Open Access

Fusobacterium nucleatum‑positive colorectal cancer (Review)

  • Authors:
    • Zhenhua Yang
    • Guang Ji
  • View Affiliations

  • Published online on: June 4, 2019     https://doi.org/10.3892/ol.2019.10433
  • Pages: 975-982
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) is an important threat to human health and the fourth leading cause of mortality worldwide. Accumulating evidence indicates that the composition of the intestinal flora is associated with the occurrence of CRC. Fusobacterium nucleatum (Fn), one of the highly enriched bacteria in CRC tissues, invades the mucosa with adhesion factors and virulence proteins, interacts with the host immune system and promotes the occurrence and development of CRC and chemoresistance. Fn infection is prevalent in human colorectal carcinoma, although the infection rate varies in different regions. Fn may be used as a prognostic indicator of CRC. It is important to understand the multi‑pathway carcinogenic mechanisms associated with CRC in order to develop novel antibacterial drugs against Fn. The current review summarizes the role of Fn and relevant research concerning CRC published in recent years, focusing on Fn infection in CRC, pathogenesis of Fn, Fn‑positive CRC treatment, screening and prevention strategies against Fn‑positive CRC.

Introduction

Colorectal cancer (CRC) was the third most common cancer type worldwide and the fourth leading cause of human cancer-associated mortality in 2012 (1). The incidence of CRC was highest in Europe, North America and Oceania, and lowest in South Asia, Central Asia and Africa between 1998–2002 (2). In China, according to epidemiological data in recent years, the five-year incidence was 74.6/100,000 and 58.3/100,000 in males and females in 2011, respectively. Each year, >376,000 people are diagnosed with CRC and ~191,000 patients succumb to CRC in China (3,4).

The pathogenesis of CRC is complex. In terms of environmental factors, epidemiological studies have indicated that high-fat diet, obesity and a western lifestyle increase the risk of CRC (57). In terms of genetic factors, the majority of CRC cases exhibit genomic instability, including microsatellite and chromosomal instability (8,9). In addition, gene mutations, including tumor suppressor gene inactivation and oncogene activation, serve an important role in the pathogenesis of CRC (8). However, there has been less focus on the role of microbial infections in the pathogenesis of CRC, despite the fact that direct evidence for an infectious cause in human cancer has been reported in the last decades (10). According to previous statistical data, 15–20% of all types of cancer are caused by infections with microbial pathogens (11).

There are >1,000 types of microorganisms in the human intestinal microbiota. A total of ~1014 microorganisms serve an important role in maintaining the physiological functions of the intestines, including energy metabolism, epithelial cell proliferation and apoptosis, and protection against pathogens (12). In addition to these beneficial roles, intestinal microbes can have a detrimental effect on human health. In recent years, a large number of studies have indicated that the intestinal flora is closely associated with the occurrence of CRC (13,14). Metagenomics and transcriptional analyses have revealed that compared with adjacent normal tissues, the enrichment of Bacteroidetes and Firmicutes is decreased in human CRC tissues, but the enrichment of Fusobacterium nucleatum (Fn) is significantly increased (15,16). Yamamura et al (17) reported that Fn is detected in 20, 10 and 45% of esophageal, gastric and CRC tissues, respectively. No Fn was detected in liver and pancreatic cancer tissues (17). Fn adheres to and invades the intestinal mucosa through its surface adhesion factors and virulence proteins, and ultimately promotes the occurrence and development of CRC (18). It has previously been identified that the absolute copy number of Fn in CRC tissues may be used as an indicator to evaluate the prognosis of patients with CRC (19). Recent studies have demonstrated that Fn is not only associated with the development of CRC, but also promotes chemotherapeutic resistance (20,21). Therefore, in depth knowledge of the mechanisms underlying Fn carcinogenesis in CRC may increase the understanding of the intestinal flora and aid the development of effective anticancer agents against the bacterium.

Fn infection in CRC

Fn is an opportunistic anaerobic bacterium in the mouth and is one of the most common gram-negative bacteria in extraoral infections (22). Fn serves an important role in periodontitis, appendicitis, gingivitis and invasive infections of the head, neck, lungs, liver, heart and brain (2325). It has been reported that Fn is widely present in pregnancy complications, including premature birth and stillbirth, as well as intrauterine infections, including neonatal sepsis (26). Previous studies have investigated the link between colonized bacteria in the mouth and extraoral infections (27,28). Fn in subgingival plaque may be transferred to the placenta and fetus, leading to acute inflammation and fetal death (24). Therefore, it is speculated that Fn is transmitted from the oral cavity through hematogenous transmission to a site that is suitable for its colonization and animal experiments support this hypothesis (2931). In addition, adhesion factors and virulence proteins on the surface of Fn facilitate its colonization and invasion (18). Studies have indicated that Fn binds to a variety of mammalian cells and macromolecules, including epithelial, endothelial, blood and immune cells, and salivary macromolecules (32). In previous years, it has been identified that human CRC tissues are generally infected with Fn (1521). At present, Fn may be detected in tissues by fluorescence quantitative (FQ)-PCR, fluorescence in situ hybridization (FISH), qPCR and droplet digital PCR (ddPCR) (33). Castellarin et al (34) compared CRC tissues in 99 cases with corresponding normal mucosa tissues using qPCR and revealed that the mean overall abundance of Fn was 415 times higher compared with the normal samples. Li et al (35) investigated 101 Chinese patients with CRC and demonstrated that the Fn infection rate was 87.1%, which was significantly higher compared with that in the adjacent normal tissues. In the USA, 598 CRC cases were analyzed by qPCR and the infection rate of Fn was identified to be higher in tumor tissues (13.0%) compared with the Fn infection rate in normal tissues (3.4%), which was significantly lower compared with studies conducted in China (36). A report from Japan demonstrated that the positive rate of Fn was 8.6%, which was similar to the rate obtained by American researchers (37). One study identified that the enrichment of Fn in CRC tissues increased gradually from 2.5% (4/157) in the rectum to 11% (19/178) in the cecum, indicating a relationship with the location of the lesion (38). Komiya et al (39) performed arbitrarily primed PCR as the strain typing method and the same Fn strain was detected in saliva specimens of 75% CRC-positive patients, suggesting Fn in CRC tissues originates in the oral cavity, and digestive tract transmission may be one mechanism underlying Fn diffusion. Bullman et al (40) investigated Fn enrichment in patients with CRC with liver metastasis and demonstrated that Fn was enriched in CRC tissues compared with normal tissues; similarly in liver metastatic CRC, the same strain of Fn was detected in liver and colon tissue, demonstrating that distant metastasis of Fn may occur along the lymphatic vessels (40). High enrichment of Fn was observed in CRC tissues and precancerous lesions, including adenomas (41). Amitay and Brenner (42) demonstrated the high enrichment of Fn in CRC tissues and identified that there was no difference in the degree of Fn enrichment between adenomas and advanced adenomas. Therefore, it is inferred that Fn is a bacterium that grows in the tissues of neoplastic lesions and propagates in favorable conditions caused by malignant tumors, but it is not the cause of CRC. Ito et al (43) investigated 465 cases of precancerous lesions, including 343 serrated lesions and 122 non-serrated adenomas, and observed that Fn is detected in 24% of hyperplastic polyps, 35% of sessile serrated adenomas, 30% of traditional serrated adenoma and 33% of non-dental adenomas, and the Fn-positive rate gradually increased from the sigmoid colon to the cecum in sessile serrated adenoma. This study demonstrated that Fn is highly enriched in the precancerous lesion stage during the occurrence and development of CRC. Further research is required to determine whether Fn is involved in the occurrence of precancerous lesions. Although studies have demonstrated that the incidence of Fn infection varies in different countries, it is concluded that Fn is generally present in CRC tissues.

