc deletion (76).

Prebiotics are defined as nondigestible food ingredients and belong mainly to a limited number of carbohydrate compounds that include fructo-oligosaccharides (FOS), inulin, galacto-oligosaccharides (GOS), and lactulose (77). They represent an alternate means to stimulate the growth of beneficial bacteria and their production of metabolites through fermentation in the colon. In addition, they can exert direct effects on the intestinal mucosa, modulate the immune system (78) and prevent infectious colonization (79). Recent studies indicate a protective effect of certain prebiotics (xylooligosaccharides/fructooligosaccharides) against colorectal cancer development in AOM-induced Sprague–Dawley rats (80). Another study showed the beneficial effects of prebiotics in reducing adenoma incidence in ApcMin mice through the interaction between prebiotic triterpenoid saponins and Bifidobacterium (81). Studies in DMH-induced Fisher 344 or Sprague–Dawley rats reported that the addition of prebiotics, such as dietary galacto-oligosaccharides (82) and resistant starch (83), reduced the numbers of aberrant crypt foci. Inulin is a galacto-oligosaccharide for which conflicting results have been reported. For example, although the addition of inulin to the food protected Fisher 344 rats against AOM-induced colorectal cancer (84), another in vitro study in Caco-2 colorectal cancer cells showed a protumorigenic effect as inulin increased the production of genotoxic colibactin by pks+ Escherichia coli in Caco-2 colorectal cancer cells (85). Based on these and other animal studies, ongoing trials in large human cohorts are exploring the potential preventive effects of prebiotics, but conclusive results are not yet available (86, 87).

Postbiotics are products of bacterial fermentation including short-chain fatty acids (SCFA), extracellular polysaccharides, and others. In colorectal cancer, the production of SCFAs such as butyrate, acetate, and propionate appears to have protective effects (88). Butyrate has been extensively studied due to the so-called butyrate paradox, which lies in its ability to safely promote healthy colonocyte proliferation in the mucosa, but at the same time to induce apoptosis in colorectal cancer cells. This effect was initially demonstrated in vitro by the ability of butyrate to inactivate multiple oncogenic pathways through histone hyperacetylation in HT-29 and HCT-116 colorectal cancer cell lines (89, 90). Subsequent in vivo studies in ApcMin mice fed a high-fat diet reported that butyrate, produced by Clostridium butyricum, protected against intestinal tumor formation by suppressing Wnt/β-catenin signaling, which decreased cell proliferation and increased apoptosis (91). Another study in TS4Cre/APClox468 conditional knockout mice showed that food enriched in dietary fiber promoted the growth of butyrate-producing bacteria and reduced polyp numbers (92). The role of butyrate in modulating pro- and anti-inflammatory cytokines and colonic Treg cell differentiation was observed in multiple murine studies (93, 94) and provided the rationale for the initiation of several clinical trials to test its ability to ameliorate ulcerative colitis, an inflammatory disease with increased risk to colorectal cancer (95, 96). Interestingly, a study using MMR-d Msh2−/−/ApcMin/+ mice demonstrated that microbial-derived butyrate can support rather than inhibit colorectal cancer cell proliferation (97), which raises questions as to whether somatic or germline mutations of the host affect the impact of postbiotics on the prevention of colorectal cancer.

Antibiotics are helpful in preventing colorectal cancer when used selectively, such as in the elimination of Fusobacterium nucleatum, a pathogen highly enriched at the adenoma stage in the colon (98, 99). Metronidazole treatment of mice transplanted with human Fusobacterium-positive colorectal cancer xenografts lowered the load of Fusobacterium and reduced cancer cell proliferation and tumor growth (100). In another example, the administration of cefoxitin (a broad-spectrum cephalosporin) to ApcΔ716 mice led to a complete clearance of previously inoculated enterotoxigenic Bacteroides fragilis, known to be associated with IBD-colorectal cancers (101) and reduced formation of intestinal microadenomas (102). Other studies used antibiotic cocktails rather than specific antibiotics, to assess their ability to prevent IBD-colorectal cancer tumorigenesis. Studies in AOM/dextran sodium sulfate (DSS)-treated BALB/c (103) and C57BL/6 mice (104, 105) showed that different combinations of antibiotics could suppress colitis-associated colorectal cancer.

Fecal microbiota transplantation (FMT) uses the transplantation of fecal matter from healthy donors to patients with intestinal pathologies (106). Several studies explored the effect of FMT from human donors on colorectal cancer development in mouse models. For example, one study showed that FMT of human stool from colorectal cancer survivors that consumed rice bran daily protected C57BL/6 mice from AOM/DSS-induced colon carcinogenesis (107). In another example of FMT in colorectal cancer prevention, FMT from volunteers who regularly used the YYFZBJS herbal medicine protected ApcMin mice from intestinal tumorigenesis (108). Similarly, restoration of a “healthy microbiota composition” in C57BL/6 mice by FMT with microbiota from wild mice (Mus musculus domesticus) reduced inflammation and improved resistance against AOM/DSS-induced colorectal tumorigenesis (109).

