Skip to main content

MINI REVIEW article

Front. Pharmacol., 20 October 2021
Sec. Gastrointestinal and Hepatic Pharmacology
This article is part of the Research Topic Colorectal Cancer: Targets, Treatment and Prevention View all 11 articles

Inflammatory Bowel Disease and Risk of Colorectal Cancer: An Overview From Pathophysiology to Pharmacological Prevention

  • 1Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
  • 2Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
  • 3Department of Life Sciences, University of Trieste, Trieste, Italy

Increased risk of colorectal cancer (CRC) in inflammatory bowel disease (IBD) patients has been attributed to long-standing chronic inflammation, with the contribution of genetic alterations and environmental factors such as the microbiota. Moreover, accumulating data indicate that IBD-associated CRC (IBD-CRC) may initiate and develop through a pathway of tumorigenesis distinct from that of sporadic CRC. This mini-review summarizes the current knowledge of IBD-CRC, focusing on the main mechanisms underlying its pathogenesis, and on the important role of immunomodulators and biologics used to treat IBD patients in interfering with the inflammatory process involved in carcinogenesis.

Introduction

Inflammatory bowel disease (IBD), comprising ulcerative colitis (UC) and Crohn’s disease (CD), is a chronic relapsing inflammatory disorder associated with an increased risk of colorectal cancer (CRC) compared to the general population (Eaden et al., 2001; Bajpai et al., 2019). Interestingly, incidence rates of IBD-associated CRC (IBD-CRC) decreased over the last decades (Castaño-Milla et al., 2014; Choi et al., 2015). Several risk factors have been described, such as disease duration and extension (Ekbom et al., 1990; Eaden et al., 2001), inflammation (Rutter et al., 2004) and primary sclerosing cholangitis (Soetikno et al., 2002). Despite the limited number of studies, younger age at onset and onset of IBD in childhood seem to be associated with an increased incidence of CRC (Jess et al., 2012; Olén et al., 2020). Unlike sporadic CRC (sCRC), in patients with IBD-CRC, long-standing chronic inflammation initiates and drives tumorigenesis and important elucidation of the multiple factors involved in the process of carcinogenesis are emerging (Ullman and Itzkowitz, 2011; Baker et al., 2018). The present mini-review summarizes the recent advances in the pathophysiology of IBD-CRC, including the role of the immunomodulators currently used in the treatment of IBD (Figure 1).

FIGURE 1
www.frontiersin.org

FIGURE 1. From IBD to CRC: possible key players in carcinogenesis within intestinal mucosa. Intestinal epithelial cells of IBD-CRC patients present a different frequency of somatic mutations, altered DNA methylation sites and deregulated microRNA in comparison to sCRC. The anti-apoptotic role of NF-κB in cancer cells enhanced their proliferation, promoting tumor progression. Moreover, an exacerbated activation of NF-κB `and STAT3 signaling, via IL6, drives M2 polarization and Th17 differentiation favouring IBD-CRC carcinogenesis. Enterobacteriaceae strains are responsible for the production of genotoxic compounds and for the reduction of intestinal levels of anti-inflammatory SCFA; Streptococcus genus could exert its tumorigenic activity through the recruitment of tumor-infiltrating myeloid cells (S. gallolyticus) and the release of chemokines and prostaglandins (S. bovis). The image was created with BioRender.com.

Pathogenesis of CRC in IBD Patients: Genetic and Epigenetic Bases

CRC tumorigenesis needs a tumor-initiating event that modifies normal intestinal epithelial cells by spontaneous mutation, environmental mutagens or inflammation-induced genetic and epigenetic alterations (Schmitt and Greten, 2021). Several lines of evidence suggest that IBD-CRC can result from a different mechanism of tumorigenesis in comparison to sCRC (Salk et al., 2009; Risques et al., 2011; Baker et al., 2019). The epithelial tumor tissue of IBD-CRC patients presents a lower frequency of somatic adenomatous polyposis coli (APC) and Kirsten rat sarcoma virus (KRAS) mutations, while tumor protein P53 (TP53) mutations and Myc proto-oncogene protein (MYC) amplifications are detected earlier during tumor progression in comparison to sCRC (Yaeger et al., 2016; Du et al., 2017; Chatila et al., 2020). Insights on germline alterations of IBD patients with CRC were provided for 25 patients with IBD-CRC, sequencing 39 genes potentially involved in predisposition to cancer (Biscaglia et al., 2021). Six patients (24%) presented pathogenic variants (International Agency for Research on Cancer, IARC class 4 or 5). Of these, four concerned the APC region, three the MLH1 gene, and the remaining ones the MSH2, MSH3, MUTYH, EPCAM, BRCA1, CHEK2, POLD1, POLE, CDKN2A and PDGFRA genes. The onset of CRC was significantly earlier in patients carrying these variants than in patients with benign or unidentified variants.

DNA and RNA-sequencing and methylation analysis were performed in 2500 CRC cases, including 31 IBD-CRC (Rajamäki et al., 2021). As expected, somatic mutations in APC and KRAS were less frequent in IBD-CRC; a significant enrichment of somatic mutations at noncoding 5′UTR of TP53 in IBD-CRC, resulting in low TP53 expression, was found. Aberrant promoter methylation patterns were detected exclusively in IBD-CRC in two genes related to mucosal immunity, in particular hypermethylation of polymeric immunoglobulin receptor (PIGR) and hypomethylation and strong overexpression of oncostatin M receptor (OSMR). Interestingly, increased levels of the interleukin-6 (IL-6) family member OSM and its receptor have been already detected in patients with active IBD and their presence was associated with failure of anti-tumor necrosis factor a (TNF) therapy (West et al., 2017), suggesting a potential role of the OSMR signaling in the molecular mechanism of IBD-associated tumorigenesis.

Chronic inflammation promotes aberrant DNA methylation in IBD, which in turn may predispose to the development of cancer (Barnicle et al., 2017; Somineni et al., 2019). A progressive increase in the percentage of methylated genes in the WNT signaling pathway from normal colon samples (n = 24) to IBD (n = 25) to IBD-CRC (n = 16) was observed, indicating their potential involvement during cancer development (Dhir et al., 2008). In particular, methylation of APC1A, APC2, SFRP1, and SFRP2 genes characterized the progression from IBD to IBD-CRC, indicating their potential role as biomarkers for early detection of cancer in IBD patients. These results have recently been validated in an additional cohort of UC CRC (Beggs et al., 2019).

The methylation status of 10 candidate genes involved in tumor suppression, cell-cycle regulation, and aging, in UC–CRC tumors and non-neoplastic tissues from both UC-CRC and UC patients (n = 114 for each) was analyzed (Garrity-Park et al., 2010). Methylation of RUNX3, MINT1, and COX-2 genes in non-neoplastic tissue was significantly associated with UC-CRC, suggesting their role as potential indicators of carcinogenesis (Garrity-Park et al., 2010). An altered methylation status of RUNX3 in the non-neoplastic sections of UC-CRC was also observed by Scarpa and others (Scarpa et al., 2016).

Among the epigenetic factors, emerging data have implicated the altered expression of specific microRNAs in IBD-associated tumorigenesis: miR-18a, -19a, -21, -31, -155 and -214 were upregulated in IBD-CRC colon tissues compared to healthy controls while miR-124, -193a and -139 were downregulated (Bocchetti et al., 2021), however, further prospective studies on large cohort of patients are needed.

