Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2295 2022-11-22 11:51:44 |
2 update layout Meta information modification 2295 2022-11-23 02:44:03 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Dacrema, M.;  Ali, A.;  Ullah, H.;  Khan, A.;  Minno, A.D.;  Xiao, J.;  Martins, A.M.C.;  Daglia, M. Colorectal Cancer Prevention via Modulation of Gut Microbiota. Encyclopedia. Available online: https://encyclopedia.pub/entry/35847 (accessed on 01 July 2024).
Dacrema M,  Ali A,  Ullah H,  Khan A,  Minno AD,  Xiao J, et al. Colorectal Cancer Prevention via Modulation of Gut Microbiota. Encyclopedia. Available at: https://encyclopedia.pub/entry/35847. Accessed July 01, 2024.
Dacrema, Marco, Arif Ali, Hammad Ullah, Ayesha Khan, Alessandro Di Minno, Jianbo Xiao, Alice Maria Costa Martins, Maria Daglia. "Colorectal Cancer Prevention via Modulation of Gut Microbiota" Encyclopedia, https://encyclopedia.pub/entry/35847 (accessed July 01, 2024).
Dacrema, M.,  Ali, A.,  Ullah, H.,  Khan, A.,  Minno, A.D.,  Xiao, J.,  Martins, A.M.C., & Daglia, M. (2022, November 22). Colorectal Cancer Prevention via Modulation of Gut Microbiota. In Encyclopedia. https://encyclopedia.pub/entry/35847
Dacrema, Marco, et al. "Colorectal Cancer Prevention via Modulation of Gut Microbiota." Encyclopedia. Web. 22 November, 2022.
Colorectal Cancer Prevention via Modulation of Gut Microbiota
Edit

Colorectal cancer (CRC) is the second most frequent cause of cancer-related mortality among all types of malignancies. Sedentary lifestyles, obesity, smoking, red and processed meat, low-fiber diets, inflammatory bowel disease, and gut dysbiosis are the most important risk factors associated with CRC pathogenesis. Alterations in gut microbiota are positively correlated with colorectal carcinogenesis, as these can dysregulate the immune response, alter the gut’s metabolic profile, modify the molecular processes in colonocytes, and initiate mutagenesis. Changes in the daily diet, and the addition of plant-based nutraceuticals, have the ability to modulate the composition and functionality of the gut microbiota, maintaining gut homeostasis and regulating host immune and inflammatory responses.

dietary spices gut microbiota colorectal cancer

1. Introduction

Colorectal cancer (CRC) is the most prevalent form of carcinoma, and represents a leading component of the global health burden. Advancements in treatment methods, colonoscopy, and avoidance of risk factors, such as smoking and red meat consumption, have contributed to a decline in CRC cases over the last three decades in the United States [1][2]. However, similar declines have only been observed in developed countries [3]. Despite innovative strategies of treatment and diagnosis, CRC remains the third most common cancer and the second leading cause of mortality across the globe. In the year 2018 alone, 1.8 million new CRC cases were recorded including 881,000 deaths [4]. CRC cases may rise to 2.5 million by the year 2035 [3]. The modifiable risk factors for CRC include obesity [5], cigarette smoking [6], heavy alcohol use [7], poor diet [8], and a sedentary lifestyle [9]. The genetic contribution towards CRC is in the range of 12–35% as demonstrated in twin and family studies [10][11]. While 60–65% of cases arise sporadically without any family history of CRC [12]. This sizeable sporadic contribution to the instigation of CRC shows the significance of environmental factors, which play a large role in causing CRC [13]. Among environmental factors, infectious agents are responsible for 15 percent of all cancers [14]. Colorectal carcinogenesis is a process involving years of development, possibly taking decades. In such scenarios, early life risk factors and lifestyle modification are pertinent contributors [15]. The current rise of CRC in the young adult population in the US is alarming [2], and this supports the concept that early life risk factors provide a major impact on CRC carcinogenesis [16].
The human microflora counts around thirty trillion bacteria without considering fungi and viruses. The microbiota is not only altered by the environment but also by the relationship between the host and the symbiotic organisms [17]. The total number of microbial cells is 10 times greater than that of human somatic cells [18][19][20][21][22] and these include over 1000 different species of bacteria populating our gut. Most of these belong to the Firmicutes and Bacteroides phyla and are linked to the protection of the host, as they can produce metabolites and bioproducts promoting a protective effect against different pathologies. The dietary compounds and vitamins produced by these bacteria are considered protective elements against the infiltration of gut pathogens and the development of pathologies [23][24][25]. The impairment of the microbiota could lead to dysbiosis, and several studies sustain this link between tumorigenesis and microbiome diversity, thanks to the combination of next-generation sequencing and computational analysis [26][27][28][29][30][31]. A well-regulated microbiome is essential for maintaining the homeostasis of the metabolism and immune response, in fact, several clinical studies underline how the immunotherapeutic response could be influenced by the gut microbiome, suggesting that treatments could be enhanced or depressed according to the gut microbiota status [31][32][33][34].
Another important role of the microbiome is the recognition of the conserved regions of Gram-negative pathogenic bacteria after the production of immunoglobin G antibodies [35][36]. However, the composition and the alteration of the microbiota are also related to different host life stages and diets [37][38][39][40][41]. It is calculated that 20% of all cancers are related to dysbiosis, and with this perspective, probiotics could be used as therapeutic agents to re-establish the normal microbial environment, enhancing the immune response to counteract tumor growth. Literature data have shown that gut microbiota may provide the missing link between dietary factors and CRC incidence [42]. Some dietary components, such as saturated fats, processed carbohydrates, red meat, and ultra-processed food can affect the gut microbiota and lead to inflammation [43], and inflammation is a known factor for 20–30% of CRC cases and is acknowledged as the principal driver of tumorigenesis [44][45][46].