These and many other studies in rodent models suggest that approaches involving prevaccination with tumor antigens or manipulation of intestinal microbiota may hold promise for the prevention of colorectal cancer in patients with familial syndromes or in patients at increased risk, such as those with inflammatory bowel disease. However, each of these approaches still need to be carefully tested in human patients to alleviate safety concerns and assess efficacy. For vaccination approaches, initial clinical trials found that three MMR-d FSP neoantigens (AIM2, HT001, and TAF1B, exhibiting one-base pair deletions at their respective coding microsatellites) are well tolerated and induced T-cell and humoral immune responses (110). The FSP-based vaccine Nous209 (encoding 209 different FSP neoantigens shared by both sporadic and hereditary MSI-positive tumors) showed broad induction of FSP-specific CD8 and CD4 T-cell responses in C56F1 mice (111). Nous209 is currently being tested in early clinical trials for the treatment of MSI-H solid tumors in combination with pembrolizumab and cancer prevention in Lynch syndrome carriers. Vaccination against TSAs such as KRAS (G12D) or mutations in other tumor driver genes that commonly occur might also be useful in colorectal cancer prevention. For example, substitution at R248Q residue is the most frequent TP53 alteration in human colorectal cancer. A recent study in humanized TP53R248Qfloxmice showed that conditional inactivation of the mutp53R248Q allele strongly reduced AOM/DSS-induced colon carcinogenesis, which suggests that vaccination against the TP53R248Q may protect against colorectal cancers expressing this TSA (112), which could be tested in mouse models.

Although studies in rodents yielded promising results suggesting the beneficial effects of probiotics, prebiotics, and postbiotics, concerns remain about their safety in the clinical setting, especially in patients with underlying medical conditions such as compromised immunity (113). Issues regarding optimal strain usage, dosage, and formulation will need to be addressed. In this regard, the beneficial use of antibiotics in colorectal cancer prevention has to be balanced with the potential risks it poses as meta-analyses in human patients found conflicting associations with both delayed and accelerated colon tumorigenesis (114). Although these studies do not directly prove a causal role for antibiotics in colon carcinogenesis, they highlight important questions regarding the safe use of these medications, as they can also induce the colonization of colorectal cancer–associated pathogens (115). Not surprisingly, the use of FMT also raises concerns as it may have the potential of transmitting multidrug-resistant bacteria and viruses or causing other host infections (116). Therefore, the manipulation of microbiota in the clinical setting is still under scientific scrutiny, and providing clinical benefit might be highly dependent on several factors, including the microbiota diversity and susceptibility to specific gut microbiota, preexisting genetic alterations, and individual immune responses in patients. It is likely that colorectal cancer prevention approaches involving the gut microbiota need to be tailored around individual patient needs, and animal models will continue to provide valuable tools to test these parameters in a controlled manner.

No disclosures were reported.

This work was supported by NIH grants (CA248536 and CA222358 and Cancer Center grant CA13330) and a Feinberg Family Donation to W. Edelmann.

1.
Dekker
E
,
Tanis
P
,
Vleugels
JA
,
Kasi
PM
,
Wallace
MB
.
Colorectal cancer
.
Lancet
2019
;
394
:
1467
80
.
2.
Czarnecka-Czapczyńska
M
,
Aebisher
D
,
Oleś
P
,
Sosna
B
,
Krupka-Olek
M
,
Dynarowicz
K
, et al
.
The role of photodynamic therapy in breast cancer: a review of in vitro research
.
Biomed Pharmacother
2021
;
144
:
112342
.
3.
Sinicrope
FA
.
Lynch syndrome–associated colorectal cancer
.
N Engl J Med
2018
;
379
:
764
73
.
4.
Stoffel
EM
,
Koeppe
E
,
Everett
J
,
Ulintz
P
,
Kiel
M
,
Osborne
J
, et al
.
Germline genetic features of young individuals with colorectal cancer
.
Gastroenterology
2018
;
154
:
897
905
.
5.
Jasperson
KW
,
Tuohy
TM
,
Neklason
DW
,
Burt
RW
.
Hereditary and familial colon cancer
.
Gastroenterology
2010
;
138
:
2044
58
.
6.
Yang
K
,
Yang
W
,
Mariadason
J
,
Velcich
A
,
Lipkin
M
,
Augenlicht
L
.
Dietary components modify gene expression: implications for carcinogenesis
.
J Nutr
2005
;
135
:
2710
4
.
7.
Johnson
CM
,
Wei
C
,
Ensor
JE
,
Smolenski
DJ
,
Amos
CI
,
Levin
B
, et al
.
Meta-analyses of colorectal cancer risk factors
.
Cancer Causes Control
2013
;
24
:
1207
22
.
8.
Waluga
M
,
Zorniak
M
,
Fichna
J
,
Kukla
M
,
Hartleb
M
.
Pharmacological and dietary factors in prevention of colorectal cancer
.
J Physiol Pharmacol
2018
;
69
. doi:
10.26402/jpp.2018.3.02
.
9.
Mathers
JC
,
Elliott
F
,
Macrae
F
,
Mecklin
J-P
,
Moslein
G
,
McRonald
FE
, et al
.
Cancer prevention with resistant starch in Lynch syndrome patients in the CAPP2-randomized placebo controlled trial: planned 10-year follow-up
.
Cancer Prev Res
2022
.;
15
:
623
34
.
10.
Hofseth
LJ
,
Hebert
JR
,
Chanda
A
,
Chen
H
,
Love
BL
,
Pena
MM
, et al
.
Early-onset colorectal cancer: initial clues and current views
.
Nat Rev Gastroenterol Hepatol
2020
;
17
:
352
64
.
11.