Inflammation and Tumorigenesis in IBD-CRC

The innate and adaptive immune system cells play an important role in the onset of IBD-CRC. Crosstalk between these cell types occurs mainly through a network of cytokines that drive and maintain inflammation and contribute to tumorigenesis via oxidative stress, epithelial cell proliferation, and angiogenesis (Long et al., 2017). In Table 1 is reported a brief summary of the effects of different inflammation-related molecules in IBD-CRC.

TABLE 1
www.frontiersin.org

TABLE 1. cytokines and/or pro-inflammatory molecules in IBD-CRC.

Among these molecules, the macrophage migration inhibitory factor (MIF), a pleiotropic cytokine that drives cellular proliferation and regulates the migration and activation state of immune cells, seems to be relevant. The pathophysiological role of MIF in a wide range of inflammatory diseases, among which IBD, was already demonstrated (Nishihira and Mitsuyama, 2009). Increased MIF in macrophages in a CRC mouse model was demonstrated, and loss of MIF expression protects mice during tumor initiation (Pacheco-Fernández et al., 2019; Klemke et al., 2021). In cancer cells from CRC patients and in an acute colitis-CRC mouse model, a tumor-specific elevation of MIF expression was demonstrated (Klemke et al., 2021). The heat shock protein 90 (HSP90) chaperone machinery stabilizes and protects MIF from degradation and supports tumor progression via macrophage recruitment and angiogenesis.

In the context of inflammation and CRC, the most investigated pathways are the nuclear factor κ light-chain enhancer (NF-κB) and IL-6/signal transducer and activator of transcription 3 (STAT3) and STAT6 signaling pathways (Grivennikov and Karin, 2011).

NF-κB plays an important role in tumorigenic process by several mechanisms: promotes the production of reactive oxygen and nitrogen species which induce DNA damage, causes chromosomal instability and epigenetic changes (Grivennikov and Karin, 2011). Furthermore, by stimulating the production of inflammatory cytokines and growth factors, NF-κB enhances the proliferation of tumor progenitor cells, favoring tumor progression. This effect is also enhanced by the anti-apoptotic role of NF-κB; in fact in a IBD-CRC mouse model, it was demonstrated that NF-κB suppresses apoptosis through the induction of the anti-apoptotic protein B-cell lymphoma-extra large (Bcl-xL) (Greten et al., 2004). The NF-κB pathway results aberrantly activated in most of colitis-associated tumors and is involved in the expression of pro-inflammatory genes including COX-2, TNF-α and IL-6 (Gambhir et al., 2015).

The impaired regulation of NF-κB in tumor cells is also mediated by STAT3 which prompts the retention of NF-κB into the nucleus and hence amplifies its effect during the tumorigenic process and increases the interactions and communication between cancer cells and the microenvironment (Onizawa et al., 2009). The pro-oncogenic effects of STAT3 are mostly evident following inactivation of the negative regulators of IL-6 signaling, such as the suppressor of cytokine signaling 3 (SOCS3), that leads to an increased phosphorylation of protein kinase B (AKT), and NF-κB, initiating the disease process in patients that will progress towards IBD-CRC (Johnson et al., 2018). An increased IL-6/p-STAT3 signaling in dysplasia and colon cancer was demonstrated (Li et al., 2010). SOCS3 expression is reduced during progression from active UC to IBD-CRC and the altered methylation of SOCS3 may be involved in tumor progression increasing STAT3 signaling.

Another mechanism by which STAT3 promotes tumor progression is by favoring immune cell recruitment via the sphingosine-1-phosphate (S1P) signaling (Liang et al., 2013). S1P is formed by two related sphingosine kinases, SphK1 and SphK2, and it has already been demonstrated that SphK1 and intracellular S1P maintain a persistent activation of NF-κB and STAT3 pathways that lead to IBD-CRC development (Kawamori et al., 2009; Liang et al., 2013). Interestingly, in mice, the knockout of SphK2 increased SphK1 and S1PR1 expression, providing a pro-inflammatory environment through the secretion of IL-6 and favoring the infiltration of macrophages and T cells into tumor tissues.

Being the most abundant immune cells in tumor microenvironment, tumor-associated macrophages might be critical players in IBD-CRC progression. The role of Wnt5a, a member of the Wnt family, was already assessed in CRC: Wnt5a stimulates macrophages to produce IL-10 through the activation of STAT3 signaling pathways, crucial events for the M2 tumorigenic phenotype (Liu et al., 2020). Wnt signaling has several functions in proliferation, differentiation, migration, and survival and is regulated also by the NF-κB pathway (Du and Geller, 2010). A crosstalk between Wnt/β-catenin and NF-κB signaling pathways can significantly influence the progression of inflammation and the onset of IBD-CRC.

A recent work demonstrated for the first time, using a mouse model of IBD-CRC, that the M2 macrophage polarization could be altered by genetic inactivation of the MAPK-activated protein kinase 2 (MK2), resulting in delayed tumor progression (Suarez-Lopez et al., 2020).

Other cell types involved in cancer-associated inflammation include natural killers, T-helpers, monocytes and regulatory T-cells. During IBD and progression to dysplasia, the regulatory T cells, expressing the Th17-related transcription factor RORγt, increase in the tumor and peripheral blood of individuals with IBD-CRC, releasing pro-inflammatory cytokines (Quandt et al., 2021). Authors linked this phenotype to enhanced Wnt–β-catenin signaling, inducing pro-inflammatory cytokine production and RORγt expression in Treg cells. In particular using a mouse model of IBD-CRC, they demonstrated that the binding of the β-catenin interacting partner, TCF-1, to DNA overlapped with Foxp3 binding at active enhancer regions of pro-inflammatory genes. As a consequence of Wnt–β-catenin activation, new accessible chromatin sites in these pro-inflammatory genes were generated, leading to their upregulated expression. Enhanced β-catenin binding to TCF-1 may alter the TCF-1–Foxp3-dependent regulation of these genes. In particular, pathway enrichment analysis revealed that co-binding of TCF-1 and Foxp3 increases the accessibility and transcription of genes involved in Th17 differentiation and T cell activation pathways such as IL-17, IFN-γ and TNF.

Role of Microbiota in the Pathogenesis of IBD-CRC

Although the exact mechanism of inflammation-associated carcinogenesis is still not completely known, the contribution of the gut microbiota, especially of some pathogenic bacterial species, seems to be relevant. There is a general consensus on the relationship between the gut microbiota and the immune system: microbes, through pathogen-associated molecular patterns (PAMPs), are capable to communicate with pattern recognition receptors (PRRs) in the innate immune system, such as Toll-like receptors (TLRs), retinoic acid-inducible gene I-like receptors (RLRs) and nucleotide-binding oligomerization domain-receptors (NLRs), and to trigger the immune response (Mogensen, 2009; Lu et al., 2018). Interestingly, the expression of TLR4 is strongly upregulated in colonic tissues of BALB/c mice treated with azoxymethane/dextran sodium sulphate (AOM/DSS) to induce IBD-CRC, and blocking TLR4 signaling slows the development of the tumor (Pastille et al., 2021). In addition, downregulation of the TLR2 gene inhibits the proliferation of IBD-CRC. In particular, knocking out the TLR2 gene in mice treated with 1,2-dimethylhydrazine-dextran sodium sulphate, reduced the shortening of colorectal length, the number and volume of tumors, the pathological score and tumor severity. Furthermore, knocking down the TLR2 gene in the colorectal cancer cell lines HCT116 and HT29 inhibited their proliferation (Meng et al., 2020). It is noteworthy that also the deficient stimulation of other PRRs, such as NOD2, leads to a higher risk of IBD-CRC: in particular, NOD2−/− mice, treated with AOM to induce IBD-CRC, presented an increase in the number and size of tumors (Couturier-Maillard et al., 2013).