While chemotherapy and radiotherapy are the key approaches employed for the treatment of patients with cancer, both are associated with serious adverse events that may outweigh their therapeutic benefits in certain cases. Drug resistance is another concern that is very common for anticancer therapies and may result in failure of the treatment [47]. Nature has provided a range of preventive and therapeutic agents with the potential to fight against the most devastating chronic disorders including cancer [48][49]. Edible plants containing phytochemicals are known to alter numerous molecular pathways that may impact anticancer effects (i.e., oxidative stress, inflammatory cascade, apoptosis, epigenetic regulation, p53 signaling pathway, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) pathway, mitogen-activated protein kinases (MAPKs), proteasome pathway, insulin-like growth factor-I mediated signal transduction pathway, matrix metalloproteinases (MMPs), vascular endothelial growth factor, Hippo signaling pathway, phosphoinositide 3-kinase–protein kinase B–mammalian target of a rapamycin signaling pathway (PI3K/Akt/mTOR), cyclooxygenase-2, and the Janus kinase–signal transducer and activator of transcription signaling pathway) [50][51][52][53][54][55][56][57].
Some spices such as turmeric, black cumin, ginger, ginseng, garlic, saffron, and black pepper, are potential sources of cancer prevention owing to their natural bioactive compounds (curcumin, thymoquinone, piperine, and capsaicin) [58][59][60]. About 80% of the world population is currently relying on phytomedicine for their primary healthcare [61], in fact, these natural products are commonly considered a safer alternative for patients, if compared to systematic chemotherapeutic drugs although their scientific validity and efficacy are currently under analysis [62][63]. These spices and herbs have been used for thousands of years in small amounts thanks to their beneficial effects. In particular, curcuma, ginger, garlic, clove, chili pepper, saffron, and flaxseed seem to inhibit CRC growth thanks to their chemotherapeutic roles [58][64][65][66]. CRC development is sustained by cancer stem cells (CSC), which are self-renewal and pluripotent stem cells able to promote carcinogenesis and the formation of heterogeneous tumors [67]. Increasing evidence sustains the link between microbiota alterations and mature tumor formation. In particular, their metabolome [68] can promote pro or anti-carcinogenic actions. The preservation of the CSC is essential and mediated by several phytochemicals such as curcumin, quercetin, lycopene, cinnamic acid, resveratrol, sibilin, and epigallocatechin-3-gallate [EGCG] [69]. The main pathways involved in the regulation of the CSC phenotype are Hedgehog, Notch, and Wnt/β-catenin [70], which are modulated thanks to the colonic microbiota transformation of phytochemicals. At the same time, these substances can modify the microbiota population. Thus, the diet can change the colonic bacteria and vice versa in a triangular rapport where is involved CRC formation.

2. Gut Dysbiosis and Carcinogenesis

The maintenance of healthy gut microbiota during an individual’s lifespan, and any potential loss of diversity, is strictly connected with their diet. The progression of a disease could also involve the long-term depletion of specific groups of bacteria, which could be induced by lifestyle changes and other societal factors [37][38]. Healthy conditions are completely different from those of patients affected by dysbiosis. In the first case, the immune system can easily recognize pathogenic microbes, promoting their consequent elimination [71], most gut bacteria are non-pathogenic, and they offer an important defense role in inhibiting colonization by pathogens. The immune cells (i.e., dendritic cells, macrophages, and phagocytes) are involved in the gut microbiome and are essential for the recognition of pathogenic bacteria [72]. Healthy individuals could suffer either mild or severe issues if bacteria translocate across the epithelial mucosa. Kupffer cells may be involved, after the production of endotoxins and viable or dead bacteria. However, in the case of dysbiosis, the commensal bacteria may also spread into extra-intestinal sites and tissues. Obviously, this event can promote septic shocks, sepsis, organ failure, and death [73] over short-term periods. The dysregulation of the microbiota is associated with various pathologies, and this could be also induced using antibiotics which are known to reduce microbiotal diversity. The state of the art sustains that diabetes types 1 and 2, obesity, arthritis, Crohn’s disease, arthritis, and celiac disease are linked with the deregulation of the microbiotal metabolism and inflammation, which promotes the incidence of these pathologies [73][74][75][76][77][78].
Obviously, the microbiota is strongly involved in the absorption and metabolization of nutrients, thanks to the expression of a great number of genes, which are not expressed in our own organism. The impairment and the downregulation of these processes can promote inflammation, which may also lead to cancer in the longer-term [79][80]. The increased incidence and prevalence of cancer over recent decades are mainly due to a higher exposure to cancer-causing molecules, but also to high-fat diets, which promote dysbiosis and the inflammation process [76]. The microbial alteration could be one of the main factors, which contribute to carcinogenesis [81], in fact, different studies have supported the importance of the relationship between carcinogenesis and lifestyle. The inflammation process remains a driving force in the progression of cancer, promoting its development through the production of inflammatory cytokines [82], with microbial dysbiosis leading to increased concentrations of interleukin (IL)-1, 6, 10, and tumor necrosis factor alpha (TNF-α). The production of IL-10 is essential for the body’s elimination of cancer, in fact it is considered the most effective anti-inflammatory cytokine involved in tumorigenesis [83][84][85]. Wnt signaling is involved with NF-ĸB and MAPKs, which together can lead to an increase in oxidative stress and inhibition of apoptosis [86][87]. Animal and human studies have shown that bacteria such as Fusobacteria, Alistipes, Porphyromonadaceae, Coriobacteridae, Staphylococcaceae, Akkermansia species and Methanobacteriales are predominantly increased in CRC, while Lactobacillus, Bifidobacterium, Faecalibacterium species, Treponema, Roseburia, and Ruminococcus are known to reduce [88].