Carcinogenic mechanisms of Fn

A number of studies have been conducted to study the carcinogenicity of Fn (44,45). There are three biomolecules that are located on the surface of Fn including lipopolysaccharides (LPS), adhesin A (FadA) and fusobacterium autotransporter protein 2 (Fap2), which are involved in and promote the occurrence of colorectal cancer (Fig. 1) (18,31,46). Chronic inflammation may promote the occurrence and development of tumors; therefore, the role of cytokines in the pathogenesis of CRC has drawn much attention (47). Tumors may form following chronic inflammation or exhibit characteristics of chronic inflammatory infiltration during progression (48,49). In an intestinal tumorigenesis APCmin/+ mouse model, Fn increases the tumor multiplicity and selectively, and recruits tumor-infiltrating myeloid cells (50). Recruiting tumor infiltrating immune cells produces a proinflammatory microenvironment, which is conducive to the occurrence of CRC (51,52). The degree of enrichment of bone marrow-derived suppressor cells (MDSCs) is significantly increased in Fn-fed mice compared with the control group (50). MDSCs have two major subpopulations, granulocytes and monocytes, both of which function as T-cell inhibitory phenotypes (5254). Therefore, it has been speculated that Fn promotes the occurrence and development of colon tumors by downregulating the T-cell-mediated adaptive immune antitumor effects. A study by Mima et al (36) supported this hypothesis, as it reported that the presence of Fn is negatively correlated with the density of CD3+ T cells in CRC tissues. Fn promotes the release of proinflammatory cytokines, including interleukin (IL)-8, IL-10 and tumor necrosis factor-α (TNF-α) (55). In addition, studies have demonstrated that Fn promotes the mRNA expression of TNF-α, IL-1β, IL-6, IL-8, prostaglandin-endoperoxide synthase 2 (PTGS2), nuclear factor κβ (NF-κβ) and matrix metallopeptidase 3, and the expression of these genes is induced in mouse and human cells treatment with Fn (56,57). The generation and release of these inflammatory cytokines constitutes to the immune microenvironment for the occurrence of CRC (58).

Fn adsorption and invasion into mucosa is a prerequisite for its role in carcinogenesis (59). A FadA virulence protein is expressed on the surface of Fn. This virulence protein is highly conserved for Fn (60). FadA exists in two forms: The non-secreted, intact FadA precursor (pre-FadA) consisting of 129 amino acid residues, and the secreted, mature FadA (mFadA) consisting of 111 amino acid residues (61,62). When mFadA is mixed with pre-FadA, they form active FadAc (FadA virulence protein) composed of non-uniform filaments (62). Fn binds to E-cadherin on CRC and non-CRC cells via FadA virulence protein (FadA) and mediates Fn attachment and invasion into epithelial cells (63). It has been demonstrated that FadA regulates E-cadherin and activates β-catenin signaling, leading to increased expression of transcription factors, oncogenes, Wnt signaling pathway and inflammatory genes, as well as proliferation of CRC cells (63). The same study revealed the oncogenic mechanism of Fn and identified FadA as a potential diagnostic and therapeutic target for CRC. Notably, another study demonstrated that Fn activated the β-catenin signaling pathway via lipopolysaccharide-mediated Toll-like receptor 4 (TLR4)/p21 activated kinase 1 (PAK1) in CRC (64). TLR4 activates the β-catenin signaling pathway, forming intestinal tumors, while PAK1 is associated with CRC progression and metastasis (64,65). An improved understanding of the signaling pathways activated by Fn may provide new avenues for the prevention and treatment of CRC. However, E-cadherin is expressed in different types of cells, and its expression levels and cell localization patterns may be different in dysplasia and tumors (66). Therefore, FadA virulence protein cannot fully explain the selection specificity of Fn. Abed et al (67) identified that a highly expressed polysaccharide, D-galactose-β-(13)-N-acetyl-D-galactosamine (Gal-GalNAc), is present in CRC tissues. The biosynthesis of Gal-GalNAc is increased gradually from normal mucosa, adenoma to CRC tissues (67). Gal-GalNAc binds to Fap2, another protein on Fn, leading to high enrichment of Fn in CRC tissues (67). Another study on Fap2 demonstrated that Fn binds to the T cell immunoreceptor with Ig and tyrosine-based inhibitory motif domains (TIGIT) via Fap2 and that TIGIT is widely present on the surface of natural killer and T cells, thereby preventing immune cells from attacking CRC cells (68). Fn also inhibited the activity of human T cells by blocking the G1 phase of the cell cycle (69). This study suggested the presence of an Fn-dependent tumor immune escape mechanism. Yang et al (70) revealed that Fn activated NF-κβ signaling via the innate immune pathway [TLR4/MYD88 innate immune signal transduction adaptor (MYD88)] and miRNA-21, and increased the proliferation of CRC cells and the development of tumors. Compared with non-neoplastic colon tissues, Fn DNA and miRNA-21 levels are significantly increased in CRC tissues, and the levels were even higher in advanced CRC tissues. This indicated that patients with both Fn DNA and high miRNA-21 exhibit a higher risk of poor prognosis. This study revealed that Fn regulates the expression of miRNAs, but the specific mechanism of this is not yet understood.