Hintze
KJ
,
Benninghoff
AD
,
Cho
CE
,
Ward
RE
.
Modeling the western diet for preclinical investigations
.
Adv Nutr
2018
;
9
:
263
71
.
12.
Siegel
RL
,
Miller
KD
,
Fedewa
SA
,
Ahnen
DJ
,
Meester
RG
,
Barzi
A
, et al
.
Colorectal cancer statistics, 2017
.
CA Cancer J Clin
2017
;
67
:
177
93
.
13.
Bürtin
F
,
Mullins
CS
,
Linnebacher
M
.
Mouse models of colorectal cancer: Past, present and future perspectives
.
World J Gastroenterol
2020
;
26
:
1394
.
14.
Taketo
MM
,
Edelmann
W
.
Mouse models of colon cancer
.
Gastroenterology
2009
;
136
:
780
98
.
15.
Li
J
,
Ma
X
,
Chakravarti
D
,
Shalapour
S
,
DePinho
RA
.
Genetic and biological hallmarks of colorectal cancer
.
Genes Dev
2021
;
35
:
787
820
.
16.
Newmark
HL
,
Yang
K
,
Kurihara
N
,
Fan
K
,
Augenlicht
LH
,
Lipkin
M
.
Western-style diet-induced colonic tumors and their modulation by calcium and vitamin D in C57Bl/6 mice: a preclinical model for human sporadic colon cancer
.
Carcinogenesis
2009
;
30
:
88
92
.
17.
Gebert
J
,
Gelincik
O
,
Oezcan-Wahlbrink
M
,
Marshall
JD
,
Hernandez-Sanchez
A
,
Urban
K
, et al
.
Recurrent frameshift neoantigen vaccine elicits protective immunity with reduced tumor burden and improved overall survival in a Lynch syndrome mouse model
.
Gastroenterology
2021
;
161
:
1288
302
.
18.
Reyes-Uribe
L
,
Wu
W
,
Gelincik
O
,
Bommi
PV
,
Francisco-Cruz
A
,
Solis
LM
, et al
.
Naproxen chemoprevention promotes immune activation in Lynch syndrome colorectal mucosa
.
Gut
2021
;
70
:
555
66
.
19.
Zhao
R
,
Coker
OO
,
Wu
J
,
Zhou
Y
,
Zhao
L
,
Nakatsu
G
, et al
.
Aspirin reduces colorectal tumor development in mice and gut microbes reduce its bioavailability and chemopreventive effects
.
Gastroenterology
2020
;
159
:
969
83
.
20.
Wagner
S
,
Mullins
CS
,
Linnebacher
M
.
Colorectal cancer vaccines: tumor-associated antigens vs neoantigens
.
World J Gastroenterol
2018
;
24
:
5418
.
21.
Jia
W
,
Zhang
T
,
Huang
H
,
Feng
H
,
Wang
S
,
Guo
Z
, et al
.
Colorectal cancer vaccines: the current scenario and future prospects
.
Front Immunol
2022
;
13
:
942235
.
22.
Azadi
A
,
Golchini
A
,
Delazar
S
,
Abarghooi Kahaki
F
,
Dehnavi
SM
,
Payandeh
Z
, et al
.
Recent advances on immune targeted therapy of colorectal cancer using bi-specific antibodies and therapeutic vaccines
.
Biol Proced Online
2021
;
23
:
1
13
.
23.
Schwitalle
Y
,
Kloor
M
,
Eiermann
S
,
Linnebacher
M
,
Kienle
P
,
Knaebel
HP
, et al
.
Immune response against frameshift-induced neopeptides in HNPCC patients and healthy HNPCC mutation carriers
.
Gastroenterology
2008
;
134
:
988
97
.
24.
Zhao
Y
,
Baldin
AV
,
Isayev
O
,
Werner
J
,
Zamyatnin
Jr
AA
,
Bazhin
AV
.
Cancer vaccines: antigen selection strategy
.
Vaccines
2021
;
9
:
85
.
25.
Yamano
T
,
Kubo
S
,
Fukumoto
M
,
Yano
A
,
Mawatari-Furukawa
Y
,
Okamura
H
, et al
.
Whole cell vaccination using immunogenic cell death by an oncolytic adenovirus is effective against a colorectal cancer model
.
Mol Ther Oncolytics
2016
;
3
:
16031
.
26.
Yarchoan
M
,
Huang
CY
,
Zhu
Q
,
Ferguson
AK
,
Durham
JN
,
Anders
RA
, et al
.
A phase 2 study of GVAX colon vaccine with cyclophosphamide and pembrolizumab in patients with mismatch repair proficient advanced colorectal cancer
.
Cancer Med
2020
;
9
:
1485
94
.
27.
Leong
ML
,
Hampl
J
,
Liu
W
,
Mathur
S
,
Bahjat
KS
,
Luckett
W
, et al
.
Impact of preexisting vector-specific immunity on vaccine potency: characterization of Listeria monocytogenes-specific humoral and cellular immunity in humans and modeling studies using recombinant vaccines in mice
.
Infect Immun
2009
;
77
:
3958
68
.
28.
Flickinger
Jr
JC
,
Staudt
RE
,
Singh
J
,
Carlson
RD
,
Barton
JR
,
Baybutt
TR
, et al
.
Chimeric adenoviral (Ad5. F35) and listeria vector prime-boost immunization is safe and effective for cancer immunotherapy
.
npj Vaccines
2022
;
7
:
61
.