Moreover, upon stimulation with PAMPs, the NF-κB pathway and Wnt signaling, mentioned before for their role in the inflammation and proliferation processes leading to tumorigenesis, can be activated (Santaolalla et al., 2013; Peng et al., 2020).

Growing evidence confirms the association between IBD and the alteration of the composition of gut microbiota, sometimes referred as dysbiosis (Kang and Martin, 2017). Overgrowth of specific bacterial species at the expense of commensals is related not only to IBD but also to the development of CRC (Kang and Martin, 2017; Fan et al., 2021).

Nevertheless, to the author’s knowledge, only one study investigated the gut microbiota composition in IBD-CRC patients (Richard et al., 2018), examining the gut microbiota from colonic mucosa of 10 healthy subjects (HS), 10 patients suffering from sCRC and seven patients affected by IBD-CRC. The bacterial microbiota of IBD-CRC patients had a reduced diversity compared to HS and a composition different from that of sCRC patients. In particular, when compared to HS, IBD-CRC patients have a decreased abundance in Firmicutes and Bacteroidetes and an increase in Proteobacteria; instead, Bradyrhizobiaceae and Enterobacteriaceae families, among Proteobacteria phylum, were overexpressed in the mucosa of IBD-CRC in comparison to sCRC patients (Richard et al., 2018). Interestingly, the Bradyrhizobiaceae and Enterobacteriaceae families, also abundantly proliferate in the mucosal and luminal gut of IBD patients, suggesting that the predominance of these microorganisms could be due to the pre-existing disease and that they could have a pathogenetic role in the inflammation-associated carcinogenesis (Swidsinski et al., 2002; Kaakoush et al., 2012; Wang et al., 2015).

The proliferation of Enterobacteriaceae is also associated with a lower concentration in short chain fatty acids (SCFAs); indeed, these metabolites counteract the competitive edge that O2 and NO2 give to this bacterial family during growth (Sorbara et al., 2019). Indeed, SCFAs, such as acetate, propionate and butyrate, are produced through the anaerobic fermentation of non-digestible dietary fibers by specific bacterial species, such as Faecalibacterium prausnitzii, Clostridium leptum, Eubacterium rectale and some Roseburia species, belonging to Firmicutes, whose abundance and diversity decreases in the gut microbiota of both IBD and IBD-CRC patients (Tan et al., 2014; Richard et al., 2018; Parada Venegas et al., 2019). SCFAs are a source of energy for colonocytes, elicit anti-inflammatory effects and exert antitumorigenic activity (Tan et al., 2014; Parada Venegas et al., 2019). In particular, SCFAs exert their anti-inflammatory properties binding to their FFAR2 and HCAR2 receptors, expressed on intestinal epithelial and immune cells, and thus inducing neutrophil chemotaxis to inflammatory sites, stimulating intestinal IgA secretion towards pathogenic bacteria and increasing the secretion of IL-18, which promotes gut epithelial integrity, repair and intestinal homeostasis, via inflammasome activation and IL-10, which promotes the differentiation of Treg cells (Tian et al., 2018). Instead, the antitumorigenic activity of SCFAs, especially butyrate, has been mainly attributed to the inhibition of the proliferation and the induction of apoptosis in cancer cells achieved through the alteration of gene transcription by inhibiting the activity of histone deacetylase (Tian et al., 2018). The administration of a mixture of SCFAs attenuated colonic inflammation and improved disease activity index, suppressing the expression of the proinflammatory cytokines IL-6, TNFα and IL-17 in BALB/c mice with AOM/DSS-induced CRC; the mixture also reduced the tumor incidence and size (Tian et al., 2018).

Among the Enterobacteriaceae family, Escherichia coli utilizes virulence factors, such as colibactin, a genotoxic compound, that promotes tumor growth in a xenograft mouse model and in mice with functioning autophagy, who lack for this reason genetic susceptibility for carcinogenesis, after treatment with AOM/DSS to induce CRC (Dalmasso et al., 2014; Salesse et al., 2021). Indeed, colibactin alkylates DNA and induces double-stranded breaks, playing thus a pro-tumorigenic role (Yang et al., 2020). Interestingly, a higher prevalence of colibactin-producing E. coli in patients affected by IBD compared to healthy individuals was demonstrated: inflammation could cause the upregulation of the colibactin gene and also facilitates the colonization of the mucosa by E. coli, leading to an increase in colibactin-induced DNA damage and allowing this bacterial strain to exert its carcinogenic activity (Yang et al., 2020).

Furthermore, a difference in microbial composition between the tumor and tumor-surrounding area, even if less pronounced than in sCRC, was evidenced; indeed, the Streptococcus genus was found to be more abundant in the IBD-CRC microbiota compared to the healthy adjacent mucosa (Richard et al., 2018). Streptococcus species, representative of the bacterial population of the mucosa and of the lumen of IBD patients, are associated with tumorigenesis (Biarc et al., 2004; Santoru et al., 2017; Zhang et al., 2018; Lo Presti et al., 2019). For instance, S. gallolyticus allows the tumor progression in C57BL/6 mice with AOM/DSS-induced CRC through the recruitment of tumor-infiltrating myeloid cells which can inhibit competence of T cells and increase proinflammatory cytokines (Zhang et al., 2018). Moreover, 12 proteins isolated from S. bovis are able to trigger the release of chemokines and prostaglandins in both human epithelial colonic Caco-2 cells and in rat colonic mucosa, and to promote pre-neoplastic lesions in AOM-treated rats (Biarc et al., 2004).

Investigating the gut microbiota in the AOM-DSS mouse model of IBD-CRC, similarly to what was encountered for IBD-CRC patients the microbial community was drastically altered by chronic colitis: in particular, in addition to Lactobacillus hamster, Bacteroides uniformis and Bacteroides ovatus, also Streptococcus luteciae, belonging to Streptococcus genus mentioned above for its pathogenic role, increases (Liang et al., 2014).

Chemopreventive Effects of Therapies for IBD

Since several observations support the important role of inflammation in the development of IBD-CRC, the use of anti-inflammatory and immunosuppressant drugs in IBD can reasonably reduce inflammation in the gut and consequently the risk of inflammation-related cancers.

The chemopreventive effect of 5-aminosalicylic acid (5-ASA) in IBD patients has been widely studied even though the results remain conflicting (Terdiman et al., 2007; Bernstein et al., 2011; Carrat et al., 2017). A systematic literature search including 164 studies and meta-analyses to identify all prognostic factors for advanced CRC in patients with IBD (Wijnands et al., 2021), showed that patients who received 5-ASA had a lower risk of advanced CRC. In a systematic review, a protective role of 5-ASA against CRC in UC patients in clinical-based studies but not in population-based studies was shown (Qiu et al., 2017). In IBD patients, 5-ASA at a dosage ≥1.2 g/day showed higher protective effects against CRC than at dosages <1.2 g/day. Interestingly, a recent observational study provided molecular evidence of changes in genes related to the carcinogenesis pathways such as CDC25A, CXCL10, IL8, NF-κB, and Ki-67 in colonic biopsies of 62 UC patients during long-term 5-ASA maintenance therapy; these changes may contribute to the chemopreventive effects observed in UC patients (Bajpai et al., 2019).