In recent years, studies have identified that Fn infection is associated with CRC-specific molecular typing. Tahara et al (71) studied CRC samples from 149 patients and reported that Fn is highly enriched in the CRC group, which was correlated with CpG island methylation phenotype (CIMP; P<0.001), tumor protein p53 (TP53) wild type (P<0.015), human mutL homolog 1 methylation (P<0.003), microsatellite instability (MSI; P<0.018) and chromodomain helicase DNA binding protein 7/8 mutation (P<0.002) phenotypes. Mima et al (72) identified that Fn is associated with MSI-high, CIMP and B-Raf proto-oncogene serine/threonine kinase (BRAF) mutations. In CRC molecular typing, CIMP and MSI are mainly identified in the right hemicolon, which may be related to the high enrichment of Fn, but the specific mechanism needs to be studied in more detail (72). Therefore, Mima et al (72) provided strong support for the pathogenic role of intestinal microbial components in CRC.

According to statistical data, 20–30% of treated patients with CRC exhibit tumor recurrence and 35% of these patients succumb to the disease within 5 years (73). Combination chemotherapy remains the main treatment for advanced CRC (74). Therefore, studies on the mechanism of CRC resistance to chemotherapy are important. A study by Yu et al (75) demonstrated that Fn abundance in recurrent CRC tissues is markedly higher compared with that in non-recurrent CRC tissues, and high-abundance Fn is associated with poor prognosis. Therefore, Fn may be used as a diagnostic marker for preventing CRC recurrence and as a prognosis predictor. In addition, a series of experiments revealed that Fn selectively decreases miRNA-18a*/4802 expression levels through the innate immune pathway (TLR4/MYD88) and subsequently activates cell autophagy via unc-51 like autophagy activating kinase 1-autophagy related 7 to become resistant to chemotherapeutic drugs. This drug resistance was overcome by the autophagy blocker chloroquine (75).

Treatment of Fn-positive CRC

Fn invasion into the mucosa promotes the release of inflammatory cytokines and is involved in the formation of the immune microenvironment for the occurrence of CRC (5558). Due to the inflammatory basis of CRC, anti-inflammatory agents may be candidates for treating or preventing CRC (76). Non-steroidal anti-inflammatory drugs (NSAIDs) are non-selective inhibitors of PTGS2 (77). PTGS2 is highly expressed in many tumor types, including CRC (78). Furthermore, Fn promotes the expression of PTGS2 (56). NSAIDs may serve a role in the prevention of CRC. A preliminary study on patients with familial adenomatous polyposis indicated that following 1 year of treatment with the NSAID sulindac, patients exhibit a decrease in polyps (79). A large-scale demographic observation study from 1991 reported that the use of NSAIDs reduces the risk of fatal CRC (80). Retrospective studies have demonstrated that NSAID treatment is associated with a decreased risk of recurrence of colorectal polyps and tumor (81,82). Patients who use low-dose aspirin for >5 years exhibit a decreased overall risk for developing CRC by 40–50% and NSAIDs have a positive effect on advanced CRC (83,84). Furthermore, non-steroidal antitumor drug therapy inhibits a tumor-promoting pathway by inhibiting Wnt signaling (85).

According to a previous report, prostaglandin E receptor 2 (PTGER2) increases the expression of NF-κβ-targeted proinflammatory genes in neutrophils. The expression of TNF-α and IL-6, PTGS2, C-X-C motif chemokine ligand 1, Wnt and other cytokines in tumor lesions are significantly higher in PTGER2-enriched compared with PTGER2-knockout mice (86). Therefore, NSAIDs and PTGER2 antagonists may be candidates for the prevention and treatment of Fn-positive CRC.

Fn is one of the most common gram-negative bacteria; therefore antibiotics against Fn are used to treat Fn-positive CRC. Bullman et al (40) transplanted Fn-infected colon cancer cell allografts into mice, maintained the animals for several weeks and then treated them with erythromycin or metronidazole. Fn is resistant to erythromycin but sensitive to metronidazole according to the drug sensitivity test. The results demonstrated that compared with the erythromycin-resistant Fn group, the volume and number of tumors is significantly reduced in mice treated with metronidazole. This indicated that antibiotic intervention may be used as a potential treatment for Fn-positive patients with CRC. Therefore, the development of targeted antibiotics with a narrow spectrum may selectively eliminate pathogens while maintaining the balance of flora and avoiding the side effects caused by the use of broad spectrum antibiotics (87). It has been reported that berberine reverses the imbalance of intestinal microbiota caused by Fn, thereby reversing the growth of CRC in vivo (88).

Tumor immunotherapy is a promising area, with significant progress being made in tumor molecular biology, particularly with regards to the use of immune checkpoint inhibitors, such as programmed cell death 1 (PDCD1) inhibitors (89). The efficacy of checkpoint inhibitors depends on the patient's gut microbiota. Complex interactions between the gut microorganisms and the immune system limit the effects of PDCD1 inhibitors (90). According to the OncoKB classification system, pembrolizumab is a Food and Drug Association-approved drug for MSI-high solid tumors (level 1) and Fn is associated with MSI-high, CIMP and BRAF mutations (71,72,91). Therefore, PDCD1 inhibitors may exhibit anticancer effects on Fn-positive CRC. However, immune checkpoint inhibitors have a number of side effects, including hepatitis, diarrhea and enterocolitis, resulting from the complex interactions between host genetics, immune responses, the environment and microbes (92). Therefore, the use of immune checkpoint inhibitors has very strict indications (92). Since Fn binds to TIGIT, an immune cell inhibitory receptor, through Fap2 to avoid attacks of immune cells, the development of an anti-Fap2 antibody may be beneficial for the antitumor immune response (68). Nedaeinia et al (93) revealed that inhibition of miRNA-21 suppresses metastasis of CRC cells through modulation of programmed cell death 4. Kumar et al (94) studied host-pathogen protein-protein interactions (HP-PPIs) and identified that Fn and CRC-related proteins have 186 interactions, including 103 host proteins and 76 Fn-pathogenic proteins. Therefore, the development of drugs targeting HP-PPIs may be used to treat Fn-positive CRC. In view of the important role of TLR4 in Fn carcinogenesis, it may be possible to develop a drug for the treatment of Fn-positive tumors (65). For patients with recurrent CRC, in addition to combination chemotherapy, the benefit of autophagy blocking agents or Fn inhibitors requires investigation in future studies.

