Next Article in Journal
Hydration of Simple Model Peptides in Aqueous Osmolyte Solutions
Next Article in Special Issue
Interactions between the Gut Microbiome, Lung Conditions, and Coronary Heart Disease and How Probiotics Affect These
Previous Article in Journal
High Resolution Analysis of Proteome Dynamics during Bacillus subtilis Sporulation
Previous Article in Special Issue
Identification of Potential Probiotics Producing Bacteriocins Active against Listeria monocytogenes by a Combination of Screening Tools
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Probiotics on Intestinal Mucositis during Chemotherapy for Colorectal Cancer: A Comprehensive Review of Animal Studies

1
General, Visceral and Transplant Surgery, Department of Surgery, Medical University of Graz, Auenbruggerpl. 2, 8036 Graz, Austria
2
Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(17), 9347; https://doi.org/10.3390/ijms22179347
Submission received: 29 June 2021 / Revised: 26 July 2021 / Accepted: 21 August 2021 / Published: 28 August 2021
(This article belongs to the Special Issue Recent Advances and Future Perspective in Microbiota and Probiotics)

Abstract

:
Colorectal cancer (CRC) is the second most commonly diagnosed cancer in females (incidence 16.4/10,000) and the third in males (incidence 23.4/10,000) worldwide. Surgery, chemotherapy (CTx), radiation therapy (RTx), or a combined treatment of those are the current treatment modalities for primary CRC. Chemotherapeutic drug-induced gastrointestinal (GIT) toxicity mainly presents as mucositis and diarrhea. Preclinical studies revealed that probiotic supplementation helps prevent CTx-induced side effects by reducing oxidative stress and proinflammatory cytokine production and promoting crypt cell proliferation. Moreover, probiotics showed significant results in preventing the loss of body weight (BW) and reducing diarrhea. However, further clinical studies are needed to elucidate the exact doses and most promising combination of strains to reduce or prevent chemotherapy-induced side effects. The aim of this review is to overview currently available literature on the impact of probiotics on CTx-induced side effects in animal studies concerning CRC treatment and discuss the potential mechanisms based on experimental studies’ outcomes.

1. Introduction

Colorectal cancer incidence and mortality rates vary markedly around the world. According to the World Health Organization (WHO), CRC is the third most commonly diagnosed cancer in males (incidence 23.4/10,000) and the second in females (incidence 16.2/10,000) globally, with 1.8 million new cases and almost 861,000 deaths in 2020 [1]. Rates are substantially higher in males than in females. The incidence of CRC is associated with modifiable and nonmodifiable risk factors. Genetic factors, gender, age, and ethnicity comprise nonmodifiable risk factors [2,3], whereas modifiable risk factors include low level of physical activity, diet and excess BW, lifestyle, inflammation, prescription drugs, smoking, alcoholic beverages, and, as recently reported, dysbiosis in the gut [4,5,6,7].
Chemotherapy, radiation therapy, and surgery are the primary treatment modalities for different cancer types, including CRC [8]. It has been proved that the efficacy of CTx alone or in combination with RTx is the foundation for treating cancer patients, including CRC patients [9]. However, CTx-induced intestinal mucositis (IM) is a major oncological problem that has been reported in 50–80% of patients, significantly affecting patients’ quality of life [10,11]. Inflammation usually accompanies cell loss in the epithelial barrier lining the gastrointestinal tract. Clinical symptoms of IM usually include nausea, bloating, vomiting, constipation or diarrhea, and weight loss. Moreover, mucositis frequently leads to dose reduction of CTx agents or even postponement, resulting in higher mortality [12,13]. Due to preclinical trials, major progress has been made in understanding the pathophysiological mechanisms of IM [14,15].
Some studies found preventive and therapeutic capacities of specific probiotic strains in different diseases such as infectious diseases, antibiotic- or CTx-induced diarrhea, liver insufficiency, lactose intolerance, inflammatory bowel disease, irritable bowel syndrome, and cancer [16,17,18,19]. Probiotics have been shown to confer beneficial effects on CRC and CTx-induced side effects because of the direct exposure of the colon to the consumed bacteria [20,21]. Moreover, it has been reported that probiotics have different abilities such as promoting crypt cell proliferation, preventing cytokine-induced apoptosis, reducing proinflammatory cytokine production, and regulating the intestinal immune system [22,23].
The objective of this comprehensive review is to provide an overview of the currently available literature on the impact of probiotics on CTx-induced side effects in animal studies concerning CRC treatment and discuss the potential mechanisms based on experimental studies’ outcomes.