29.
Carreno
BM
,
Magrini
V
,
Becker-Hapak
M
,
Kaabinejadian
S
,
Hundal
J
,
Petti
AA
, et al
.
A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells
.
Science
2015
;
348
:
803
8
.
30.
Ho
WW
,
Gomes-Santos
IL
,
Aoki
S
,
Datta
M
,
Kawaguchi
K
,
Talele
NP
, et al
.
Dendritic cell paucity in mismatch repair–proficient colorectal cancer liver metastases limits immune checkpoint blockade efficacy
.
Proc Natl Acad Sci
2021
;
118
:
e2105323118
.
31.
Santos
P
,
Almeida
F
.
Exosome-based vaccines: history, current state, and clinical trials
.
Front Immunol
2021
;
12
:
711565
.
32.
Tampa
M
,
Sarbu
M-I
,
Matei
C
,
Mitran
C-I
,
Mitran
M-I
,
Caruntu
C
, et al
.
Photodynamic therapy: a hot topic in dermato-oncology
.
Oncol Lett
2019
;
17
:
4085
93
.
33.
Linnebacher
M
,
Gebert
J
,
Rudy
W
,
Woerner
S
,
Yuan
YP
,
Bork
P
, et al
.
Frameshift peptide-derived T-cell epitopes: a source of novel tumor-specific antigens
.
Int J Cancer
2001
;
93
:
6
11
.
34.
Hernandez-Sanchez
A
,
Grossman
M
,
Yeung
K
,
Sei
SS
,
Lipkin
S
,
Kloor
M
.
Vaccines for immunoprevention of DNA mismatch repair deficient cancers
.
J Immunother Cancer
2022
;
10
:
e004416
.
35.
Majumder
S
,
Shah
R
,
Elias
J
,
Manoharan
M
,
Shah
P
,
Kumari
A
, et al
.
A cancer vaccine approach for personalized treatment of Lynch syndrome
.
Sci Rep
2018
;
8
:
1
14
.
36.
Sakamoto
K
,
Kamada
Y
,
Sameshima
T
,
Yaguchi
M
,
Niida
A
,
Sasaki
S
, et al
.
K-Ras (G12D)-selective inhibitory peptides generated by random peptide T7 phage display technology
.
Biochem Biophys Res Commun
2017
;
484
:
605
11
.
37.
Sakamoto
K
,
Lin
B
,
Nunomura
K
,
Izawa
T
,
Nakagawa
S
.
The K-Ras (G12D)-inhibitory peptide KS-58 suppresses growth of murine CT26 colorectal cancer cell-derived tumors
.
Sci Rep
2022
;
12
:
8121
.
38.
Saha
A
,
Bhattacharya-Chatterjee
M
,
Foon
KA
,
Celis
E
,
Chatterjee
SK
.
Stimulatory effects of CpG oligodeoxynucleotide on dendritic cell-based immunotherapy of colon cancer in CEA/HLA-A2 transgenic mice
.
Int J Cancer
2009
;
124
:
877
88
.
39.
Saha
A
,
Chatterjee
SK
,
Foon
KA
,
Celis
E
,
Bhattacharya-Chatterjee
M
.
Therapy of established tumors in a novel murine model transgenic for human carcinoembryonic antigen and HLA-A2 with a combination of anti-idiotype vaccine and CTL peptides of carcinoembryonic antigen
.
Cancer Res
2007
;
67
:
2881
92
.
40.
Hance
KW
,
Zeytin
HE
,
Greiner
JW
.
Mouse models expressing human carcinoembryonic antigen (CEA) as a transgene: evaluation of CEA-based cancer vaccines
.
Mutat Res
2005
;
576
:
132
54
.
41.
Niv
Y
.
MUC1 and colorectal cancer pathophysiology considerations
.
World J Gastroenterol
2008
;
14
:
2139
.
42.
Acres
B
,
Apostolopoulos
V
,
Balloul
J-M
,
Wreschner
D
,
Xing
P-X
,
Ali-Hadji
D
, et al
.
MUC1-specific immune responses in human MUC1 transgenic mice immunized with various human MUC1 vaccines
.
Cancer Immunol Immunother
2000
;
48
:
588
94
.
43.
Mukherjee
P
,
Pathangey
L
,
Bradley
J
,
Tinder
T
,
Basu
G
,
Akporiaye
E
, et al
.
MUC1-specific immune therapy generates a strong anti-tumor response in a MUC1-tolerant colon cancer model
.
Vaccine
2007
;
25
:
1607
18
.
44.
Beatty
PL
,
Narayanan
S
,
Gariépy
J
,
Ranganathan
S
,
Finn
OJ
.
Vaccine against MUC1 antigen expressed in inflammatory bowel disease and cancer lessens colonic inflammation and prevents progression to colitis-associated colon CancerMUC1 vaccine controls IBD and prevents colon cancer
.
Cancer Prev Res
2010
;
3
:
438
46
.
45.
Guo
M
,
Luo
B
,
Pan
M
,
Li
M
,
Xu
H
,
Zhao
F
, et al
.
Colorectal cancer stem cell vaccine with high expression of MUC1 serves as a novel prophylactic vaccine for colorectal cancer
.
Int Immunopharmacol
2020
;
88
:
106850
.
46.
Kimura
T
,
McKolanis
JR
,
Dzubinski
LA
,
Islam
K
,
Potter
DM
,
Salazar
AM
, et al
.