One of the most recent systematic review and meta-analysis, including 11 cohort and 16 case-control studies and involving 95,397 patients, highlighted that the use of thiopurines, azathioprine and mercaptopurine, was associated with a reduced risk of CRC; this chemopreventive effect was confirmed in patients with long disease duration (Beaugerie et al., 2013) but not in those with extensive colitis or primary sclerosing cholangitis (Zhu et al., 2018). Studies conducted on CESAME (19,486 patients) and ENEIDA (831 patients) cohorts confirmed that the risk for CRC is lower among IBD patients receiving thiopurine therapy (Beaugerie et al., 2013; Gordillo et al., 2015). In a well-established murine model, the thiopurine thioguanine inhibits colitis-associated cancer by decreasing β-catenin activation/nuclear translocation, providing important evidence in support of the potential therapeutic utility of this class of drugs (Sheng et al., 2021).

The impact of biological drugs on IBD-CRC development has yet to be definitely confirmed and long-term follow-up studies will be extremely important. Considering the role of TNFα in the initiation and progression of IBD-CRC (Popivanova et al., 2008; Wilson, 2008) the use of anti-TNFα agents may be useful in preventing CRC in patients with IBD (Biancone et al., 2009). A large-scale database study showing the inverse association of CRC with anti-TNFα therapy in the IBD population (225,090 CD and 188,420 UC) was recently published (Alkhayyat et al., 2021). In this study, patients with IBD who received any of the anti-TNFα agents and those who received combined treatment (anti-TNFs plus immunomodulators) had a lower risk of developing CRC. The relationship between anti-TNFα and CRC in IBD is also supported by the results obtained in animal models treated with infliximab in which CRC carcinogenesis associated with chronic colitis was reduced (Kim et al., 2010).

Conclusion

In summary, this mini-review summarizes the recent advances in the knowledge of the pathophysiology of IBD-CRC, a complex disease associated with multifactorial causes. Inflammatory pathways seem to be the major drivers of tumorigenesis in IBD patients even if the mechanisms that link inflammation and carcinogenesis remain not well characterized in patients. In this context, development of therapies targeting specific proinflammatory cytokines involved in tumorigenesis can provide a novel approach to prevent tumor initiation or progression. Further studies with large numbers of subjects are needed to address the existing gaps in the knowledge of the role of epigenetics in the process of carcinogenesis and to validate the predictive power and clinical value of the data collected so far. The identification of predictive and prognostic epigenetic markers could favor an early detection of IBD patients with increased risk of CRC. These analyses could also consider purified cellular populations, in particular epithelial cells.

Since the mucosal associated microbiota of IBD-CRC patients is characterized by the overgrowth of bacterial species playing a role in the pathogenesis of IBD-CRC, it could be assumed that increasing the levels of beneficial bacteria with probiotics could enhance the levels of the anti-inflammatory bacterial products SCFAs, restore the equilibrium and possibly ameliorate IBD-CRC condition. In addition, IBD-CRC may be at least partially prevented through mucosal healing of intestinal lesions, and the power of the potential anti-tumor effects of IBD drugs should be evaluated by rigorous prospective studies in the near future.

Author Contributions

ML, DC and MF: Conceptualization and drafting of the initial manuscript. GD and GS: writing review and editing.

Funding

This work was supported by the Institute for Maternal and Child Health “Burlo Garofolo,” Trieste, Italy (grant number RC 01/17, 10/19).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Alkhayyat, M., Abureesh, M., Gill, A., Khoudari, G., Abou Saleh, M., Mansoor, E., et al. (2021). Lower Rates of Colorectal Cancer in Patients with Inflammatory Bowel Disease Using Anti-TNF Therapy. Inflamm. Bowel Dis. 27 (7), 1052–1060. doi:10.1093/ibd/izaa252

PubMed Abstract | CrossRef Full Text | Google Scholar

Araki, A., Jin, L., Nara, H., Takeda, Y., Nemoto, N., Gazi, M. Y., et al. (2019). IL-21 Enhances the Development of Colitis-Associated Colon Cancer: Possible Involvement of Activation-Induced Cytidine Deaminase Expression. J. Immunol. 202 (11), 3326–3333. doi:10.4049/jimmunol.1800550

PubMed Abstract | CrossRef Full Text | Google Scholar

Bajpai, M., Seril, D. N., Van Gurp, J., Geng, X., Alvarez, J., Minacapelli, C. D., et al. (2019). Effect of Long-Term Mesalamine Therapy on Cancer-Associated Gene Expression in Colonic Mucosa of Patients with Ulcerative Colitis. Dig. Dis. Sci. 64 (3), 740–750. doi:10.1007/s10620-018-5378-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Baker, A. M., Cross, W., Curtius, K., Al Bakir, I., Choi, C. R., Davis, H. L., et al. (2019). Evolutionary History of Human Colitis-Associated Colorectal Cancer. Gut 68 (6), 985–995. doi:10.1136/gutjnl-2018-316191

PubMed Abstract | CrossRef Full Text | Google Scholar

Baker, K. T., Salk, J. J., Brentnall, T. A., and Risques, R. A. (2018). Precancer in Ulcerative Colitis: the Role of the Field Effect and its Clinical Implications. Carcinogenesis 39 (1), 11–20. doi:10.1093/carcin/bgx117

PubMed Abstract | CrossRef Full Text | Google Scholar

Barnicle, A., Seoighe, C., Greally, J. M., Golden, A., and Egan, L. J. (2017). Inflammation-associated DNA Methylation Patterns in Epithelium of Ulcerative Colitis. Epigenetics 12 (8), 591–606. doi:10.1080/15592294.2017.1334023

PubMed Abstract | CrossRef Full Text | Google Scholar

Beaugerie, L., Svrcek, M., Seksik, P., Bouvier, A. M., Simon, T., Allez, M., et al. (2013). Risk of Colorectal High-Grade Dysplasia and Cancer in a Prospective Observational Cohort of Patients with Inflammatory Bowel Disease. Gastroenterology 145 (1), 166. doi:10.1053/j.gastro.2013.03.044

PubMed Abstract | CrossRef Full Text | Google Scholar

Beggs, A. D., Mehta, S., Deeks, J. J., James, J. D., Caldwell, G. M., Dilworth, M. P., et al. (2019). Validation of Epigenetic Markers to Identify Colitis Associated Cancer: Results of Module 1 of the ENDCAP-C Study. EBioMedicine 39, 265–271. doi:10.1016/j.ebiom.2018.11.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Bernstein, C. N., Nugent, Z., and Blanchard, J. F. (2011). 5-aminosalicylate Is Not Chemoprophylactic for Colorectal Cancer in IBD: a Population Based Study. Am. J. Gastroenterol. 106 (4), 731–736. doi:10.1038/ajg.2011.50

CrossRef Full Text | Google Scholar

Biancone, L., Petruzziello, C., Calabrese, E., Zorzi, F., Naccarato, P., Onali, S., et al. (2009). Long-term Safety of Infliximab for the Treatment of Inflammatory Bowel Disease: Does Blocking TNFalpha Reduce Colitis-Associated Colorectal Carcinogenesis? Gut 58 (12), 1703. doi:10.1136/gut.2008.176461