2. Materials and Methods

The literature search was performed in the PubMed, Web of Science, EMBASE, and clinicalTrials.gov online databases. The following combination of Medical Subject Headings (MeSH) and keywords with the employment of “AND” or “OR” Boolean operators were used: “Preclinical trials” OR “Gastrointestinal mucositis” OR “Chemotherapy side effects” OR “Cancer” OR “Diarrhea” OR “Colorectal cancer” OR “Intestinal microbiota” OR “Chemotherapy induced mucositis” OR “Chemotherapy induced diarrhea” OR “Chemotherapy” OR “Gut microbiota” AND “Probiotics”.
The search was restricted to English language only without a time limitation. Most recent search was performed on 17 May 2021.
At least two researchers reviewed the abstracts for the inclusion. After relevant abstracts were identified, full-text articles were retrieved and re-reviewed. Reference lists from selected studies were examined, and relevant articles included.

3. Comprehensive Review

3.1. Probiotics

Probiotics are bacteria with health benefits ingested as a supplement or food constituent that have been consumed increasingly in recent years [24]. The Food and Agriculture Organization (FAO) and World Health Organization (WHO) have defined probiotics as “live microorganisms that, when administered in adequate amounts confer a health benefit on the host” [25]. According to the current definition, the term probiotics implies alive, viable bacteria; it does not apply to dead bacterial components. Moreover, probiotics should have several certain characteristics to exert maximum therapeutic effects, including resistance to the gastrointestinal tract environment (low pH and bile salt), because bacteria must remain viable, able to adhere to the intestinal mucosa, and able to colonize the intestinal tract [26]. There are many different microorganisms currently used as probiotics, with the most common group of probiotics belonging to the lactic acid bacteria of the genera Lactobacillus and Bifidobacterium (Table 1) [27].
Compared to pathogenic bacteria, probiotics are considered safe, and infections caused by probiotics are extremely rare. Probiotics are noninvasive despite strong adherence to the intestinal epithelium. Most studies did not report a statistically significant increase in adverse events compared to control groups [28,29]. Usually, probiotic bacteria colonize the intestine only transiently without producing toxins or metabolites dangerous for the host [30]. However, there are also clinical trials from which deaths were reported [31].
Probiotics are used to improve the homeostasis of internal microbiota in order maintain intestinal health [32]. As a result, the number of harmful bacteria that cannot survive in the acidic environment decreases while the beneficial bacteria that thrive in the acidic environment proliferate, balancing the intestinal microbiota [33].
Scientific evidence supports the important role that probiotics can play in the digestive system, having significant effects in alleviating the symptoms of GIT diseases such as: irritable bowel syndrome, inflammatory bowel disease, GIT infections, constipation, food allergies, antibiotic- or CTx-induced diarrhea, and colorectal cancer [21,34]. The orally administered probiotic cocktail VSL#3 has been shown to be effective in inducing remission in patients with mild to moderate ulcerative colitis by decreasing expression of TLR-4, NF-κB, and inducible nitric oxide synthase [35,36]. In pouchitis, it appears to exert several anti-inflammatory mechanisms of action, including alteration of cytokine profile and expression of nitric oxide synthase and matrix metalloproteinases [37]. Moreover, Saccharomyces boulardii prevented relapse from active disease in patients with Crohn’s disease and infections caused by Clostridium difficile [38]. Probiotics exhibited antiproliferative and proapoptotic properties against gastrointestinal cancers [39,40]. Furthermore, Lactobacillus casei, Bifidobacterium longum, and L. acidophilus showed beneficial effects on tumor cell apoptosis [41]. Moreover, in cellular lines, it has been observed that Bifidobacterium adolescentis inhibited the proliferation of three human colon cancer cell lines including HT-29, SW 480, and Caco-2 [42]. Studies have shown that at least 107–109 viable bacteria must reach the intestine for health benefits to be achieved for the organism [26,43].