The production of toxins can also influence the tumorigenesis process, with Helicobacter pylori, Escherichia coli, and Shigella flexneri, for example, inducing double-strand DNA cuts causing apoptosis or alteration of the cell cycle [89]. Starting from E. coli, colibactin and cytolethal distengin toxins induce genomic instability, promoting breaks in the host’s DNA and tumorigenesis [87]. S. Flexneri instead produces cysteine proteases, such as virulence gene A and the inisitol phosphate phosphatase D, with the final response, in this case, being necrosis, with the development of cancer and cell death due to the degradation of the p53 gene and host damage [90]. Fusobacterium nucleatum disrupts the junction of β-catenin through the effector adhesin A (FadA); moreover, it is responsible for the production of virulence factor (Fap2) but in this case, it is through the mediation of blocks of natural killer cells (NK cells) through the binding of the NK inhibitory receptor [90][91][92]. Bacteroides fragilis produces a toxin responsible for DNA damage after the production of reactive oxygen species and hydrogen peroxide [93], the same is the case for Enterococcus fecalis, which is responsible for the production of extracellular superoxide, able to trigger mutations in host DNA [55]. Finally, Lactobacillus casei is responsible for the production of the ferrichrome siderophore, which activates c-Jun N-terminal kinase (JNK) signaling and consequent apoptosis [94].

3. Gut Microbial Alteration, Chemotherapy, and Cancer Prevention

Our gut contains trillions of microorganisms interacting with the host, and it is important to underline their essential role in bodily function. Digestion, secretion of metabolites, and the intervention of the immune system as cited above, are strictly related to the microbiota. Bacteria-free mouse models underline how dysbiosis is related to immunoglobulin A, lymphadenitis, and the absence of mucus [95][96]. Cancers very often become resistant to the drugs most used for their treatment [97][98], and unfortunately in 90% of cases, this phenomenon is responsible for the patient’s death [99][100][101]. Obviously, this problem requires attention and time to promote the development of new treatments, and the gut microbiota in particular may also influence the efficacy of antitumor therapies [102].
The negative impact of the absence of a microbiota is becoming clearer year after year, with different studies on mice treated with antibiotics underlining the efficacy of chemotherapy and immunotherapy [103]. Moreover, it is possible that the efficacy of chemotherapy treatments may be heightened under normal conditions, promoting the destruction of cancer through the intervention of T-lymphocytes and myeloid cells. The antibiotic treatments applied in certain mice studies [104] can impair the presence of bacteria and the production of cytokines, however further clinical studies are required to confirm these preliminary findings. The combination of metabolomics and metagenomics underlines the importance of the gut-brain axis [105], which regulates the composition of the gut flora through the production of neuro-hormones and hormones. The case of cyclophosphamide is particularly interesting, a chemotherapeutic drug able to promote the T-cell immune response in the presence of commensal microbiota, which translocates from the spleen to the lymph nodes promoting their anticancer effect [92][106]. It appears that Bifidobacterium can enhance dendritic cells, promoting the activation of T CD8-positive cells and enhancing the efficiency of anti-programmed death ligand (PDL-1) therapy [107]. The five-year survival rate was found to have increased by 80% for 1000 sarcoma patients treated with killed microorganism activate (Serratia and Streptococcus) [108]. The T lymphocytes associated with antigen 4 (CTLA-4) seem to have anticancer effects, promoting the production of CTLA-4 inhibitors. In the absence of CTLA-4, germ-free mice registered a positive response against cancer following an exposure to Bacteroides [109] underlying the anticancer effects of these molecules.
Only a few studies to date appear to sustain the relationship between cancer prevention and the microbiota. The production of short-chain fatty acids (SCFAs) by microbiota (i.e., Propionibacteria such as P. freudenreichii) [110][111][112] has an anti-cancer effect [113], inhibiting the deacetylases of cancer cells. Indeed, a lower concentration of butyrate is registered in cancer patients. The production of SCFAs stimulates the production of IL-18, promoting the healing process in mucosal tissues [114]. Probiotic administration also exhibits interesting effects, as it seems to trigger the immune response with an antitumor effect. Gram-negative bacteria activate TLR4 and T-cells, with Salmonella enterica, for example, appearing to be very effective against cervical cancer [115]. Finally, L. casei stimulates apoptosis in cancer cells thanks to ferricrome production, through the activation of the JNK signaling pathway [87].

References

  1. Vogelaar, I.; van Ballegooijen, M.; Schrag, D.; Boer, R.; Winawer, S.J.; Habbema, J.D.F.; Zauber, A.G. How Much Can Current Interventions Reduce Colorectal Cancer Mortality in the US? Mortality Projections for Scenarios of Risk-factor Modification, Screening, and Treatment. Cancer 2006, 107, 1624–1633.
  2. Siegel, R.L.; Miller, K.D.; Fedewa, S.A.; Ahnen, D.J.; Meester, R.G.; Barzi, A.; Jemal, A. Colorectal Cancer Statistics, 2017. CA Cancer J. Clin. 2017, 67, 177–193.
  3. Dekker, E.; Tanis, P.J.; Vleugels, J.L.; Kasi, P.M.; Wallace, M.B. Colorectal Cancer. Lancet 2019, 10207, 1467–1480.
  4. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2018, 68, 394–424.
  5. Dong, Y.; Zhou, J.; Zhu, Y.; Luo, L.; He, T.; Hu, H.; Liu, H.; Zhang, Y.; Luo, D.; Xu, S.; et al. Abdominal Obesity and Colorectal Cancer Risk: Systematic Review and Meta-Analysis of Prospective Studies. Biosci. Rep. 2017, 37, BSR20170945.