MUC1 vaccine for individuals with advanced adenoma of the colon: a cancer immunoprevention feasibility StudyMUC1 vaccine clinical trial for colon cancer prevention
.
Cancer Prev Res
2013
;
6
:
18
26
.
47.
Timani
KA
,
Liu
Y
,
Fan
Y
,
Mohammad
KS
,
He
JJ
.
Tip110 regulates the cross talk between p53 and hypoxia-inducible factor 1α under hypoxia and promotes survival of cancer cells
.
Mol Cell Biol
2015
;
35
:
2254
64
.
48.
Ito
M
,
Shichijo
S
,
Miyagi
Y
,
Kobayashi
T
,
Tsuda
N
,
Yamada
A
, et al
.
Identification of SART3-derived peptides capable of inducing HLA-A2-restricted and tumor-specific CTLs in cancer patients with different HLA-A2 subtypes
.
Int J Cancer
2000
;
88
:
633
9
.
49.
Miyagi
Y
,
Imai
N
,
Sasatomi
T
,
Yamada
A
,
Mine
T
,
Katagiri
K
, et al
.
Induction of cellular immune responses to tumor cells and peptides in colorectal cancer patients by vaccination with SART3 peptides
.
Clin Cancer Res
2001
;
7
:
3950
62
.
50.
Liu
Y
,
Timani
K
,
Ou
X
,
Broxmeyer
HE
,
He
JJ
.
C-MYC controlled TIP110 protein expression regulates OCT4 mRNA splicing in human embryonic stem cells
.
Stem Cells Dev
2013
;
22
:
689
94
.
51.
Sasatomi
T
,
Suefuji
Y
,
Matsunaga
K
,
Yamana
H
,
Miyagi
Y
,
Araki
Y
, et al
.
Expression of tumor rejection antigens in colorectal carcinomas
.
Cancer
2002
;
94
:
1636
41
.
52.
Minami
T
,
Matsueda
S
,
Takedatsu
H
,
Tanaka
M
,
Noguchi
M
,
Uemura
H
, et al
.
Identification of SART3-derived peptides having the potential to induce cancer-reactive cytotoxic T lymphocytes from prostate cancer patients with HLA-A3 supertype alleles
.
Cancer Immunol Immunother
2007
;
56
:
689
98
.
53.
Furugaki
K
,
Cui
L
,
Kunisawa
Y
,
Osada
K
,
Shinkai
K
,
Tanaka
M
, et al
.
Intraperitoneal administration of a tumor-associated antigen SART3, CD40L, and GM-CSF gene-loaded polyplex micelle elicits a vaccine effect in mouse tumor models
.
PLoS One
2014
;
9
:
e101854
.
54.
Tang
Q
,
Efe
G
,
Chiarella
AM
,
Leung
J
,
Chen
M
,
Yamazoe
T
, et al
.
Mutant p53 regulates Survivin to foster lung metastasis
.
Genes Dev
2021
;
35
:
528
41
.
55.
Fenstermaker
RA
,
Figel
SA
,
Qiu
J
,
Barone
TA
,
Dharma
SS
,
Winograd
EK
, et al
.
Survivin monoclonal antibodies detect survivin cell surface expression and inhibit tumor growth in VivoSurvivin mAbs inhibit tumor growth
.
Clin Cancer Res
2018
;
24
:
2642
52
.
56.
Onodi
F
,
Maherzi-Mechalikh
C
,
Mougel
A
,
Ben Hamouda
N
,
Taboas
C
,
Gueugnon
F
, et al
.
High therapeutic efficacy of a new survivin LSP-cancer vaccine containing CD4+ and CD8+ T-cell epitopes
.
Front Oncol
2018
;
8
:
517
.
57.
Fong
W
,
Li
Q
,
Yu
J
.
Gut microbiota modulation: a novel strategy for prevention and treatment of colorectal cancer
.
Oncogene
2020
;
39
:
4925
43
.
58.
Gill
SR
,
Pop
M
,
DeBoy
RT
,
Eckburg
PB
,
Turnbaugh
PJ
,
Samuel
BS
, et al
.
Metagenomic analysis of the human distal gut microbiome
.
Science
2006
;
312
:
1355
9
.
59.
Wong
SH
,
Yu
J
.
Gut microbiota in colorectal cancer: mechanisms of action and clinical applications
.
Nat Rev Gastroenterol Hepatol
2019
;
16
:
690
704
.
60.
Wirbel
J
,
Pyl
PT
,
Kartal
E
,
Zych
K
,
Kashani
A
,
Milanese
A
, et al
.
Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer
.
Nat Med
2019
;
25
:
679
89
.
61.
Arthur
JC
,
Perez-Chanona
E
,
Mühlbauer
M
,
Tomkovich
S
,
Uronis
JM
,
Fan
T-J
, et al
.
Intestinal inflammation targets cancer-inducing activity of the microbiota
.
Science
2012
;
338
:
120
3
.
62.
Yang
Y
,
Weng
W
,
Peng
J
,
Hong
L
,
Yang
L
,
Toiyama
Y
, et al
.
Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating toll-like receptor 4 signaling to nuclear factor− κB, and up-regulating expression of microRNA-21
.
Gastroenterology
2017
;
152
:
851
66
.
63.
Wu
S
,
Rhee
K-J
,
Albesiano
E
,
Rabizadeh
S
,
Wu
X
,
Yen
H-R
, et al
.