PubMed Abstract | CrossRef Full Text | Google Scholar

Biarc, J., Nguyen, I. S., Pini, A., Gossé, F., Richert, S., Thiersé, D., et al. (2004). Carcinogenic Properties of Proteins with Pro-inflammatory Activity from Streptococcus Infantarius (Formerly S.Bovis). Carcinogenesis 25 (8), 1477–1484. doi:10.1093/carcin/bgh091

PubMed Abstract | CrossRef Full Text | Google Scholar

Biscaglia, G., Latiano, A., Castellana, S., Fontana, R., Gentile, A., Latiano, T., et al. (2021). Germline Alterations in Patients with IBD-Associated Colorectal Cancer. Inflamm. Bowel Dis., izab195. doi:10.1093/ibd/izab195

PubMed Abstract | CrossRef Full Text | Google Scholar

Bocchetti, M., Ferraro, M. G., Ricciardiello, F., Ottaiano, A., Luce, A., Cossu, A. M., et al. (2021). The Role of microRNAs in Development of Colitis-Associated Colorectal Cancer. Int. J. Mol. Sci. 22 (8), 3967. doi:10.3390/ijms22083967

CrossRef Full Text | Google Scholar

Carrat, F., Seksik, P., Colombel, J. F., Peyrin-Biroulet, L., and Beaugerie, L. (2017). The Effects of Aminosalicylates or Thiopurines on the Risk of Colorectal Cancer in Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 45 (4), 533–541. doi:10.1111/apt.13897

PubMed Abstract | CrossRef Full Text | Google Scholar

Castaño-Milla, C., Chaparro, M., and Gisbert, J. P. (2014). Systematic Review with Meta-Analysis: the Declining Risk of Colorectal Cancer in Ulcerative Colitis. Aliment. Pharmacol. Ther. 39 (7), 645–659. doi:10.1111/apt.12651

PubMed Abstract | CrossRef Full Text | Google Scholar

Chatila, W. K., Walch, H. S., Benhamida, J., Hechtman, J. F., Barriga, F. M., Kundra, R., et al. (2020). Genomic Alterations in Colitis-Associated Cancers in Comparison to Those Found in Sporadic Colorectal Cancer and Present in Precancerous Dysplasia. Jco 38 (4_Suppl. l), 191. doi:10.1200/jco.2020.38.4_suppl.191

CrossRef Full Text | Google Scholar

Choi, C. H., Rutter, M. D., Askari, A., Lee, G. H., Warusavitarne, J., Moorghen, M., et al. (2015). Forty-Year Analysis of Colonoscopic Surveillance Program for Neoplasia in Ulcerative Colitis: An Updated Overview. Am. J. Gastroenterol. 110 (7), 1022–1034. doi:10.1038/ajg.2015.65

PubMed Abstract | CrossRef Full Text | Google Scholar

Couturier-Maillard, A., Secher, T., Rehman, A., Normand, S., De Arcangelis, A., Haesler, R., et al. (2013). NOD2-mediated Dysbiosis Predisposes Mice to Transmissible Colitis and Colorectal Cancer. J. Clin. Invest. 123 (2), 700–711. doi:10.1172/JCI62236

CrossRef Full Text | Google Scholar

Dalmasso, G., Cougnoux, A., Delmas, J., Darfeuille-Michaud, A., and Bonnet, R. (2014). The Bacterial Genotoxin Colibactin Promotes colon Tumor Growth by Modifying the Tumor Microenvironment. Gut microbes 5 (5), 675–680. doi:10.4161/19490976.2014.969989

PubMed Abstract | CrossRef Full Text | Google Scholar

Dhir, M., Montgomery, E. A., Glöckner, S. C., Schuebel, K. E., Hooker, C. M., Herman, J. G., et al. (2008). Epigenetic Regulation of WNT Signaling Pathway Genes in Inflammatory Bowel Disease (IBD) Associated Neoplasia. J. Gastrointest. Surg. 12 (10), 1745–1753. doi:10.1007/s11605-008-0633-5

CrossRef Full Text | Google Scholar

Du, L., Kim, J. J., Shen, J., Chen, B., and Dai, N. (2017). KRAS and TP53 Mutations in Inflammatory Bowel Disease-Associated Colorectal Cancer: a Meta-Analysis. Oncotarget 8 (13), 22175–22186. doi:10.18632/oncotarget.14549

PubMed Abstract | CrossRef Full Text | Google Scholar

Du, Q., and Geller, D. (2010). Cross-Regulation between WNT and NF-κB Signaling Pathways. Immunopathol Dis. Therap. 1, 155–181. doi:10.1615/ForumImmunDisTher.v1.i3

PubMed Abstract | CrossRef Full Text | Google Scholar

Eaden, J. A., Abrams, K. R., and Mayberry, J. F. (2001). The Risk of Colorectal Cancer in Ulcerative Colitis: a Meta-Analysis. Gut 48 (4), 526–535. doi:10.1136/gut.48.4.526

PubMed Abstract | CrossRef Full Text | Google Scholar

Ekbom, A., Helmick, C., Zack, M., and Adami, H. O. (1990). Ulcerative Colitis and Colorectal Cancer. A Population-Based Study. N. Engl. J. Med. 323 (18), 1228–1233. doi:10.1056/NEJM199011013231802

CrossRef Full Text | Google Scholar

Fan, X., Jin, Y., Chen, G., Ma, X., and Zhang, L. (2021). Gut Microbiota Dysbiosis Drives the Development of Colorectal Cancer. Digestion 102 (4), 508–515. doi:10.1159/000508328

PubMed Abstract | CrossRef Full Text | Google Scholar

Gambhir, S., Vyas, D., Hollis, M., Aekka, A., and Vyas, A. (2015). Nuclear Factor Kappa B Role in Inflammation Associated Gastrointestinal Malignancies. World J. Gastroenterol. 21 (11), 3174–3183. doi:10.3748/wjg.v21.i11.3174

CrossRef Full Text | Google Scholar

Garrity-Park, M. M., Loftus, E. V., Sandborn, W. J., Bryant, S. C., and Smyrk, T. C. (2010). Methylation Status of Genes in Non-neoplastic Mucosa from Patients with Ulcerative Colitis-Associated Colorectal Cancer. Am. J. Gastroenterol. 105 (7), 1610–1619. doi:10.1038/ajg.2010.22

CrossRef Full Text | Google Scholar

Gordillo, J., Cabré, E., Garcia-Planella, E., Ricart, E., Ber-Nieto, Y., Márquez, L., et al. (2015). Thiopurine Therapy Reduces the Incidence of Colorectal Neoplasia in Patients with Ulcerative Colitis. Data from the ENEIDA Registry. J. Crohns Colitis 9 (12), 1063–1070. doi:10.1093/ecco-jcc/jjv145

PubMed Abstract | CrossRef Full Text | Google Scholar

Greten, F. R., Eckmann, L., Greten, T. F., Park, J. M., Li, Z. W., Egan, L. J., et al. (2004). IKKbeta Links Inflammation and Tumorigenesis in a Mouse Model of Colitis-Associated Cancer. Cell 118 (3), 285–296. doi:10.1016/j.cell.2004.07.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Grivennikov, S., Karin, E., Terzic, J., Mucida, D., Yu, G. Y., Vallabhapurapu, S., et al. (2009). IL-6 and Stat3 Are Required for Survival of Intestinal Epithelial Cells and Development of Colitis-Associated Cancer. Cancer Cell 15 (2), 103–113. doi:10.1016/j.ccr.2009.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Grivennikov, S. I., and Karin, M. (2011). Inflammatory Cytokines in Cancer: Tumour Necrosis Factor and Interleukin 6 Take the Stage. Ann. Rheum. Dis. 70 (Suppl. 1), i104–8. doi:10.1136/ard.2010.140145