3.2. The Role of Microbiota

The gut microbiome maintains a symbiotic relationship with the gut mucosa performing specific metabolic, protective, trophic, and immunomodulatory functions in the organism. Metabolic functions, including production of vitamin K and several components of vitamin B, digestion, and fermentation of the carbohydrates that escaped proximal digestion and indigestible oligosaccharides, result in the synthesis of short-chain fatty acids (SCFA) such as butyrate, propionate, and acetate, which are rich sources of energy for the host. Protective functions are associated with degradation and prevention of the resident pathogen overgrowth, while trophic functions involve control of integrity of the intestinal epithelium and ensure immune system homeostasis [44,45].
The integrity of the intestinal barrier is a hallmark of a eubiotic intestinal ecosystem [33]. Dysbiosis, an imbalance in function or structure of gut microbiota, may be caused by extrinsic factors such as drugs, chemotherapy, radiotherapy, and poor nutrition. Intrinsic factors causing dysbiosis comprise various diseases, such as colitis, inflammatory bowel disease, obesity, and colorectal cancer [46,47].
Ingested probiotic bacteria, which are capable of colonizing the intestinal tract, are reported to restore eubiotic conditions by producing antimicrobial substances such as bacteriocins and lowering the pH in order to inhibit the growth of other pathogenic bacteria [33,48]. Generally, CTx causes a decrease in Lactobacillus, Bifidobacterium, and other protective bacteria and an increase in specific pathogenic species [49]. In addition, the beneficial probiotic microflora, dominated by Bifidobacteria and Lactobacilli, are able to modify the gut microbiota by reducing the risk of cancer following their capacity to decrease β-glucoronidase and carcinogen levels [33].
In the GIT, cancer treatment by CTx agents results in intestinal crypt apoptosis and villous atrophy that may affect the composition of luminal microbiota and increase intestinal permeability [50]. Consuming probiotic bacteria can affect the rebuilding of the epithelial barrier by modulating the expression and distribution of tight junction proteins (e.g., occluding, zonula occludens (ZO)-1) [51,52,53]. Both Bifidobacteria and Lactobacilli increase tight junction protein expression and restore intestinal permeability [54]. Some studies have shown that SCFAs, by activating 5′-adenosine monophosphate-activated protein kinase, a key agent in regulating energy metabolism in colonocytes, leads to a strengthening of the intestinal epithelial tight junctions and creation of a strong and healthy barrier [55].
The epithelial mucus layer is another protective factor; it is regulated by gut bacteria playing an essential role in protecting the host against bacterial invasion and in maintaining the integrity of the intestinal epithelium. Chemotherapy regimens have been shown to alter mucin (MUC) dynamics, potentially reducing intestinal barrier function [56]. Both in vivo and in vitro studies showed the ability of probiotics to increase Muc gene expression and enhance the secretion of mucus by goblet cells [35,57].
Probiotic bacteria may activate cytoprotective pathways in epithelial cells, counteract reactive oxygen species (ROS) displace pathogenic bacteria, interact with tight junctions, and subsequently activate the NF-κB signaling pathway to enhance mucosal integrity and ensure the development of innate immune response. Thus, it contributes to the control of intestinal homeostasis, protection of the gut against injury, promotion of tissue regeneration, maintenance of the barrier function, and eubiotic intestinal microbiota [58,59,60].