A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses
.
Nat Med
2009
;
15
:
1016
22
.
64.
Long
X
,
Wong
CC
,
Tong
L
,
Chu
ES
,
Ho Szeto
C
,
Go
MY
, et al
.
Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity
.
Nature microbiology
2019
;
4
:
2319
30
.
65.
Ternes
D
,
Tsenkova
M
,
Pozdeev
VI
,
Meyers
M
,
Koncina
E
,
Atatri
S
, et al
.
The gut microbial metabolite formate exacerbates colorectal cancer progression
.
Nature metabolism
2022
;
4
:
458
75
.
66.
Kroemer
G
,
Zitvogel
L
.
The breakthrough of the microbiota
.
Nat Rev Immunol
2018
;
18
:
87
8
.
67.
Hill
C
,
Guarner
F
,
Reid
G
,
Gibson
GR
,
Merenstein
DJ
,
Pot
B
, et al
.
Expert consensus document: the International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic
.
Nat Rev Gastroenterol Hepatol
2014
;
11
:
506
14
.
68.
O'mahony
L
,
Feeney
M
,
O'halloran
S
,
Murphy
L
,
Kiely
B
,
Fitzgibbon
J
, et al
.
Probiotic impact on microbial flora, inflammation and tumour development in IL-10 knockout mice
.
Aliment Pharmacol Ther
2001
;
15
:
1219
25
.
69.
Drewes
JL
,
Chen
J
,
Markham
NO
,
Knippel
RJ
,
Domingue
JC
,
Tam
AJ
, et al
.
Human colon cancer–derived Clostridioides difficile strains drive colonic tumorigenesis in mice
.
Cancer Discov
2022
;
12
:
1873
85
.
70.
Wu
Z
,
Xu
Q
,
Gu
S
,
Chen
Y
,
Lv
L
,
Zheng
B
, et al
.
Akkermansia muciniphila ameliorates clostridioides difficile infection in mice by modulating the intestinal microbiome and metabolites
.
Front Microbiol
2022
;
13
:
841920
.
71.
Nagao-Kitamoto
H
,
Leslie
JL
,
Kitamoto
S
,
Jin
C
,
Thomsson
KA
,
Gillilland
III
MG
, et al
.
Interleukin-22-mediated host glycosylation prevents Clostridioides difficile infection by modulating the metabolic activity of the gut microbiota
.
Nat Med
2020
;
26
:
608
17
.
72.
Li
Q
,
Hu
W
,
Liu
W-X
,
Zhao
L-Y
,
Huang
D
,
Liu
X-D
, et al
.
Streptococcus thermophilus inhibits colorectal tumorigenesis through secreting β-galactosidase
.
Gastroenterology
2021
;
160
:
1179
93
.
73.
Kuugbee
ED
,
Shang
X
,
Gamallat
Y
,
Bamba
D
,
Awadasseid
A
,
Suliman
MA
, et al
.
Structural change in microbiota by a probiotic cocktail enhances the gut barrier and reduces cancer via TLR2 signaling in a rat model of colon cancer
.
Dig Dis Sci
2016
;
61
:
2908
20
.
74.
Chen
Z-F
,
Ai
L-Y
,
Wang
J-L
,
Ren
L-L
,
Yu
Y-N
,
Xu
J
, et al
.
Probiotics clostridium butyricum and bacillus subtilis ameliorate intestinal tumorigenesis
.
Future microbiology
2015
;
10
:
1433
45
.
75.
Kahouli
I
,
Malhotra
M
,
Westfall
S
,
Alaoui-Jamali
MA
,
Prakash
S
.
Design and validation of an orally administrated active L. fermentum-L. acidophilus probiotic formulation using colorectal cancer Apc Min/+ mouse model
.
Appl Microbiol Biotechnol
2017
;
101
:
1999
2019
.
76.
Khazaie
K
,
Zadeh
M
,
Khan
MW
,
Bere
P
,
Gounari
F
,
Dennis
K
, et al
.
Abating colon cancer polyposis by Lactobacillus acidophilus deficient in lipoteichoic acid
.
Proc Natl Acad Sci
2012
;
109
:
10462
7
.
77.
Gibson
GR
,
Hutkins
R
,
Sanders
ME
,
Prescott
SL
,
Reimer
RA
,
Salminen
SJ
, et al
.
Expert consensus document: the International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics
.
Nat Rev Gastroenterol Hepatol
2017
;
14
:
491
502
.
78.
Ito
H
,
Takemura
N
,
Sonoyama
K
,
Kawagishi
H
,
Topping
DL
,
Conlon
MA
, et al
.
Degree of polymerization of inulin-type fructans differentially affects number of lactic acid bacteria, intestinal immune functions, and immunoglobulin A secretion in the rat cecum
.
J Agric Food Chem
2011
;
59
:
5771
8
.
79.
Shoaf
K
,
Mulvey
GL
,
Armstrong
GD
.,
Hutkins RW. Prebiotic galactooligosaccharides reduce adherence of enteropathogenic Escherichia coli to tissue culture cells
.
Infect Immun
2006
;
74
:
6920
8
.
80.
Hsu
C-K
,
Liao
J-W
,
Chung
Y-C
,
Hsieh
C-P
,
Chan
Y-C
.
Xylooligosaccharides and fructooligosaccharides affect the intestinal microbiota and precancerous colonic lesion development in rats
.