  6. Liang, P.S.; Chen, T.Y.; Giovannucci, E. Cigarette Smoking and Colorectal Cancer Incidence and Mortality: Systematic Review and Meta-analysis. Int. J. Cancer 2009, 124, 2406–2415.
  7. McNabb, S.; Harrison, T.A.; Albanes, D.; Berndt, S.I.; Brenner, H.; Caan, B.J.; Campbell, P.T.; Cao, Y.; Chang-Claude, J.; Chan, A.; et al. Meta-analysis of 16 Studies of the Association of Alcohol with Colorectal Cancer. Int. J. Cancer 2020, 146, 861–873.
  8. Zheng, X.; Hur, J.; Nguyen, L.H.; Liu, J.; Song, M.; Wu, K.; Smith-Warner, S.A.; Ogino, S.; Willett, W.C.; Chan, A.T.; et al. Comprehensive Assessment of Diet Quality and Risk of Precursors of Early-Onset Colorectal Cancer. J. Natl. Cancer Inst. 2021, 113, 543–552.
  9. de Rezende, L.F.M.; de Sá, T.H.; Markozannes, G.; Rey-López, J.P.; Lee, I.M.; Tsilidis, K.K.; Ioannidis, J.P.; Eluf-Neto, J. Physical Activity and Cancer: An Umbrella Review of the Literature Including 22 Major Anatomical Sites and 770,000 Cancer Cases. Br. J. Sports Med. 2018, 52, 826–833.
  10. Lichtenstein, P.; Holm, N.V.; Verkasalo, P.K.; Iliadou, A.; Kaprio, J.; Koskenvuo, M.; Pukkala, E.; Skytthe, A.; Hemminki, K. Environmental and Heritable Factors in the Causation of Cancer—Analyses of Cohorts of Twins from Sweden, Denmark, and Finland. N. Engl. J. Med. 2000, 343, 78–85.
  11. Graff, R.E.; Möller, S.; Passarelli, M.N.; Witte, J.S.; Skytthe, A.; Christensen, K.; Tan, Q.; Adami, H.O.; Czene, K.; Harris, J.R.; et al. Familial Risk and Heritability of Colorectal Cancer in the Nordic Twin Study of Cancer. Clin. Gastroenterol. Hepatol. 2017, 15, 1256–1264.
  12. Jasperson, K.W.; Tuohy, T.M.; Neklason, D.W.; Burt, R.W. Hereditary and Familial Colon Cancer. Gastroenterology 2010, 138, 2044–2058.
  13. Wong, S.H.; Yu, J. Gut Microbiota in Colorectal Cancer: Mechanisms of Action and Clinical Applications. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 690–704.
  14. Plummer, M.; de Martel, C.; Vignat, J.; Ferlay, J.; Bray, F.; Franceschi, S. Global Burden of Cancers Attributable to Infections in 2012: A Synthetic Analysis. Lancet Glob. Health 2016, 4, e609–e616.
  15. Hughes, L.A.; van den Brandt, P.A.; Goldbohm, R.A.; de Goeij, A.F.; de Bruïne, A.P.; van Engeland, M.; Weijenberg, M.P. Childhood and Adolescent Energy Restriction and Subsequent Colorectal Cancer Risk: Results from the Netherlands Cohort Study. Int. J. Epidemiol. 2010, 39, 1333–1344.
  16. Nimptsch, K.; Wu, K. Is Timing Important? The Role of Diet and Lifestyle during Early Life on Colorectal Neoplasia. Curr. Colorectal Cancer Rep. 2018, 14, 1–11.
  17. Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the Human Microbiome. Nutr. Rev. 2012, 70, S38–S44.
  18. Bull, M.J.; Plummer, N.T. Part 1: The Human Gut Microbiome in Health and Disease. Integr. Med. 2014, 13, 17–22.
  19. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533.
  20. Gao, R.; Gao, Z.; Huang, L.; Qin, H. Gut Microbiota and Colorectal Cancer. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 757–769.
  21. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the Gut Microbiota in Nutrition and Health. BMJ 2018, 361, k2179.
  22. Leeming, E.R.; Louca, P.; Gibson, R.; Menni, C.; Spector, T.D.; le Roy, C.I. The Complexities of the Diet-Microbiome Relationship: Advances and Perspectives. Genome Med. 2021, 13, 10.
  23. Vaishnava, S.; Behrendt, C.L.; Ismail, A.S.; Eckmann, L.; Hooper, L.V. Paneth Cells Directly Sense Gut Commensals and Maintain Homeostasis at the Intestinal Host-Microbial Interface. Proc. Natl. Acad. Sci. USA 2008, 105, 20858–20863.
  24. Belkaid, Y.; Naik, S. Compartmentalized and Systemic Control of Tissue Immunity by Commensals. Nat. Immunol. 2013, 14, 646–653.
  25. Magnúsdóttir, S.; Ravcheev, D.; de Crécy-Lagard, V.; Thiele, I. Systematic Genome Assessment of B-Vitamin Biosynthesis Suggests Co-Operation among Gut Microbes. Front. Genet. 2015, 6, 148.
  26. Grice, E.A.; Segre, J.A. The Human Microbiome: Our Second Genome. Annu. Rev. Genomics Hum. Genet. 2012, 13, 151–170.
  27. Geva-Zatorsky, N.; Sefik, E.; Kua, L.; Pasman, L.; Tan, T.G.; Ortiz-Lopez, A.; Yanortsang, T.B.; Yang, L.; Jupp, R.; Mathis, D.; et al. Mining the Human Gut Microbiota for Immunomodulatory Organisms. Cell 2017, 168, 928–943.