PubMed Abstract | CrossRef Full Text | Google Scholar

Hnatyszyn, A., Hryhorowicz, S., Kaczmarek-Ryś, M., Lis, E., Słomski, R., Scott, R. J., et al. (2019). Colorectal Carcinoma in the Course of Inflammatory Bowel Diseases. Hered. Cancer Clin. Pract. 17, 18. doi:10.1186/s13053-019-0118-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Jess, T., Rungoe, C., and Peyrin-Biroulet, L. (2012). Risk of Colorectal Cancer in Patients with Ulcerative Colitis: a Meta-Analysis of Population-Based Cohort Studies. Clin. Gastroenterol. Hepatol. 10 (6), 639–645. doi:10.1016/j.cgh.2012.01.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnson, D. E., O'Keefe, R. A., and Grandis, J. R. (2018). Targeting the IL-6/JAK/STAT3 Signalling axis in Cancer. Nat. Rev. Clin. Oncol. 15 (4), 234–248. doi:10.1038/nrclinonc.2018.8

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaakoush, N. O., Day, A. S., Huinao, K. D., Leach, S. T., Lemberg, D. A., Dowd, S. E., et al. (2012). Microbial Dysbiosis in Pediatric Patients with Crohn's Disease. J. Clin. Microbiol. 50 (10), 3258–3266. doi:10.1128/JCM.01396-12

CrossRef Full Text | Google Scholar

Kang, M., and Martin, A. (2017). Microbiome and Colorectal Cancer: Unraveling Host-Microbiota Interactions in Colitis-Associated Colorectal Cancer Development. Semin. Immunol. 32, 3–13. doi:10.1016/j.smim.2017.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Kawamori, T., Kaneshiro, T., Okumura, M., Maalouf, S., Uflacker, A., Bielawski, J., et al. (2009). Role for Sphingosine Kinase 1 in colon Carcinogenesis. FASEB J. 23 (2), 405–414. doi:10.1096/fj.08-117572

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, Y. J., Hong, K. S., Chung, J. W., Kim, J. H., and Hahm, K. B. (2010). Prevention of Colitis-Associated Carcinogenesis with Infliximab. Cancer Prev. Res. 3 (10), 1314–1333. doi:10.1158/1940-6207.CAPR-09-0272

PubMed Abstract | CrossRef Full Text | Google Scholar

Klemke, L., De Oliveira, T., Witt, D., Winkler, N., Bohnenberger, H., Bucala, R., et al. (2021). Hsp90-stabilized MIF Supports Tumor Progression via Macrophage Recruitment and Angiogenesis in Colorectal Cancer. Cell Death Dis. 12 (2), 155. doi:10.1038/s41419-021-03426-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Kohno, H., Suzuki, R., Sugie, S., and Tanaka, T. (2005). Suppression of Colitis-Related Mouse colon Carcinogenesis by a COX-2 Inhibitor and PPAR Ligands. BMC Cancer 5 (1), 46. doi:10.1186/1471-2407-5-46

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., de Haar, C., Chen, M., Deuring, J., Gerrits, M. M., Smits, R., et al. (2010). Disease-related Expression of the IL6/STAT3/SOCS3 Signalling Pathway in Ulcerative Colitis and Ulcerative Colitis-Related Carcinogenesis. Gut 59 (2), 227–235. doi:10.1136/gut.2009.184176

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, J., Nagahashi, M., Kim, E. Y., Harikumar, K. B., Yamada, A., Huang, W. C., et al. (2013). Sphingosine-1-phosphate Links Persistent STAT3 Activation, Chronic Intestinal Inflammation, and Development of Colitis-Associated Cancer. Cancer Cell 23 (1), 107–120. doi:10.1016/j.ccr.2012.11.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Liang, X., Li, H., Tian, G., and Li, S. (2014). Dynamic Microbe and Molecule Networks in a Mouse Model of Colitis-Associated Colorectal Cancer. Sci. Rep. 4, 4985. doi:10.1038/srep04985

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Q., Yang, C., Wang, S., Shi, D., Wei, C., Song, J., et al. (2020). Wnt5a-induced M2 Polarization of Tumor-Associated Macrophages via IL-10 Promotes Colorectal Cancer Progression. Cell Commun Signal 18 (1), 51. doi:10.1186/s12964-020-00557-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Lo Presti, A., Zorzi, F., Del Chierico, F., Altomare, A., Cocca, S., Avola, A., et al. (2019). Fecal and Mucosal Microbiota Profiling in Irritable Bowel Syndrome and Inflammatory Bowel Disease. Front. Microbiol. 10, 1655. doi:10.3389/fmicb.2019.01655

PubMed Abstract | CrossRef Full Text | Google Scholar

Long, A. G., Lundsmith, E. T., and Hamilton, K. E. (2017). Inflammation and Colorectal Cancer. Curr. Colorectal Cancer Rep. 13 (4), 341–351. doi:10.1007/s11888-017-0373-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, Y., Li, X., Liu, S., Zhang, Y., and Zhang, D. (2018). Toll-like Receptors and Inflammatory Bowel Disease. Front. Immunol. 9, 72. doi:10.3389/fimmu.2018.00072

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, C., and Zhang, H. (2017). The Role of Proinflammatory Pathways in the Pathogenesis of Colitis-Associated Colorectal Cancer. Mediators Inflamm. 2017, 5126048. doi:10.1155/2017/5126048

PubMed Abstract | CrossRef Full Text | Google Scholar

Meng, S., Li, Y., Zang, X., Jiang, Z., Ning, H., and Li, J. (2020). Effect of TLR2 on the Proliferation of Inflammation-Related Colorectal Cancer and Sporadic Colorectal Cancer. Cancer Cel Int. 20, 95. doi:10.1186/s12935-020-01184-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Mizuno, R., Kawada, K., and Sakai, Y. (2019). Prostaglandin E2/EP Signaling in the Tumor Microenvironment of Colorectal Cancer. Int. J. Mol. Sci. 20 (24), 6254. doi:10.3390/ijms20246254

PubMed Abstract | CrossRef Full Text | Google Scholar

Mogensen, T. H. (2009). Pathogen Recognition and Inflammatory Signaling in Innate Immune Defenses. Clin Microbiol Rev. 22 (2), 240–273. doi:10.1128/CMR.00046-08

PubMed Abstract | CrossRef Full Text | Google Scholar

Nishihira, J., and Mitsuyama, K. (2009). Overview of the Role of Macrophage Migration Inhibitory Factor (MIF) in Inflammatory Bowel Disease. Curr. Pharm. Des. 15 (18), 2104–2109. doi:10.2174/138161209788489113