Screening and prevention of Fn-positive CRC

Fn can be detected in formalin-fixed paraffin-embedded CRC tissues, frozen CRC tissues and feces of patients with CRC using FQ-PCR, FISH, qPCR and ddPCR (33). For screening, it is not convenient to obtain tissue specimens, whereas feces and serum samples are more appropriate. A number of studies have identified that Fn infection leads to elevated anti-Fn antibody IgA levels in the serum of patients with CRC (95,96). Serum anti-Fn antibody IgA combined with carbohydrate antigen 19-9 (CA19-9) and carcinoembryonic antigen (CEA) is more sensitive compared with only CA19-9 and CEA in screening for early stage CRC, suggesting that serum Fn antibodies may be a potential marker for the diagnosis of early stage CRC (97). The fecal immunochemical test (FIT) is recommended as a non-invasive screening test for CRC. However, it is less sensitive in advanced adenomas (98). Wong et al (99) detected Fn in feces by qPCR combined with FIT, which increases the detection rate of CRC and advanced adenomas. Its sensitivity and specificity for CRC are 92.3 and 93.0%, respectively, and for advanced adenomas 38.6 and 89.0%, respectively, are reported. Huang et al (100) employed loop-mediated isothermal amplification technology to quickly and efficiently identify the Fn virulence factor FadA through the nusG gene, which increases the sensitivity by 10-fold compared with qPCR. Therefore, Fn may be used as a microbial marker to increase the diagnostic efficiency for CRC in the future.

Previous studies have demonstrated that a diet rich in fruits, vegetables and whole grains is associated with a lower risk of colon cancer compared with Western dietary patterns, which are dominated by red and processed meats (57,101). A high-fat diet and overconsumption of red meat may increase the risk of CRC (102104). Although the mechanism of the association between diet and CRC remains unclear, it is speculated that the gut microbiota may play an intermediary role. It has been reported that diet influences the composition of gut microbiota, although long-term dietary patterns outweigh short-term changes in diet (105,106). Considering that different groups of gut microbiota have different metabolic capacities, particular dietary patterns may allow for the selection of certain microbes and inhibits others (107,108). A previous study demonstrated that certain microflora may become extinct. By providing low-fiber diets to mice, reversible changes are observed in the microbiota. However, following low-fiber diets for several successive generations, the diversity of microbiota is gradually lost and certain microbiota are undetectable (108).

Previous studies have attempted to change the enrichment of Fn in the intestines through diet. Specific nutrients may cause intestinal inflammation, characterized by elevated circulatory levels of IL-6, creatine protein (CRP) and TNF receptor superfamily 1B (TNFRSF1B), and promote CRC (109). Inflammatory effects of different diets may be estimated using the Empirical Dietary Inflammatory Pattern (EDIP) score, which is calculated based on the intake of 18 foods associated with levels of IL-6, CRP and TNFRSF1B in the plasma (110). Liu et al (111) demonstrated that higher EDIP scores are associated with increased risk of Fn-positive CRC, and high EDIP scores are associated with proximal Fn-positive CRC. The aforementioned studies indicated that the negative association between a diet rich in fruits, vegetables and whole grains and risk of CRC is more pronounced in the Fn-positive CRC subgroup compared with in the Fn-negative CRC group.

Conclusions

In summary, Fn infection is associated with the occurrence of many human diseases and Fn infection is prevalent in human CRC tissues. Fn invades the intestinal epithelial mucosa via its own adhesion factors and virulence proteins, and subsequently interacts with the host immune system to promote the occurrence and development of CRC and chemoresistance through a variety of molecular pathways. This ultimately affects the efficacy of CRC treatment. Fn-positive CRC is characterized by shorter survival times and poor prognosis. Fn may be used as an evaluation index to predict the clinical outcomes of CRC. Eradication therapy for Fn may not only treat Fn-positive CRC but also prevent Fn-positive CRC recurrence. At present, there are no clinically effective drugs that target Fn-positive CRC. Therefore, it is expected that targeted immunotherapy drugs and biological agents with minimal side effects will be developed in the future.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

GJ designed the study, and ZY drafted the manuscript and revised the manuscript. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global patterns and trends in colorectal cancer incidence and mortality. Gut. 66:683–691. 2017. View Article : Google Scholar : PubMed/NCBI

2 

Center MM, Jemal A, Smith RA and Ward E: Worldwide variations in colorectal cancer. CA Cancer J Clin. 59:366–378. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Zheng R, Zeng H, Zhang S, Chen T and Chen W: National estimates of cancer prevalence in China, 2011. Cancer Lett. 370:33–38. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Berg A: Nutrition, development, and population growth. Popul Bull. 29:3–37. 1973.PubMed/NCBI

6 

Alsheridah N and Akhtar S: Diet, obesity and colorectal carcinoma risk: Results from a national cancer registry-based middle-eastern study. BMC Cancer. 18:12272018. View Article : Google Scholar : PubMed/NCBI

7 

Mafiana RN, Al Lawati AS, Waly MI, Al Farsi Y, Al Kindi M and Al Moundhri M: Association between dietary and lifestyle indices and colorectal cancer in Oman: A case-control study. Asian Pac J Cancer Prev. 19:3117–3122. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Markowitz SD and Bertagnolli MM: Molecular origins of cancer: Molecular basis of colorectal cancer. N Engl J Med. 361:2449–2460. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S and Tabernero J: Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer. 17:2682017. View Article : Google Scholar : PubMed/NCBI

10 

Brower V: Connecting viruses to cancer: How research moves from association to causation. J Natl Cancer Inst. 96:256–257. 2004. View Article : Google Scholar : PubMed/NCBI

11 

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, . Biological agents. Volume 100 B. A review of human carcinogens. IARC Monogr Eval Carcinog Risks Hum. 100:1–441. 2012.