  28. Haber, A.L.; Biton, M.; Rogel, N.; Herbst, R.H.; Shekhar, K.; Smillie, C.; Burgin, G.; Delorey, T.M.; Howitt, M.R.; Katz, Y.; et al. A Single-Cell Survey of the Small Intestinal Epithelium. Nature 2017, 551, 333–339.
  29. Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N.; et al. Environment Dominates over Host Genetics in Shaping Human Gut Microbiota. Nature 2018, 555, 210–215.
  30. Korem, T.; Zeevi, D.; Suez, J.; Weinberger, A.; Avnit-Sagi, T.; Pompan-Lotan, M.; Matot, E.; Jona, G.; Harmelin, A.; Cohen, N.; et al. Growth Dynamics of Gut Microbiota in Health and Disease Inferred from Single Metagenomic Samples. Science (1979) 2015, 349, 1101–1106.
  31. Gopalakrishnan, V.; Helmink, B.A.; Spencer, C.N.; Reuben, A.; Wargo, J.A. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell 2018, 33, 570–580.
  32. Shindo, Y.; Hazama, S. Novel Biomarkers for Personalized Cancer Immunotherapy. Cancers 2019, 11, 1223.
  33. Picardo, S.L.; Coburn, B.; Hansen, A.R. The Microbiome and Cancer for Clinicians. Crit. Rev. Oncol. Hematol. 2019, 141, 1–12.
  34. Wilson, I.D.; Nicholson, J.K. Gut Microbiome Interactions with Drug Metabolism, Efficacy, and Toxicity. Transl. Res. 2017, 179, 204–222.
  35. Brenchley, J.M.; Douek, D.C. Microbial Translocation across the GI Tract. Annu. Rev. Immunol. 2012, 30, 149.
  36. Zeng, M.Y.; Cisalpino, D.; Varadarajan, S.; Hellman, J.; Warren, H.S.; Cascalho, M.; Inohara, N.; Núñez, G. Gut Microbiota-Induced Immunoglobulin G Controls Systemic Infection by Symbiotic Bacteria and Pathogens. Immunity 2016, 44, 647–658.
  37. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet Rapidly and Reproducibly Alters the Human Gut Microbiome. Nature 2014, 505, 559–563.
  38. Xu, Z.; Knight, R. Dietary Effects on Human Gut Microbiome Diversity. Br. J. Nutr. 2015, 113, S1–S5.
  39. Sonnenburg, E.D.; Smits, S.A.; Tikhonov, M.; Higginbottom, S.K.; Wingreen, N.S.; Sonnenburg, J.L. Diet-Induced Extinctions in the Gut Microbiota Compound over Generations. Nature 2016, 529, 212–215.
  40. Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The Role of the Microbiome in Cancer Development and Therapy. CA Cancer J. Clin. 2017, 67, 326–344.
  41. David, L.A.; Materna, A.C.; Friedman, J.; Campos-Baptista, M.I.; Blackburn, M.C.; Perrotta, A.; Erdman, S.E.; Alm, E.J. Host Lifestyle Affects Human Microbiota on Daily Timescales. Genome Biol. 2014, 15, R89.
  42. De Filippo, C.; Cavalieri, D.; di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of Diet in Shaping Gut Microbiota Revealed by a Comparative Study in Children from Europe and Rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696.
  43. Shi, Z. Gut Microbiota: An Important Link between Western Diet and Chronic Diseases. Nutrients 2019, 11, 2287.
  44. Brennan, C.A.; Garrett, W.S. Gut Microbiota, Inflammation, and Colorectal Cancer. Annu. Rev. Microbiol. 2016, 70, 395.
  45. Choi, C.H.R.; Bakir, I.A.; Hart, A.L.; Graham, T.A. Clonal Evolution of Colorectal Cancer in IBD. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 218–229.
  46. Lasry, A.; Zinger, A.; Ben-Neriah, Y. Inflammatory Networks Underlying Colorectal Cancer. Nat. Immunol. 2016, 17, 230–240.
  47. Alam, W.; Ullah, H.; Santarcangelo, C.; di Minno, A.; Khan, H.; Daglia, M.; Arciola, C.R. Micronutrient Food Supplements in Patients with Gastro-Intestinal and Hepatic Cancers. Int. J. Mol. Sci. 2021, 22, 8014.
  48. Kim, D.H.; Khan, H.; Ullah, H.; Hassan, S.T.S.; Šmejkal, K.; Efferth, T.; Mahomoodally, M.F.; Xu, S.; Habtemariam, S.; Filosa, R.; et al. MicroRNA Targeting by Quercetin in Cancer Treatment and Chemoprotection. Pharmacol. Res. 2019, 147, 104346.
  49. Shankar, E.; Kanwal, R.; Candamo, M.; Gupta, S. Dietary Phytochemicals as Epigenetic Modifiers in Cancer: Promise and Challenges. Semin. Cancer Biol. 2016, 40, 82–99.
  50. Khan, H.; Ullah, H.; Martorell, M.; Valdes, S.E.; Belwal, T.; Tejada, S.; Sureda, A.; Kamal, M.A. Flavonoids Nanoparticles in Cancer: Treatment, Prevention and Clinical Prospects. Semin. Cancer Biol. 2021, 69, 200–211.
  51. Khan, H.; Reale, M.; Ullah, H.; Sureda, A.; Tejada, S.; Wang, Y.; Zhang, Z.J.; Xiao, J. Anti-Cancer Effects of Polyphenols via Targeting P53 Signaling Pathway: Updates and Future Directions. Biotechnol. Adv. 2020, 38, 107385.