PubMed Abstract | CrossRef Full Text | Google Scholar

Olén, O., Erichsen, R., Sachs, M. C., Pedersen, L., Halfvarson, J., Askling, J., et al. (2020). Colorectal Cancer in Ulcerative Colitis: a Scandinavian Population-Based Cohort Study. Lancet 395 (10218), 123–131. doi:10.1016/S0140-6736(19)32545-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Onizawa, M., Nagaishi, T., Kanai, T., Nagano, K., Oshima, S., Nemoto, Y., et al. (2009). Signaling Pathway via TNF-alpha/NF-kappaB in Intestinal Epithelial Cells May Be Directly Involved in Colitis-Associated Carcinogenesis. Am. J. Physiol. Gastrointest. Liver Physiol. 296 (4), G850–G859. doi:10.1152/ajpgi.00071.2008

CrossRef Full Text | Google Scholar

Pacheco-Fernández, T., Juárez-Avelar, I., Illescas, O., Terrazas, L. I., Hernández-Pando, R., Pérez-Plasencia, C., et al. (2019). Macrophage Migration Inhibitory Factor Promotes the Interaction between the Tumor, Macrophages, and T Cells to Regulate the Progression of Chemically Induced Colitis-Associated Colorectal Cancer. Mediators Inflamm. 2019, 2056085. doi:10.1155/2019/2056085

PubMed Abstract | CrossRef Full Text | Google Scholar

Parada Venegas, D., De la Fuente, M. K., Landskron, G., González, M. J., Quera, R., Dijkstra, G., et al. (2019). Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 10, 277. doi:10.3389/fimmu.2019.00277

PubMed Abstract | CrossRef Full Text | Google Scholar

Pastille, E., Faßnacht, T., Adamczyk, A., Ngo Thi Phuong, N., Buer, J., and Westendorf, A. M. (2021). Inhibition of TLR4 Signaling Impedes Tumor Growth in Colitis-Associated Colon Cancer. Front. Immunol. 12, 669747. doi:10.3389/fimmu.2021.669747

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, C., Ouyang, Y., Lu, N., and Li, N. (2020). The NF-κB Signaling Pathway, the Microbiota, and Gastrointestinal Tumorigenesis: Recent Advances. Front. Immunol. 11, 1387. doi:10.3389/fimmu.2020.01387

PubMed Abstract | CrossRef Full Text | Google Scholar

Popivanova, B. K., Kitamura, K., Wu, Y., Kondo, T., Kagaya, T., Kaneko, S., et al. (2008). Blocking TNF-Alpha in Mice Reduces Colorectal Carcinogenesis Associated with Chronic Colitis. J. Clin. Invest. 118 (2), 560–570. doi:10.1172/JCI32453

CrossRef Full Text | Google Scholar

Qiu, X., Ma, J., Wang, K., and Zhang, H. (2017). Chemopreventive Effects of 5-aminosalicylic Acid on Inflammatory Bowel Disease-Associated Colorectal Cancer and Dysplasia: a Systematic Review with Meta-Analysis. Oncotarget 8 (1), 1031–1045. doi:10.18632/oncotarget.13715

PubMed Abstract | CrossRef Full Text | Google Scholar

Quandt, J., Arnovitz, S., Haghi, L., Woehlk, J., Mohsin, A., Okoreeh, M., et al. (2021). Wnt-β-catenin Activation Epigenetically Reprograms Treg Cells in Inflammatory Bowel Disease and Dysplastic Progression. Nat. Immunol. 22 (4), 471–484. doi:10.1038/s41590-021-00889-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Rajamäki, K., Taira, A., Katainen, R., Välimäki, N., Kuosmanen, A., Plaketti, R.-M., et al. (2021). Genetic and Epigenetic Characteristics of Inflammatory Bowel Disease-Associated Colorectal Cancer. Gastroenterology 161 (2), 592–607. doi:10.1053/j.gastro.2021.04.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Richard, M. L., Liguori, G., Lamas, B., Brandi, G., da Costa, G., Hoffmann, T. W., et al. (2018). Mucosa-associated Microbiota Dysbiosis in Colitis Associated Cancer. Gut Microbes 9 (2), 131–142. doi:10.1080/19490976.2017.1379637

PubMed Abstract | CrossRef Full Text | Google Scholar

Risques, R. A., Lai, L. A., Himmetoglu, C., Ebaee, A., Li, L., Feng, Z., et al. (2011). Ulcerative Colitis-Associated Colorectal Cancer Arises in a Field of Short Telomeres, Senescence, and Inflammation. Cancer Res. 71 (5), 1669–1679. doi:10.1158/0008-5472.CAN-10-1966

PubMed Abstract | CrossRef Full Text | Google Scholar

Rutter, M., Saunders, B., Wilkinson, K., Rumbles, S., Schofield, G., Kamm, M., et al. (2004). Severity of Inflammation Is a Risk Factor for Colorectal Neoplasia in Ulcerative Colitis. Gastroenterology 126 (2), 451–459. doi:10.1053/j.gastro.2003.11.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Salesse, L., Lucas, C., Hoang, M. H. T., Sauvanet, P., Rezard, A., Rosenstiel, P., et al. (2021). Colibactin-Producing Escherichia coli Induce the Formation of Invasive Carcinomas in a Chronic Inflammation-Associated Mouse Model. Cancers (Basel) 13 (9), 2060. doi:10.3390/cancers13092060

PubMed Abstract | CrossRef Full Text | Google Scholar

Salk, J. J., Salipante, S. J., Risques, R. A., Crispin, D. A., Li, L., Bronner, M. P., et al. (2009). Clonal Expansions in Ulcerative Colitis Identify Patients with Neoplasia. Proc. Natl. Acad. Sci. U S A. 106 (49), 20871–20876. doi:10.1073/pnas.0909428106

PubMed Abstract | CrossRef Full Text | Google Scholar

Santaolalla, R., Sussman, D. A., Ruiz, J. R., Davies, J. M., Pastorini, C., España, C. L., et al. (2013). TLR4 Activates the β-catenin Pathway to Cause Intestinal Neoplasia. PloS one 8 (5), e63298. doi:10.1371/journal.pone.0063298

PubMed Abstract | CrossRef Full Text | Google Scholar

Santoru, M. L., Piras, C., Murgia, A., Palmas, V., Camboni, T., Liggi, S., et al. (2017). Cross Sectional Evaluation of the Gut-Microbiome Metabolome axis in an Italian Cohort of IBD Patients. Sci. Rep. 7 (1), 9523. doi:10.1038/s41598-017-10034-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Scarpa, M., Scarpa, M., Castagliuolo, I., Erroi, F., Kotsafti, A., Basato, S., et al. (2016). Aberrant Gene Methylation in Non-neoplastic Mucosa as a Predictive Marker of Ulcerative Colitis-Associated CRC. Oncotarget 7 (9), 10322–10331. doi:10.18632/oncotarget.7188

PubMed Abstract | CrossRef Full Text | Google Scholar

Schmitt, M., and Greten, F. R. (2021). The Inflammatory Pathogenesis of Colorectal Cancer. Nat. Rev. Immunol. 21 (10), 653–667. doi:10.1038/s41577-021-00534-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sheng, Y. H., Giri, R., Davies, J., Schreiber, V., Alabbas, S., Movva, R., et al. (2021). A Nucleotide Analog Prevents Colitis-Associated Cancer via Beta-Catenin Independently of Inflammation and Autophagy. Cell Mol Gastroenterol Hepatol 11 (1), 33–53. doi:10.1016/j.jcmgh.2020.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Soetikno, R. M., Lin, O. S., Heidenreich, P. A., Young, H. S., and Blackstone, M. O. (2002). Increased Risk of Colorectal Neoplasia in Patients with Primary Sclerosing Cholangitis and Ulcerative Colitis: a Meta-Analysis. Gastrointest. Endosc. 56 (1), 48–54. doi:10.1067/mge.2002.125367