3.3. Pathogenesis of CTx-Induced Mucositis

Almost immediately after initiation of CTx, cellular damage in the intestinal villi becomes evident, whereas clinical evidence of mucositis onset is reported within 24–48 h after treatment start [61,62]. CTx is linked to a range of symptoms such as abdominal pain, diarrhea, constipation, nausea, vomiting, and anorexia. In some cases, dehydration, malnutrition, infections, and sepsis may also occur. These symptoms occur primarily because of direct mucosal damage [63,64].
The pathogenesis of mucositis involves not only the epithelium but also the cells and tissues within the submucosa. Signaling from damaged endothelium, fibroblasts, and infiltrating leukocyte cells contributes to apoptosis, loss of renewal, atrophy, and ulceration. These changes occur slowly in stratified mucosa, whereas in single layers of the small intestine, changes seem to manifest abruptly [65,66].
Animal and human studies revealed mucositis development as a five-step model, entailing complex signaling pathways: (I) An initiation phase with direct DNA injury, the formation of ROS and release of endogenous damage-associated molecular pattern molecules from injured cells of the basal epithelial layers, submucosa, and endothelium. (II) A primary damage response phase with inflammation and apoptosis. This phase starts immediately when DNA strand breaks and the generation of ROS leads to the activation of redox-sensitive transcription factors such as Wnt/β-catenin, p53, caspase-1/3, Bcl-2 and NF-κB, and their associated pathways [67,68,69,70]. The activation of NF-κB leads to the release of proinflammatory cytokines such as tumor necrosis factor (TNF-α), interleukin (IL)-6, IL-1β, IL-6, IL-1, IL-18, IL-33, and cyclooxygenase-2 (COX-2) [63,71,72,73]. The timing of histological lesions, peak tissue levels of NF-κB, and proinflammatory cytokines are different according to the CTx agent (irinotecan, methotrexate, or 5-fluorouracil (5-FU)) [71,72,74]. (III) A signaling and amplification phase, increasing inflammation and apoptosis. For instance, NF-κB activates TNF-α release, which in turn activates more NF-κB. (IV) An ulceration phase, leading to ablation of the epithelial villi, disruption of epithelial cell adhesion, and discontinuity of the epithelial barrier, promoting bacterial translocation and immune cells into lamina propria. (V) A healing phase, with epithelial cell proliferation, migration, and differentiation once chemotherapy or radiotherapy has ceased [12,58,75]. These overlapping steps might be driven by the activation of NF-κB, subsequently promoting key proinflammatory cytokines, causing further mucosal injury, and eliciting further tissue damage [76].
Moreover, the small intestine is most often affected. Different CTx agents may target different parts of the cell cycle or metabolism; their effect on intestinal integrity is consistent and characterized by enterocyte hyperplasia, decreased crypt length, blunting and fusion of intestinal villi, and increased apoptosis. Studies with CTx agents suggest that levels of TNFα, IL-1β, and IL-6 are altered in different sites of the alimentary tract prior to histological evidence of damage following CTx [77].

3.4. Effects of Probiotics on CTx Side Effects in CRC

The use of probiotics to improve safety and gastrointestinal side effects during cancer treatment has been investigated by evaluating the potential benefits of probiotics during and after CTx. Gastrointestinal toxicity is mainly related to mucosal damage by CTx, decreased colonization resistance, and alteration of the natural host microflora. Probiotics may decrease the risk and severity of CTx-related toxicity, and thus may reduce side effects associated with cancer treatment [78,79].
Probiotics were evaluated mainly in the prevention of infectious complications of CTx, weight loss, and CTx-related diarrhea. In animal models, promising results have been reported (Table 2). Preclinical trials, although using diverging study design, animal populations, and probiotic products, revealed that animals receiving probiotics before, during, and after CTx developed fewer episodes of high-grade diarrhea and proved the safety of use of probiotics.
Studies in mice and rats developing diarrhea following intraperitoneal application of 5-fluorouracil discovered that the symptoms were alleviated after treatment with multistrain probiotics containing Lactobacillus and Bifidobacterium (LaBi). All studies with a LaBi mixture showed a protective effect against weight loss compared to the 5-FU group. Average jejunal crypt depth increased significantly returning to near control levels after administration of LaBi in the CTx group. Expression of TLR2 and TLR4, TNF-α, IL-1β, IL-4, IL-6, IL-17, and IFNγ in intestinal tissue were significantly reduced after probiotic strains were given to 5-FU-treated mice [76,83,89].
Treating 5-FU-induced side effects with a single probiotic strain in mice and rat studies showed that B. infantis, B. bifidum, or L. acidophilus administration diminished the severity of intestinal damage. This led to reduced MPO activity, TNF-α expression, and IL-1β expression; it also increased GSH and IL10 concentrations, prevented the loss in BW, and reduced the occurrence of diarrhea as well as the decrease in villus height [78,84,88]. The best effect of B. infantis was observed at a dose of 109 CFU/mouse. Interestingly, after the first injection of 5-FU, B. bifidum failed to prevent the initial induction of apoptosis at 24 h. These findings suggest that B. bifidum does not prevent the induction of apoptosis but is able to suppress the secondary inflammatory responses during the progression of 5-FU-induced IM [84]. Moreover, Justino et al. measured gastric emptying and intestinal transit, revealing that L. acidophilus reversed 5-FU-induced changes in GIT motility, which enhanced intestinal transit and gastric emptying and decreased retention in the distal bowel segment [78].
Bowen et al. [80] evaluated the multistrain probiotic VSL#3 in the prevention of single intraperitoneal dose irinotecan-induced diarrhea and mucositis. Maximal protective effects of probiotics were achieved when the probiotics were given before and after chemotherapy. VSL#3 significantly reduced intestinal apoptosis, and thus helped to prevent mucosal breakdown and crypt damage. It also increased epithelial proliferation, prevented moderate or severe diarrhea, prevented weight loss, and prevented irinotecan-induced loss in goblet cell numbers.
Another study [81] underlined the activity of Saccharomyces cerevisiae in reducing the severity of diarrhea and weight loss in mice after administration of both viable and heat-killed probiotic yeast. Furthermore, only viable probiotic yeast prevented the loss of goblet cells, preserved the architecture of intestinal mucosa, and reduced mucosal inflammation. S. cerevisiae decreased oxidative stress induced by irinotecan. Most importantly, intestinal concentration of SN-38 (an active metabolite of irinotecan) remained stable under the yeast treatment, whereas lower intestinal concentrations of active SN-38 could contribute to a decrease of the chemotherapeutic efficacy of irinotecan. Sezer et al. [82] investigated the efficiency of another probiotic from the Saccharomyces genus—Saccharomyces boulardii—on irinotecan-induced diarrhea and mucosal damage in rats. In rats receiving probiotics, mucosal damage was significantly less and improvement on diarrhea was recorded.
A study by Ching-Wei Chang et al. [19] showed that CTx with FOLFOX is associated with a change in microbial diversity, and oral administration of single strain probiotic Lactobacillus rhamnosus (Lcr35) restored this compositional change. Their taxonomic analysis indicated that FOLFOX significantly increased the abundance of Firmicutes, decreased the abundance of Bacteroidetes, and increased the F/B (Firmicutes/Bacteroidetes) ratio. Furthermore, Lcr35 administration restored the crypt depth and alleviated villus height-to-crypt depth ratio in CTx-treated mice, although the levels did not reach those observed in the normal saline group. Lcr35 administration was able to restore the healthy microbiome as well as reduced the severity of diarrhea and intestinal mucositis by modulation of the proinflammatory responses with suppression of intrinsic apoptosis without affecting the antitumor effect of FOLFOX.
Hui Mi et al. [85] used the CRC rat model and showed that B. infantis administration prevented the loss of BW and the decrease in villus height, reduced the occurrence of diarrhea, and reduced the severity of intestinal damage caused by 5-FU and oxaliplatin by suppressing Th1 and Th17 responses.
Not all probiotics ameliorate side effects caused by CTx. Hanru Wang [86], Whitford [87], and Smith CL [11] investigated the effects of S. thermophilus and L. fermentum in a rat model of CTx-induced mucositis. They showed that S. thermophilus and L. fermentum only partially prevented the loss of BW and partially reduced jejunal inflammation, but neither treatment was effective at reducing structural and functional changes in the GIT.