  52. Bose, S.; Banerjee, S.; Mondal, A.; Chakraborty, U.; Pumarol, J.; Croley, C.R.; Bishayee, A. Targeting the JAK/STAT Signaling Pathway Using Phytocompounds for Cancer Prevention and Therapy. Cells 2020, 9, 1451.
  53. Tewari, D.; Patni, P.; Bishayee, A.; Sah, A.N.; Bishayee, A. Natural Products Targeting the PI3K-Akt-MTOR Signaling Pathway in Cancer: A Novel Therapeutic Strategy. Semin. Cancer Biol. 2019, 80, 1–17.
  54. Moloudizargari, M.; Asghari, M.H.; Nabavi, S.F.; Gulei, D.; Berindan-Neagoe, I.; Bishayee, A.; Nabavi, S.M. Targeting Hippo Signaling Pathway by Phytochemicals in Cancer Therapy. Semin. Cancer Biol. 2020, 80, 183–194.
  55. Tewari, D.; Priya, A.; Bishayee, A.; Bishayee, A. Targeting Transforming Growth Factor-β Signalling for Cancer Prevention and Intervention: Recent Advances in Developing Small Molecules of Natural Origin. Clin. Transl. Med. 2022, 12, e795.
  56. Patra, S.; Mishra, S.R.; Behera, B.P.; Mahapatra, K.K.; Panigrahi, D.P.; Bhol, C.S.; Praharaj, P.P.; Sethi, G.; Patra, S.K.; Bhutia, S.K. Autophagy-Modulating Phytochemicals in Cancer Therapeutics: Current Evidences and Future Perspectives. Semin. Cancer Biol. 2020, 80, 205–217.
  57. Khan, H.; Ullah, H.; Castilho, P.C.M.F.; Gomila, A.S.; D’Onofrio, G.; Filosa, R.; Wang, F.; Nabavi, S.M.; Daglia, M.; Silva, A.S.; et al. Targeting NF-ΚB Signaling Pathway in Cancer by Dietary Polyphenols. Crit. Rev. Food Sci. Nutr. 2019, 60, 2790–2800.
  58. Zheng, J.; Zhou, Y.; Li, Y.; Xu, D.P.; Li, S.; Li, H.B. Spices for Prevention and Treatment of Cancers. Nutrients 2016, 8, 495.
  59. Chen, H.; Yang, H.; Fan, D.; Deng, J. The Anticancer Activity and Mechanisms of Ginsenosides: An Updated Review. eFood 2020, 1, 226–241.
  60. Jing, H. Black Garlic: Processing, Composition Change, and Bioactivity. eFood 2020, 1, 242–246.
  61. Ekor, M. The Growing Use of Herbal Medicines: Issues Relating to Adverse Reactions and Challenges in Monitoring Safety. Front. Pharmacol. 2014, 4, 177.
  62. Hossain, M.; Urbi, Z. Effect of Naphthalene Acetic Acid on the Adventitious Rooting in Shoot Cuttings of Andrographis paniculata (Burm. f.) Wall. Ex Nees: An Important Therapeutical Herb. Int. J. Agron. 2016, 2016, 1–6.
  63. Hossain, M.S.; Sharfaraz, A.; Dutta, A.; Ahsan, A.; Masud, M.A.; Ahmed, I.A.; Goh, B.H.; Urbi, Z.; Sarker, M.M.R.; Ming, L.C. A Review of Ethnobotany, Phytochemistry, Antimicrobial Pharmacology and Toxicology of Nigella sativa L. Biomed. Pharmacother. 2021, 143, 112182.
  64. DeLuca, J.A.; Garcia-Villatoro, E.L.; Allred, C.D. Flaxseed Bioactive Compounds and Colorectal Cancer Prevention. Curr. Oncol. Rep. 2018, 20, 59.
  65. Jaksevicius, A.; Carew, M.; Mistry, C.; Modjtahedi, H.; Opara, E.I. Inhibitory Effects of Culinary Herbs and Spices on the Growth of HCA-7 Colorectal Cancer Cells and Their COX-2 Expression. Nutrients 2017, 9, 1051.
  66. Narayanankutty, A. PI3K/Akt/MTOR Pathway as a Therapeutic Target for Colorectal Cancer: A Review of Preclinical and Clinical Evidence. Curr. Drug Targets 2019, 20, 1217–1226.
  67. Zheng, S.; Xin, L.; Liang, A.; Fu, Y. Cancer Stem Cell Hypothesis: A Brief Summary and Two Proposals. Cytotechnology 2013, 65, 505–512.
  68. Alexander, J.L.; Scott, A.J.; Pouncey, A.L.; Marchesi, J.; Kinross, J.; Teare, J. Colorectal Carcinogenesis: An Archetype of Gut Microbiota–Host Interaction. Ecancermedicalscience 2018, 12, 865.
  69. Scarpa, E.S.; Ninfali, P. Phytochemicals as Innovative Therapeutic Tools against Cancer Stem Cells. Int. J. Mol. Sci. 2015, 16, 15727–15742.
  70. Koury, J.; Zhong, L.; Hao, J. Targeting Signaling Pathways in Cancer Stem Cells for Cancer Treatment. Stem Cells Int. 2017, 2017, 1–10.
  71. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Reddy, D.N. Role of the Normal Gut Microbiota. World J. Gastroenterol. 2015, 21, 8787–8803.
  72. Jiao, Y.; Wu, L.; Huntington, N.D.; Zhang, X. Crosstalk between Gut Microbiota and Innate Immunity and Its Implication in Autoimmune Diseases. Front. Immunol. 2020, 11, 282.