12 

Tremaroli V and Bäckhed F: Functional interactions between the gut microbiota and host metabolism. Nature. 489:242–249. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Yamashiro Y: Gut microbiota in health and disease. Ann Nutr Metab. 71:242–246. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Sears CL and Garrett WS: Microbes, microbiota, and colon cancer. Cell Host Microbe. 15:317–328. 2014. View Article : Google Scholar : PubMed/NCBI

15 

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

16 

Repass J, Maherali N and Owen K; Reproducibility Project, : Cancer Biology; Reproducibility Project Cancer Biology: Registered report: Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. ELife. 5(pii): e100122016. View Article : Google Scholar : PubMed/NCBI

17 

Yamamura K, Baba Y, Miyake K, Nakamura K, Shigaki H, Mima K, Kurashige J, Ishimoto T, Iwatsuki M, Sakamoto Y, et al: Fusobacterium nucleatum in gastroenterological cancer: Evaluation of measurement methods using quantitative polymerase chain reaction and a literature review. Oncol Lett. 14:6373–6378. 2017.PubMed/NCBI

18 

Han YW: Fusobacterium nucleatum: A commensal-turned pathogen. Curr Opin Microbiol. 23:141–147. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Yamaoka Y, Suehiro Y, Hashimoto S, Hoshida T, Fujimoto M, Watanabe M, Imanaga D, Sakai K, Matsumoto T, Nishioka M, et al: Fusobacterium nucleatum as a prognostic marker of colorectal cancer in a Japanese population. J Gastroenterol. 53:517–524. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Ramos A and Hemann MT: Drugs, Bugs, and Cancer: Fusobacterium nucleatum promotes chemoresistance in colorectal cancer. Cell. 170:411–413. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Zhang S, Yang Y, Weng W, Guo B, Cai G, Ma Y and Cai S: Fusobacterium nucleatum promotes chemoresistance to 5-fluorouracil by upregulation of BIRC3 expressio n in colorectal cancer. J Exp Clin Cancer Res. 38:142019. View Article : Google Scholar : PubMed/NCBI

22 

Han YW, Shi W, Huang GT, Kinder Haake S, Park NH, Kuramitsu H and Genco RJ: Interactions between periodontal bacteria and human oral epithelial cells: Fusobacterium nucleatum adheres to and invades epithelial cells. Infect Immun. 68:3140–3146. 2000. View Article : Google Scholar : PubMed/NCBI

23 

Heckmann JG, Lang CJ, Hartl H and Tomandl B: Multiple brain abscesses caused by Fusobacterium nucleatum treated conservatively. Can J Neurol Sci. 30:266–268. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Han YW, Fardini Y, Chen C, Iacampo KG, Peraino VA, Shamonki JM and Redline RW: Term stillbirth caused by oral Fusobacterium nucleatum. Obstet Gynecol. 115:442–445. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Wells CD, Balan V and Smilack JD: Pyogenic liver abscess after colonoscopy in a patient with ulcerative colitis. Clin Gastroenterol Hepatol. 3:xxiv2005. View Article : Google Scholar : PubMed/NCBI

26 

Wang X, Buhimschi CS, Temoin S, Bhandari V, Han YW and Buhimschi IA: Comparative microbial analysis of paired amniotic fluid and cord blood from pregnancies complicated by preterm birth and early-onset neonatal sepsis. PLoS One. 8:e561312013. View Article : Google Scholar : PubMed/NCBI

27 

Hill GB: Investigating the source of amniotic fluid isolates of fusobacteria. Clin Infect Dis. 16 (Suppl 4):S423–S424. 1993. View Article : Google Scholar : PubMed/NCBI

28 

Han YW, Redline RW, Li M, Yin L, Hill GB and McCormick TS: Fusobacterium nucleatum induces premature and term stillbirths in pregnant mice: Implication of oral bacteria in preterm birth. Infect Immun. 72:2272–2279. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Han YW: Can oral bacteria cause pregnancy complications? Womens Health (Lond). 7:401–404. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Han YW: Oral health and adverse pregnancy outcomes-what's next? J Dent Res. 90:289–293. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Liu H, Redline RW and Han YW: Fusobacterium nucleatum induces fetal death in mice via stimulation of TLR4-mediated placental inflammatory response. J Immunol. 179:2501–2508. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Bachrach G, Ianculovici C, Naor R and Weiss EI: Fluorescence based measurements of Fusobacterium nucleatum coaggregation and of fusobacterial attachment to mammalian cells. FEMS Microbiol Lett. 248:235–240. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Shang FM and Liu HL: Fusobacterium nucleatum and colorectal cancer: A review. World J Gastrointest Oncol. 10:71–81. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Castellarin M, Warren RL, Freeman JD, Dreolini L, Krzywinski M, Strauss J, Barnes R, Watson P, Allen-Vercoe E, Moore RA and Holt RA: Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 22:299–306. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Li YY, Ge QX, Cao J, Zhou YJ, Du YL, Shen B, Wan YJ and Nie YQ: Association of Fusobacterium nucleatum infection with colorectal cancer in Chinese patients. World J Gastroenterol. 22:3227–3233. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Mima K, Sukawa Y, Nishihara R, Qian ZR, Yamauchi M, Inamura K, Kim SA, Masuda A, Nowak JA, Nosho K, et al: Fusobacterium nucleatum and T cells in colorectal carcinoma. JAMA Oncol. 1:653–661. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Nosho K, Sukawa Y, Adachi Y, Ito M, Mitsuhashi K, Kurihara H, Kanno S, Yamamoto I, Ishigami K, Igarashi H, et al: Association of Fusobacterium nucleatum with immunity and molecular alterations in colorectal cancer. World J Gastroenterol. 22:557–566. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Mima K, Cao Y, Chan AT, Qian ZR, Nowak JA, Masugi Y, Shi Y, Song M, da Silva A, Gu M, et al: Fusobacterium nucleatum in colorectal carcinoma tissue according to tumor location. Clin Transl Gastroenterol. 7:e2002016. View Article : Google Scholar : PubMed/NCBI