PubMed Abstract | CrossRef Full Text | Google Scholar

Somineni, H. K., Venkateswaran, S., Kilaru, V., Marigorta, U. M., Mo, A., Okou, D. T., et al. (2019). Blood-Derived DNA Methylation Signatures of Crohn's Disease and Severity of Intestinal Inflammation. Gastroenterology 156 (8), 2254. doi:10.1053/j.gastro.2019.01.270

PubMed Abstract | CrossRef Full Text | Google Scholar

Sorbara, M. T., Dubin, K., Littmann, E. R., Moody, T. U., Fontana, E., Seok, R., et al. (2019). Inhibiting Antibiotic-Resistant Enterobacteriaceae by Microbiota-Mediated Intracellular Acidification. J. Exp. Med. 216 (1), 84–98. doi:10.1084/jem.20181639

CrossRef Full Text | Google Scholar

Suarez-Lopez, L., Kong, Y. W., Sriram, G., Patterson, J. C., Rosenberg, S., Morandell, S., et al. (2020). MAPKAP Kinase-2 Drives Expression of Angiogenic Factors by Tumor-Associated Macrophages in a Model of Inflammation-Induced Colon Cancer. Front. Immunol. 11, 607891. doi:10.3389/fimmu.2020.607891

PubMed Abstract | CrossRef Full Text | Google Scholar

Swidsinski, A., Ladhoff, A., Pernthaler, A., Swidsinski, S., Loening-Baucke, V., Ortner, M., et al. (2002). Mucosal flora in Inflammatory Bowel Disease. Gastroenterology 122 (1), 44–54. doi:10.1053/gast.2002.30294

PubMed Abstract | CrossRef Full Text | Google Scholar

Tan, J., McKenzie, C., Potamitis, M., Thorburn, A. N., Mackay, C. R., and Macia, L. (2014). The Role of Short-Chain Fatty Acids in Health and Disease. Adv. Immunol. 121, 91–119. doi:10.1016/B978-0-12-800100-4.00003-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Terdiman, J. P., Steinbuch, M., Blumentals, W. A., Ullman, T. A., and Rubin, D. T. (2007). 5-Aminosalicylic Acid Therapy and the Risk of Colorectal Cancer Among Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 13 (4), 367–371. doi:10.1002/ibd.20074

PubMed Abstract | CrossRef Full Text | Google Scholar

Tian, Y., Xu, Q., Sun, L., Ye, Y., and Ji, G. (2018). Short-chain Fatty Acids Administration Is Protective in Colitis-Associated Colorectal Cancer Development. J. Nutr. Biochem. 57, 103–109. doi:10.1016/j.jnutbio.2018.03.007

CrossRef Full Text | Google Scholar

Ullman, T. A., and Itzkowitz, S. H. (2011). Intestinal Inflammation and Cancer. Gastroenterology 140 (6), 1807–1816. doi:10.1053/j.gastro.2011.01.057

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C., Gong, G., Sheh, A., Muthupalani, S., Bryant, E. M., Puglisi, D. A., et al. (2017). Interleukin-22 Drives Nitric Oxide-dependent DNA Damage and Dysplasia in a Murine Model of Colitis-Associated Cancer. Mucosal Immunol. 10 (6), 1504–1517. doi:10.1038/mi.2017.9

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, W., Jovel, J., Halloran, B., Wine, E., Patterson, J., Ford, G., et al. (2015). Metagenomic Analysis of Microbiome in colon Tissue from Subjects with Inflammatory Bowel Diseases Reveals Interplay of Viruses and Bacteria. Inflamm. Bowel Dis. 21 (6), 1419–1427. doi:10.1097/MIB.0000000000000344

PubMed Abstract | CrossRef Full Text | Google Scholar

West, N. R., Hegazy, A. N., Owens, B. M. J., Bullers, S. J., Linggi, B., Buonocore, S., et al. (2017). Oncostatin M Drives Intestinal Inflammation and Predicts Response to Tumor Necrosis Factor-Neutralizing Therapy in Patients with Inflammatory Bowel Disease. Nat. Med. 23 (5), 579–589. doi:10.1038/nm.4307

PubMed Abstract | CrossRef Full Text | Google Scholar

Wijnands, A. M., de Jong, M. E., Lutgens, M. W. M. D., Hoentjen, F., Elias, S. G., and Oldenburg, B. (2021). Prognostic Factors for Advanced Colorectal Neoplasia in Inflammatory Bowel Disease: Systematic Review and Meta-Analysis. Gastroenterology 160 (5), 1584–1598. doi:10.1053/j.gastro.2020.12.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilson, J. A. (2008). Tumor Necrosis Factor Alpha and Colitis-Associated colon Cancer. N. Engl. J. Med. 358 (25), 2733–2734. doi:10.1056/NEJMcibr0803116

CrossRef Full Text | Google Scholar

Yaeger, R., Shah, M. A., Miller, V. A., Kelsen, J. R., Wang, K., Heins, Z. J., et al. (2016). Genomic Alterations Observed in Colitis-Associated Cancers Are Distinct from Those Found in Sporadic Colorectal Cancers and Vary by Type of Inflammatory Bowel Disease. Gastroenterology 151 (2), 278–e6. doi:10.1053/j.gastro.2016.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, Y., Gharaibeh, R. Z., Newsome, R. C., and Jobin, C. (2020). Amending Microbiota by Targeting Intestinal Inflammation with TNF Blockade Attenuates Development of Colorectal Cancer. Nat. Cancer 1 (7), 723–734. doi:10.1038/s43018-020-0078-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Weng, Y., Gan, H., Zhao, X., and Zhi, F. (2018). Streptococcus Gallolyticus Conspires Myeloid Cells to Promote Tumorigenesis of Inflammatory Bowel Disease. Biochem. Biophys. Res. Commun. 506 (4), 907–911. doi:10.1016/j.bbrc.2018.10.136

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, Z., Mei, Z., Guo, Y., Wang, G., Wu, T., Cui, X., et al. (2018). Reduced Risk of Inflammatory Bowel Disease-Associated Colorectal Neoplasia with Use of Thiopurines: a Systematic Review and Meta-Analysis. J. Crohns Colitis 12 (5), 546–558. doi:10.1093/ecco-jcc/jjy006

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: inflammatory bowel deases, colorectal cancer, inflammation, epigenetics, microbiota, immunomodulators

Citation: Lucafò M, Curci D, Franzin M, Decorti G and Stocco G (2021) Inflammatory Bowel Disease and Risk of Colorectal Cancer: An Overview From Pathophysiology to Pharmacological Prevention. Front. Pharmacol. 12:772101. doi: 10.3389/fphar.2021.772101

Received: 07 September 2021; Accepted: 06 October 2021;
Published: 20 October 2021.

Edited by:

Mireille Alhouayek, Catholic University of Louvain, Belgium

Reviewed by:

John Kisiel, Mayo Clinic, United States
Joan Bestard-Escalas, Catholic University of Louvain, Belgium

Copyright © 2021 Lucafò, Curci, Franzin, Decorti and Stocco. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Giuliana Decorti, giuliana.decorti@burlo.trieste.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.