  73. Guarner, F.; Malagelada, J.R. Gut Flora in Health and Disease. Lancet 2003, 361, 512–519.
  74. Jeyamogan, S.; Khan, N.A.; Anwar, A.; Shah, M.R.; Siddiqui, R. Cytotoxic Effects of Benzodioxane, Naphthalene Diimide, Porphyrin and Acetamol Derivatives on HeLa Cells. SAGE Open Med. 2018, 6.
  75. Jeyamogan, S.; Khan, N.A.; Siddiqui, R. Application and Importance of Theranostics in the Diagnosis and Treatment of Cancer. Arch. Med. Res. 2021, 52, 131–142.
  76. Bultman, S.J. Emerging Roles of the Microbiome in Cancer. Carcinogenesis 2014, 35, 249–255.
  77. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2015. CA Cancer J. Clin. 2015, 65, 5–29.
  78. Agrawal, B. New Therapeutic Targets for Cancer: The Interplay between Immune and Metabolic Checkpoints and Gut Microbiota. Clin. Transl. Med. 2019, 8, 23.
  79. Chen, W.; Liu, F.; Ling, Z.; Tong, X.; Xiang, C. Human Intestinal Lumen and Mucosa-Associated Microbiota in Patients with Colorectal Cancer. PLoS One 2012, 7, e39743.
  80. Wu, N.; Yang, X.; Zhang, R.; Li, J.; Xiao, X.; Hu, Y.; Chen, Y.; Yang, F.; Lu, N.; Wang, Z.; et al. Dysbiosis Signature of Fecal Microbiota in Colorectal Cancer Patients. Microb. Ecol. 2013, 66, 462–470.
  81. Khatoon, J.; Rai, R.P.; Prasad, K.N. Role of Helicobacter pylori in Gastric Cancer: Updates. World J. Gastroenterol. 2016, 8, 147.
  82. Hattori, N.; Ushijima, T. Epigenetic Impact of Infection on Carcinogenesis: Mechanisms and Applications. Genome Med. 2016, 8, 1–13.
  83. Arthur, J.C.; Perez-Chanona, E.; Mühlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal Inflammation Targets Cancer-Inducing Activity of the Microbiota. Science (1979) 2012, 338, 120–123.
  84. Nougayrède, J.P.; Homburg, S.; Taieb, F.; Boury, M.; Brzuszkiewicz, E.; Gottschalk, G.; Buchrieser, C.; Hacker, J.; Dobrindt, U.; Oswald, E. Escherichia coli Induces DNA Double-Strand Breaks in Eukaryotic Cells. Science (1979) 2006, 313, 848–851.
  85. Dennis, K.L.; Blatner, N.R.; Gounari, F.; Gounari, F. Current Status of Interleukin-10 and Regulatory T-Cells in Cancer. Curr. Opin. Oncol. 2013, 25, 637–645.
  86. Al-Hebshi, N.N.; Borgnakke, W.S.; Johnson, N.W. The Microbiome of Oral Squamous Cell Carcinomas: A Functional Perspective. Curr. Oral Health Rep. 2019, 6, 145–160.
  87. Vivarelli, S.; Salemi, R.; Candido, S.; Falzone, L.; Santagati, M.; Stefani, S.; Torino, F.; Banna, G.L.; Tonini, G.; Libra, M. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers (Basel) 2019, 11, 38.
  88. Borges-Canha, M.; Portela-Cidade, J.P.; Dinis-Ribeiro, M.; Leite-Moreira, A.F.; Pimentel-Nunes, P. Role of Colonic Microbiota in Colorectal Carcinogenesis: A Systematic Review. Rev. Esp. Enferm. Dig. 2015, 107, 659–671.
  89. Klampfer, L. Cytokines, Inflammation and Colon Cancer. Curr. Cancer Drug Targets 2011, 11, 451–464.
  90. Bergounioux, J.; Elisee, R.; Prunier, A.L.; Donnadieu, F.; Sperandio, B.; Sansonetti, P.; Arbibe, L. Calpain Activation by the Shigella flexneri Effector VirA Regulates Key Steps in the Formation and Life of the Bacterium’s Epithelial Niche. Cell Host Microbe 2012, 11, 240–252.
  91. Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum Promotes Colorectal Carcinogenesis by Modulating E-Cadherin/β-Catenin Signaling via Its FadA Adhesin. Cell Host Microbe 2013, 14, 195–206.
  92. Cheng, W.Y.; Wu, C.Y.; Yu, J. The Role of Gut Microbiota in Cancer Treatment: Friend or Foe? Gut 2013, 69, 1867–1876.
  93. Goodwin, A.C.; Shields, C.E.D.; Wu, S.; Huso, D.L.; Wu, X.; Murray-Stewart, T.R.; Hacker-Prietz, A.; Rabizadeh, S.; Woster, P.M.; Sears, C.L.; et al. Polyamine Catabolism Contributes to Enterotoxigenic Bacteroides fragilis-Induced Colon Tumorigenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 15354–15359.
  94. Konishi, H.; Fujiya, M.; Tanaka, H.; Ueno, N.; Moriichi, K.; Sasajima, J.; Ikuta, K.; Akutsu, H.; Tanabe, H.; Kohgo, Y. Probiotic-Derived Ferrichrome Inhibits Colon Cancer Progression via JNK-Mediated Apoptosis. Nat. Commun. 2016, 7, 12365.
  95. Johansson, M.E.; Jakobsson, H.E.; Holmén-Larsson, J.; Schütte, A.; Ermund, A.; Rodríguez-Piñeiro, A.M.; Arike, L.; Wising, C.; Svensson, F.; Bäckhed, F.; et al. Normalization of Host Intestinal Mucus Layers Requires Long-Term Microbial Colonization. Cell Host Microbe 2015, 18, 582–592.