39 

Komiya Y, Shimomura Y, Higurashi T, Sugi Y, Arimoto J, Umezawa S, Uchiyama S, Matsumoto M and Nakajima A: Patients with colorectal cancer have identical strains of Fusobacterium nucleatum in their colorectal cancer and oral cavity. Gut. Jun 22–2018.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

40 

Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Huang K, Guevara F, Nelson T, et al: Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. 358:1443–1448. 2017. View Article : Google Scholar : PubMed/NCBI

41 

McCoy AN, Araújo-Pérez F, Azcárate-Peril A, Yeh JJ, Sandler RS and Keku TO: Fusobacterium is associated with colorectal adenomas. PLoS One. 8:e536532013. View Article : Google Scholar : PubMed/NCBI

42 

Amitay EL and Brenner H: Response to comments on ‘Fusobacterium and colorectal cancer: Causal factor or passenger? Results from a large colorectal cancer screening study’. Carcinogenesis. 39:852018. View Article : Google Scholar : PubMed/NCBI

43 

Ito M, Kanno S, Nosho K, Sukawa Y, Mitsuhashi K, Kurihara H, Igarashi H, Takahashi T, Tachibana M, Takahashi H, et al: Association of Fusobacterium nucleatum with clinical and molecular features in colorectal serrated pathway. Int J Cancer. 137:1258–1268. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Koi M, Okita Y and Carethers JM: Fusobacterium nucleatum infection in colorectal cancer: Linking inflammation, DNA mismatch repair and genetic and epigenetic alterations. J Anus Rectum Colon. 2:37–46. 2018. View Article : Google Scholar : PubMed/NCBI

45 

Zhang S, Cai S and Ma Y: Association between Fusobacterium nucleatum and colorectal cancer: Progress and future directions. J Cancer. 9:1652–1659. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Coppenhagen-Glazer S, Sol A, Abed J, Naor R, Zhang X, Han YW and Bachrach G: Fap2 of Fusobacterium nucleatum is a galactose-inhibitable adhesin involved in coaggregation, cell adhesion, and preterm birth. Infect Immun. 83:1104–1113. 2015. View Article : Google Scholar : PubMed/NCBI

47 

West NR, McCuaig S, Franchini F and Powrie F: Emerging cytokine networks in colorectal cancer. Nat Rev Immunol. 15:615–629. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Aggarwal BB, Vijayalekshmi RV and Sung B: Targeting inflammatory pathways for prevention and therapy of cancer: Short-term friend, long-term foe. Clin Cancer Res. 15:425–430. 2009. View Article : Google Scholar : PubMed/NCBI

49 

Balkwill FR and Mantovani A: Cancer-related inflammation: Common themes and therapeutic opportunities. Semin Cancer Biol. 22:33–40. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Kostic AD, Chun E, Robertson L, Glickman JN, Gallini CA, Michaud M, Clancy TE, Chung DC, Lochhead P, Hold GL, et al: Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe. 14:207–215. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Ma N, Liu Q, Hou L, Wang Y and Liu Z: MDSCs are involved in the protumorigenic potentials of GM-CSF in colitis-associated cancer. Int J Immunopathol Pharmacol. 30:152–162. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Gabrilovich DI, Ostrand-Rosenberg S and Bronte V: Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 12:253–268. 2012. View Article : Google Scholar : PubMed/NCBI

53 

Qian BZ and Pollard JW: Macrophage diversity enhances tumor progression and metastasis. Cell. 141:39–51. 2010. View Article : Google Scholar : PubMed/NCBI

54 

Gabrilovich DI and Nagaraj S: Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 9:162–174. 2009. View Article : Google Scholar : PubMed/NCBI

55 

Quah SY, Bergenholtz G and Tan KS: Fusobacterium nucleatum induces cytokine production through Toll-like-receptor-independent mechanism. Int Endod J. 47:550–559. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Dharmani P, Strauss J, Ambrose C, Allen-Vercoe E and Chadee K: Fusobacterium nucleatum infection of colonic cells stimulates MUC2 mucin and tumor necrosis factor alpha. Infect Immun. 79:2597–2607. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Trinchieri G: Cancer and inflammation: An old intuition with rapidly evolving new concepts. Annu Rev Immunol. 30:677–706. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Taniguchi K and Karin M: NF-κB, inflammation, immunity and cancer: Coming of age. Nat Rev Immunol. 18:309–324. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Fardini Y, Wang X, Témoin S, Nithianantham S, Lee D, Shoham M and Han YW: Fusobacterium nucleatum adhesin FadA binds vascular endothelial cadherin and alters endothelial integrity. Mol Microbiol. 82:1468–1480. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Han YW, Ikegami A, Rajanna C, Kawsar HI, Zhou Y, Li M, Sojar HT, Genco RJ, Kuramitsu HK and Deng CX: Identification and characterization of a novel adhesin unique to oral fusobacteria. J Bacteriol. 187:5330–5340. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Xu M, Yamada M, Li M, Liu H, Chen SG and Han YW: FadA from Fusobacterium nucleatum utilizes both secreted and nonsecreted forms for functional oligomerization for attachment and invasion of host cell. J Biol Chem. 282:25000–25009. 2007. View Article : Google Scholar : PubMed/NCBI

62 

Temoin S, Wu KL, Wu V, Shoham M and Han YW: Signal peptide of FadA adhesin from Fusobacterium nucleatum plays a novel structural role by modulating the filament's length and width. FEBS Lett. 586:1–6. 2012. View Article : Google Scholar : PubMed/NCBI

63 

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

64 

Chen Y, Peng Y, Yu J, Chen T, Wu Y, Shi L, Li Q, Wu J and Fu X: Invasive Fusobacterium nucleatum activates beta-catenin signaling in colorectal cancer via a TLR4/P-PAK1 cascade. Oncotarget. 8:31802–31814. 2017.PubMed/NCBI