J Nutr
2004
;
134
:
1523
8
.
81.
Chen
L
,
Brar
MS
,
Leung
FC
,
Hsiao
WW
.
Triterpenoid herbal saponins enhance beneficial bacteria, decrease sulfate-reducing bacteria, modulate inflammatory intestinal microenvironment and exert cancer preventive effects in ApcMin/+ mice
.
Oncotarget
2016
;
7
:
31226
.
82.
Wijnands
M
,
Schoterman
H
,
Bruijntjes
J
,
Hollanders
V
,
Woutersen
R
.
Effect of dietary galacto-oligosaccharides on azoxymethane-induced aberrant crypt foci and colorectal cancer in Fischer 344 rats
.
Carcinogenesis
2001
;
22
:
127
32
.
83.
Bauer-Marinovic
M
,
Florian
S
,
Müller-Schmehl
K
,
Glatt
H
,
Jacobasch
G
.
Dietary resistant starch type 3 prevents tumor induction by 1, 2-dimethylhydrazine and alters proliferation, apoptosis and dedifferentiation in rat colon
.
Carcinogenesis
2006
;
27
:
1849
59
.
84.
Verghese
M
,
Rao
D
,
Chawan
C
,
Williams
LL
,
Shackelford
L
.
Dietary inulin suppresses azoxymethane-induced aberrant crypt foci and colon tumors at the promotion stage in young Fisher 344 rats
.
J Nutr
2002
;
132
:
2809
13
.
85.
Oliero
M
,
Calvé
A
,
Fragoso
G
,
Cuisiniere
T
,
Hajjar
R
,
Dobrindt
U
, et al
.
Oligosaccharides increase the genotoxic effect of colibactin produced by pks+ Escherichia coli strains
.
BMC Cancer
2021
;
21
:
1
10
.
86.
Rafter
J
,
Bennett
M
,
Caderni
G
,
Clune
Y
,
Hughes
R
,
Karlsson
PC
, et al
.
Dietary synbiotics reduce cancer risk factors in polypectomized and colon cancer patients
.
Am J Clin Nutr
2007
;
85
:
488
96
.
87.
Ishikawa
H
,
Akedo
I
,
Otani
T
,
Suzuki
T
,
Nakamura
T
,
Takeyama
I
, et al
.
Randomized trial of dietary fiber and Lactobacillus casei administration for prevention of colorectal tumors
.
Int J Cancer
2005
;
116
:
762
7
.
88.
Scharlau
D
,
Borowicki
A
,
Habermann
N
,
Hofmann
T
,
Klenow
S
,
Miene
C
, et al
.
Mechanisms of primary cancer prevention by butyrate and other products formed during gut flora-mediated fermentation of dietary fibre
.
Mutat Res
2009
;
682
:
39
53
.
89.
Hinnebusch
BF
,
Meng
S
,
Wu
JT
,
Archer
SY
,
Hodin
RA
.
The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation
.
J Nutr
2002
;
132
:
1012
7
.
90.
Donohoe
DR
,
Collins
LB
,
Wali
A
,
Bigler
R
,
Sun
W
,
Bultman
SJ
.
The Warburg effect dictates the mechanism of butyrate-mediated histone acetylation and cell proliferation
.
Mol Cell
2012
;
48
:
612
26
.
91.
Chen
D
,
Jin
D
,
Huang
S
,
Wu
J
,
Xu
M
,
Liu
T
, et al
.
Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota
.
Cancer Lett
2020
;
469
:
456
67
.
92.
Bishehsari
F
,
Engen
PA
,
Preite
NZ
,
Tuncil
YE
,
Naqib
A
,
Shaikh
M
, et al
.
Dietary fiber treatment corrects the composition of gut microbiota, promotes SCFA production, and suppresses colon carcinogenesis
.
Genes
2018
;
9
:
102
.
93.
Furusawa
Y
,
Obata
Y
,
Fukuda
S
,
Endo
TA
,
Nakato
G
,
Takahashi
D
, et al
.
Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells
.
Nature
2013
;
504
:
446
50
.
94.
Smith
PM
,
Howitt
MR
,
Panikov
N
,
Michaud
M
,
Gallini
CA
,
Bohlooly-y
M
, et al
.
The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis
.
Science
2013
;
341
:
569
73
.
95.
Hodgkinson
K
,
El Abbar
F
,
Dobranowski
P
,
Manoogian
J
,
Butcher
J
,
Figeys
D
, et al
.
Butyrate's role in human health and the current progress towards its clinical application to treat gastrointestinal disease
.
Clin Nutr
2022
;
42
:
61
75
.
96.
Mulita
F
,
Iliopoulos
F
,
Liolis
E
,
Tchabashvili
L
,
Drakos
N
,
Tsilivigkos
C
, et al
.
Ulcerative colitis: the role of elective and emergency surgery
.
Albanian J Truma Emerg Surg.
2021
;
5
:
829
33
.
97.
Belcheva
A
,
Irrazabal
T
,
Robertson
SJ
,
Streutker
C
,
Maughan
H
,
Rubino
S
, et al
.
Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells
.
Cell
2014
;
158
:
288
99
.
98.
Castellarin
M
,
Warren
RL
,
Freeman
JD
,
Dreolini
L
,
Krzywinski
M
,
Strauss
J
, et al
.
Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma
.
Genome Res
2012
;
22
:
299
306
.
99.