  96. Spiljar, M.; Merkler, D.; Trajkovski, M. The Immune System Bridges the Gut Microbiota with Systemic Energy Homeostasis: Focus on TLRs, Mucosal Barrier, and SCFAs. Front. Immunol. 2017, 8, 1353.
  97. Wang, Q.; Rao, Y.; Guo, X.; Liu, N.; Liu, S.; Wen, P.; Li, S.; Li, Y. Oral Microbiome in Patients with Oesophageal Squamous Cell Carcinoma. Sci. Rep. 2019, 9, 19055.
  98. Nair, M.; Sandhu, S.S.; Sharma, A.K. Cancer Molecular Markers: A Guide to Cancer Detection and Management. Semin. Cancer Biol. 2018, 52, 39–55.
  99. Housman, G.; Byler, S.; Heerboth, S.; Lapinska, K.; Longacre, M.; Snyder, N.; Sarkar, S. Drug Resistance in Cancer: An Overview. Cancers (Basel) 2014, 6, 1769–1792.
  100. Rueff, J.; Rodrigues, A.S. Cancer Drug Resistance: A Brief Overview from a Genetic Viewpoint. Methods Mol. Biol. 2016, 1395, 1–18.
  101. Alfarouk, K.O.; Stock, C.-M.; Taylor, S.; Walsh, M.; Muddathir, A.K.; Verduzco, D.; Bashir, A.H.H.; Mohammed, O.Y.; Elhassan, G.O.; Harguindey, S.; et al. Resistance to Cancer Chemotherapy: Failure in Drug Response from ADME to P-Gp. Cancer Cell Int. 2015, 15, 71.
  102. Schwabe, R.F.; Jobin, C. The Microbiome and Cancer. Nat. Rev. Cancer 2013, 13, 800–812.
  103. Zitvogel, L.; Daillère, R.; Roberti, M.P.; Routy, B.; Kroemer, G. Anticancer Effects of the Microbiome and Its Products. Nat. Rev. Microbiol. 2017, 15, 465–478.
  104. Iida, N.; Dzutsev, A.; Stewart, C.; Smith, L.; Bouladoux, N.; Weingarten, R.; Molina, D.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal Bacteria Control Cancer Response to Therapy by Modulating the Tumor Microenvironment. Science (1979) 2013, 342, 967–970.
  105. Kuwahara, A.; Matsuda, K.; Kuwahara, Y.; Asano, S.; Inui, T.; Marunaka, Y. Microbiota-Gut-Brain Axis: Enteroendocrine Cells and the Enteric Nervous System Form an Interface between the Microbiota and the Central Nervous System. Biomed. Res. 2020, 41, 199–216.
  106. Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillère, R.; Hannani, D.; Enot, D.; Pfirschke, C.; Engblom, C.; Pittet, M.; et al. The Intestinal Microbiota Modulates the Anticancer Immune Effects of Cyclophosphamide. Science (1979) 2013, 342, 971–976.
  107. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Man Lei, Y.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium Promotes Antitumor Immunity and Facilitates Anti–PD-L1 Efficacy. Science (1979) 2015, 350, 1084–1089.
  108. Lin, C.; Cai, X.; Zhang, J.; Wang, W.; Sheng, Q.; Hua, H.; Zhou, X. Role of Gut Microbiota in the Development and Treatment of Colorectal Cancer. Digestion 2019, 100, 72–78.
  109. Vétizou, M.; Pitt, J.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.; Duong, C.; et al. Anticancer Immunotherapy by CTLA-4 Blockade Relies on the Gut Microbiota. Science (1979) 2015, 350, 1079–1084.
  110. Jan, G.B.A.S.; Belzacq, A.S.; Haouzi, D.; Rouault, A.; Metivier, D.; Kroemer, G.; Brenner, C. Propionibacteria Induce Apoptosis of Colorectal Carcinoma Cells via Short-Chain Fatty Acids Acting on Mitochondria. Cell Death Differ. 2002, 9, 179–188.
  111. Wei, W.; Sun, W.; Yu, S.; Yang, Y.; Ai, L. Butyrate Production from High-Fiber Diet Protects against Lymphoma Tumor. Leuk. Lymphoma 2016, 57, 2401–2408.
  112. Zhang, J.; Xia, Y.; Sun, J. Breast and Gut Microbiome in Health and Cancer. Genes Dis. 2021, 8, 581–589.
  113. Sánchez-Alcoholado, L.; Ramos-Molina, B.; Otero, A.; Laborda-Illanes, A.; Ordóñez, R.; Medina, J.A.; Gómez-Millán, J.; Queipo-Ortuño, M.I. The Role of the Gut Microbiome in Colorectal Cancer Development and Therapy Response. Cancers (Basel) 2020, 12, 1406.
  114. Salcedo, R.; Worschech, A.; Cardone, M.; Jones, Y.; Gyulai, Z.; Dai, R.M.; Wang, E.; Ma, W.; Haines, D.; O’hUigin, C.; et al. MyD88-Mediated Signaling Prevents Development of Adenocarcinomas of the Colon: Role of Interleukin 18. J. Exp. Med. 2010, 207, 1625–1636.
  115. Paavonen, J.; Naud, P.; Salmerón, J.; Wheeler, C.; Chow, S.; Apter, D.; Kitchener, H.; Castellsague, X.; Teixeira, J.; Skinner, S.; et al. Efficacy of Human Papillomavirus (HPV)-16/18 AS04-Adjuvanted Vaccine against Cervical Infection and Precancer Caused by Oncogenic HPV Types (PATRICIA): Final Analysis of a Double-Blind, Randomised Study in Young Women. Lancet 2009, 374, 301–314.
More
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 471
Entry Collection: Gastrointestinal Disease
Revisions: 2 times (View History)
Update Date: 23 Nov 2022
1000/1000
Video Production Service