65 

Wu Y, Wu J, Chen T, Li Q, Peng W, Li H, Tang X and Fu X: Fusobacterium nucleatum potentiates intestinal tumorigenesis in mice via a Toll-like receptor 4/p21-activated kinase 1 cascade. Dig Dis Sci. 63:1210–1218. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Jiang WG, Sanders AJ, Katoh M, Ungefroren H, Gieseler F, Prince M, Thompson SK, Zollo M, Spano D, Dhawan P, et al: Tissue invasion and metastasis: Molecular, biological and clinical perspectives. Semin Cancer Biol. 35 (Suppl):S244–S275. 2015. View Article : Google Scholar : PubMed/NCBI

67 

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

68 

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

69 

Shenker BJ and Datar S: Fusobacterium nucleatum inhibits human T-cell activation by arresting cells in the mid-G1 phase of the cell cycle. Infect Immun. 63:4830–4836. 1995.PubMed/NCBI

70 

Yang Y, Weng W, Peng J, Hong L, Yang L, Toiyama Y, Gao R, Liu M, Yin M, Pan C, 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. 152:851–866.e24. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Tahara T, Yamamoto E, Suzuki H, Maruyama R, Chung W, Garriga J, Jelinek J, Yamano HO, Sugai T, An B, et al: Fusobacterium in colonic flora and molecular features of colorectal carcinoma. Cancer Res. 74:1311–1318. 2014. View Article : Google Scholar : PubMed/NCBI

72 

Mima K, Nishihara R, Qian ZR, Cao Y, Sukawa Y, Nowak JA, Yang J, Dou R, Masugi Y, Song M, et al: Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut. 65:1973–1980. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Ryuk JP, Choi GS, Park JS, Kim HJ, Park SY, Yoon GS, Jun SH and Kwon YC: Predictive factors and the prognosis of recurrence of colorectal cancer within 2 years after curative resection. Ann Surg Treat Res. 86:143–151. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Cartwright TH: Treatment decisions after diagnosis of metastatic colorectal cancer. Clin Colorectal Cancer. 11:155–166. 2012. View Article : Google Scholar : PubMed/NCBI

75 

Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, et al: Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell. 170:548–563.e16. 2017. View Article : Google Scholar : PubMed/NCBI

76 

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

77 

Huang WW, Hsieh KP, Huang RY and Yang YH: Role of cyclooxygenase-2 inhibitors in the survival outcome of colorectal cancer patients: A population-based cohort study. Kaohsiung J Med Sci. 33:308–314. 2017. View Article : Google Scholar : PubMed/NCBI

78 

Williams CS, Shattuck-Brandt RL and DuBois RN: The role of COX-2 in intestinal cancer. Expert Opin Investig Drugs. 8:1–12. 1999. View Article : Google Scholar : PubMed/NCBI

79 

Waddell WR, Ganser GF, Cerise EJ and Loughry RW: Sulindac for polyposis of the colon. Am J Surg. 157:175–179. 1989. View Article : Google Scholar : PubMed/NCBI

80 

Rothwell PM, Wilson M, Elwin CE, Norrving B, Algra A, Warlow CP and Meade TW: Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials. Lancet. 376:1741–1750. 2010. View Article : Google Scholar : PubMed/NCBI

81 

Tsoi KK, Chan FC, Hirai HW and Sung JJ: Risk of gastrointestinal bleeding and benefit from colorectal cancer reduction from long-term use of low-dose aspirin: A retrospective study of 612 509 patients. J Gastroenterol Hepatol. 33:1728–1736. 2018. View Article : Google Scholar : PubMed/NCBI

82 

Johnson CC, Jankowski M, Rolnick S, Yood MU and Alford SH: Influence of NSAID use among colorectal cancer survivors on cancer outcomes. Am J Clin Oncol. 40:370–374. 2017. View Article : Google Scholar : PubMed/NCBI

83 

Sandler RS, Halabi S, Baron JA, Budinger S, Paskett E, Keresztes R, Petrelli N, Pipas JM, Karp DD, Loprinzi CL, et al: A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer. N Engl J Med. 348:883–890. 2003. View Article : Google Scholar : PubMed/NCBI

84 

Ng K, Meyerhardt JA, Chan AT, Sato K, Chan JA, Niedzwiecki D, Saltz LB, Mayer RJ, Benson AB III, Schaefer PL, et al: Aspirin and COX-2 inhibitor use in patients with stage III colon cancer. J Natl Cancer Inst. 107:3452014.PubMed/NCBI

85 

Bos CL, Kodach LL, van den Brink GR, Diks SH, van Santen MM, Richel DJ, Peppelenbosch MP and Hardwick JC: Effect of aspirin on the Wnt/beta-catenin pathway is mediated via protein phosphatase 2A. Oncogene. 25:6447–6456. 2006. View Article : Google Scholar : PubMed/NCBI

86 

Ma X, Aoki T, Tsuruyama T and Narumiya S: Definition of prostaglandin E2-EP2 signals in the colon tumor microenvironment that amplify inflammation and tumor growth. Cancer Res. 75:2822–2832. 2015. View Article : Google Scholar : PubMed/NCBI

87 

Haak BW, Lankelma JM, Hugenholtz F, Belzer C, de Vos WM and Wiersinga WJ: Long-term impact of oral vancomycin, ciprofloxacin and metronidazole on the gut microbiota in healthy humans. J Antimicrob Chemother. 74:782–786. 2019. View Article : Google Scholar : PubMed/NCBI

88 

Yu YN, Yu TC, Zhao HJ, Sun TT, Chen HM, Chen HY, An HF, Weng YR, Yu J, Li M, et al: Berberine may rescue Fusobacterium nucleatum-induced colorectal tumorigenesis by modulating the tumor microenvironment. Oncotarget. 6:32013–32026. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Bever KM and Le DT: An expanding role for immunotherapy in colorectal cancer. J Natl Compr Canc Netw. 15:401–410. 2017. View Article : Google Scholar : PubMed/NCBI

90 

Bhatt AP, Redinbo MR and Bultman SJ: The role of the microbiome in cancer development and therapy. CA Cancer J Clin. 67:326–344. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Chakravarty D, Gao J, Phillips SM, Kundra R, Zhang H, Wang J, Rudolph JE, Yaeger R, Soumerai T, Nissan MH, et al: OncoKB: A precision oncology knowledge base. JCO Precis Oncol 2017. 2017. View Article : Google Scholar