Kostic
AD
,
Gevers
D
,
Pedamallu
CS
,
Michaud
M
,
Duke
F
,
Earl
AM
, et al
.
Genomic analysis identifies association of Fusobacterium with colorectal carcinoma
.
Genome Res
2012
;
22
:
292
8
.
100.
Bullman
S
,
Pedamallu
CS
,
Sicinska
E
,
Clancy
TE
,
Zhang
X
,
Cai
D
, et al
.
Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer
.
Science
2017
;
358
:
1443
8
.
101.
Ulger Toprak
N
,
Yagci
A
,
Gulluoglu
B
,
Akin
M
,
Demirkalem
P
,
Celenk
T
, et al
.
A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer
.
Clin Microbiol Infect
2006
;
12
:
782
6
.
102.
DeStefano Shields
CE
,
Van Meerbeke
SW
,
Housseau
F
,
Wang
H
,
Huso
DL
,
Casero
Jr
RA
, et al
.
Reduction of murine colon tumorigenesis driven by enterotoxigenic Bacteroides fragilis using cefoxitin treatment
.
J Infect Dis
2016
;
214
:
122
9
.
103.
Hattori
N
,
Niwa
T
,
Ishida
T
,
Kobayashi
K
,
Imai
T
,
Mori
A
, et al
.
Antibiotics suppress colon tumorigenesis through inhibition of aberrant DNA methylation in an azoxymethane and dextran sulfate sodium colitis model
.
Cancer Sci
2019
;
110
:
147
56
.
104.
Zackular
JP
,
Baxter
NT
,
Chen
GY
,
Schloss
PD
.
Manipulation of the gut microbiota reveals role in colon tumorigenesis
.
mSphere
2016
;
1
:
e00001
15
.
105.
Lee
JG
,
Eun
CS
,
Jo
SV
,
Lee
A-r
Park
CH
,
Han
DS
.
The impact of gut microbiota manipulation with antibiotics on colon tumorigenesis in a murine model
.
PLoS One
2019
;
14
:
e0226907
.
106.
Smits
LP
,
Bouter
KE
,
De Vos
WM
,
Borody
TJ
,
Nieuwdorp
M
.
Therapeutic potential of fecal microbiota transplantation
.
Gastroenterology
2013
;
145
:
946
53
.
107.
Parker
KD
,
Maurya
AK
,
Ibrahim
H
,
Rao
S
,
Hove
PR
,
Kumar
D
, et al
.
Dietary rice bran-modified human gut microbial consortia confers protection against colon carcinogenesis following fecal transfaunation
.
Biomedicines
2021
;
9
:
144
.
108.
Sui
H
,
Zhang
L
,
Gu
K
,
Chai
N
,
Ji
Q
,
Zhou
L
, et al
.
YYFZBJS ameliorates colorectal cancer progression in ApcMin/+ mice by remodeling gut microbiota and inhibiting regulatory T-cell generation
.
Cell Commun Signal
2020
;
18
:
1
17
.
109.
Rosshart
SP
,
Vassallo
BG
,
Angeletti
D
,
Hutchinson
DS
,
Morgan
AP
,
Takeda
K
, et al
.
Wild mouse gut microbiota promotes host fitness and improves disease resistance
.
Cell
2017
;
171
:
1015
28
.
110.
Kloor
M
,
Reuschenbach
M
,
Pauligk
C
,
Karbach
J
,
Rafiyan
M-R
,
Al-Batran
S-E
, et al
.
A frameshift peptide neoantigen-based vaccine for mismatch repair-deficient cancers: a phase I/IIa clinical TrialFrameshift peptide vaccine for MSI cancer
.
Clin Cancer Res
2020
;
26
:
4503
10
.
111.
Leoni
G
,
D'Alise
AM
,
Cotugno
G
,
Langone
F
,
Garzia
I
,
De Lucia
M
, et al
.
A genetic vaccine encoding shared cancer neoantigens to treat tumors with microsatellite instability: a vaccine for MSI tumors
.
Cancer Res
2020
;
80
:
3972
82
.
112.
Schulz-Heddergott
R
,
Stark
N
,
Edmunds
SJ
,
Li
J
,
Conradi
L-C
,
Bohnenberger
H
, et al
.
Therapeutic ablation of gain-of-function mutant p53 in colorectal cancer inhibits Stat3-mediated tumor growth and invasion
.
Cancer Cell
2018
;
34
:
298
314
.
113.
Suez
J
,
Zmora
N
,
Segal
E
,
Elinav
E
.
The pros, cons, and many unknowns of probiotics
.
Nat Med
2019
;
25
:
716
29
.
114.
McDowell
R
,
Perrott
S
,
Murchie
P
,
Cardwell
C
,
Hughes
C
,
Samuel
L
.
Oral antibiotic use and early-onset colorectal cancer: findings from a case-control study using a national clinical database
.
Br J Cancer
2022
;
126
:
957
67
.
115.
Willing
BP
,
Russell
SL
,
Finlay
BB
.
Shifting the balance: antibiotic effects on host–microbiota mutualism
.
Nat Rev Microbiol
2011
;
9
:
233
43
.
116.
Food
U
.
Drug administration: important safety alert regarding use of fecal microbiota for transplantation and risk of serious adverse reactions due to transmission of multidrug resistant organisms
.
June 13, 2019
.
Washington, DC
.
2019
; https://www.fda.gov/media/86440/download.