92 

Cramer P and Bresalier RS: Gastrointestinal and hepatic complications of immune checkpoint inhibitors. Curr Gastroenterol Rep. 19:32017. View Article : Google Scholar : PubMed/NCBI

93 

Nedaeinia R, Sharifi M, Avan A, Kazemi M, Nabinejad A, Ferns GA, Ghayour-Mobarhan M and Salehi R: Inhibition of microRNA-21 via locked nucleic acid-anti-miR suppressed metastatic features of colorectal cancer cells through modulation of programmed cell death 4. Tumour Biol. 39:10104283176922612017. View Article : Google Scholar : PubMed/NCBI

94 

Kumar A, Thotakura PL, Tiwary BK and Krishna R: Target identification in Fusobacterium nucleatum by subtractive genomics approach and enrichment analysis of host-pathogen protein-protein interactions. BMC Microbiol. 16:842016. View Article : Google Scholar : PubMed/NCBI

95 

Brook I and Frazier EH: Immune response to Fusobacterium nucleatum and Prevotella intermedia in the sputum of patients with acute exacerbation of chronic bronchitis. Chest. 124:832–833. 2003. View Article : Google Scholar : PubMed/NCBI

96 

Velsko IM, Chukkapalli SS, Rivera-Kweh MF, Chen H, Zheng D, Bhattacharyya I, Gangula PR, Lucas AR and Kesavalu L: Fusobacterium nucleatum alters atherosclerosis risk factors and enhances inflammatory markers with an atheroprotective immune response in ApoE(null) mice. PLoS One. 10:e01297952015. View Article : Google Scholar : PubMed/NCBI

97 

Wang HF, Li LF, Guo SH, Zeng QY, Ning F, Liu WL and Zhang G: Evaluation of antibody level against Fusobacterium nucleatum in the serological diagnosis of colorectal cancer. Sci Rep. 6:334402016. View Article : Google Scholar : PubMed/NCBI

98 

Hundt S, Haug U and Brenner H: Comparative evaluation of immunochemical fecal occult blood tests for colorectal adenoma detection. Ann Intern Med. 150:162–169. 2009. View Article : Google Scholar : PubMed/NCBI

99 

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

100 

Huang S, Yang Z, Zou D, Dong D, Liu A, Liu W and Huang L: Rapid detection of nusG and fadA in Fusobacterium nucleatum by loop-mediated isothermal amplification. J Med Microbiol. 65:760–769. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Song M, Garrett WS and Chan AT: Nutrients, foods, and colorectal cancer prevention. Gastroenterology. 148:1244–1260.e16. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Norat T, Bingham S, Ferrari P, Slimani N, Jenab M, Mazuir M, Overvad K, Olsen A, Tjønneland A, Clavel F, et al: Meat, fish, and colorectal cancer risk: The European prospective investigation into cancer and nutrition. J Natl Cancer Inst. 97:906–916. 2005. View Article : Google Scholar : PubMed/NCBI

103 

Larsson SC and Wolk A: Meat consumption and risk of colorectal cancer: A meta-analysis of prospective studies. Int J Cancer. 119:2657–2664. 2006. View Article : Google Scholar : PubMed/NCBI

104 

Magalhães B, Peleteiro B and Lunet N: Dietary patterns and colorectal cancer: Systematic review and meta-analysis. Eur J Cancer Prev. 21:15–23. 2012. View Article : Google Scholar : PubMed/NCBI

105 

David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, et al: Diet rapidly and reproducibly alters the human gut microbiome. Nature. 505:559–563. 2014. View Article : Google Scholar : PubMed/NCBI

106 

Xu Z and Knight R: Dietary effects on human gut microbiome diversity. Br J Nutr. 113 (Suppl):S1–S5. 2015. View Article : Google Scholar : PubMed/NCBI

107 

Haro C, García-Carpintero S, Rangel-Zúñiga OA, Alcalá-Díaz JF, Landa BB, Clemente JC, Pérez-Martínez P, López-Miranda J, Pérez-Jiménez F and Camargo A: Consumption of two healthy dietary patterns restored microbiota dysbiosis in obese patients with metabolic dysfunction. Mol Nutr Food Res. 61:2017. View Article : Google Scholar

108 

Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS and Sonnenburg JL: Diet-induced extinctions in the gut microbiota compound over generations. Nature. 529:212–215. 2016. View Article : Google Scholar : PubMed/NCBI

109 

Tabung FK, Liu L, Wang W, Fung TT, Wu K, Smith-Warner SA, Cao Y, Hu FB, Ogino S, Fuchs CS and Giovannucci EL: Association of dietary inflammatory potential with colorectal cancer risk in men and women. JAMA Oncol. 4:366–373. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Tabung FK, Smith-Warner SA, Chavarro JE, Wu K, Fuchs CS, Hu FB, Chan AT, Willett WC and Giovannucci EL: Development and validation of an empirical dietary inflammatory index. J Nutr. 146:1560–1570. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Liu L, Tabung FK, Zhang X, Nowak JA, Qian ZR, Hamada T, Nevo D, Bullman S, Mima K, Kosumi K, et al: Diets that promote colon inflammation associate with risk of colorectal carcinomas that contain Fusobacterium nucleatum. Clin Gastroenterol Hepatol. 16:1622–1631.e3. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 18 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang Z and Yang Z: Fusobacterium nucleatum‑positive colorectal cancer (Review). Oncol Lett 18: 975-982, 2019
APA
Yang, Z., & Yang, Z. (2019). Fusobacterium nucleatum‑positive colorectal cancer (Review). Oncology Letters, 18, 975-982. https://doi.org/10.3892/ol.2019.10433
MLA
Yang, Z., Ji, G."Fusobacterium nucleatum‑positive colorectal cancer (Review)". Oncology Letters 18.2 (2019): 975-982.
Chicago
Yang, Z., Ji, G."Fusobacterium nucleatum‑positive colorectal cancer (Review)". Oncology Letters 18, no. 2 (2019): 975-982. https://doi.org/10.3892/ol.2019.10433