4. Conclusions

Animal studies showed that use of probiotics may reduce different side effects of CRC CTx treatment including GIT injury, IM, weight loss, and diarrhea. IM is the main side effect after CTx in CRC. The development of mucositis involves changes in gut microbiota and activation of NF-κB. Activated NF-κB results in apoptotic signals and proinflammatory cytokine production, sequentially contributing to GIT injury, diarrhea, and weight loss. Probiotics seem to have potential capacities in prevention of CTx-induced side effects in CRC treatment by modulating the gut microbiota and proinflammatory responses with suppression of intrinsic apoptosis and appear to be a promising alternative therapeutic strategy that targets both the deregulated immune response and the intestinal dysbiosis. Further animal and human studies aiming to investigate the effective dose and combination of different probiotic strains, the effectiveness of probiotics supplementation intervention in reducing inflammatory markers, and the side effects of CTx are required.

Author Contributions

P.S. (Philipp Stiegler) was responsible for the study concept, design, and critical revision of the drafted manuscript. P.M., R.Z., P.S. (Peter Schemmer), B.L. and K.S. were responsible for the literature review, interpretation and drafting of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

5-FU5-fluorouracil
BWbody weight
COX-2cyclooxygenase-2
CRCcolorectal cancer
CTxchemotherapy
DM#1B. breve, L. acidophilus, L. casei, S. thermophilus
FAOThe Food and Agriculture Organisation
F/BFirmicutes/Bacteroidetes
FOLFOX5-fluorouracil (5-FU), leucovorin (LV) and oxaliplatin
GITgastrointestinal tract
GSHglutathione
IFNinterferon
ILinterleukin
IMintestinal mucositis
LaBiLactobacillus and Bifidobacterium
Lcr35Lactobacillus casei variety rhamnosus
MPOmyeloperoxidase
MUCmucin
NF-κBnuclear factor kappa-light-chain-enhancer
PCNAproliferating cell nuclear antigen
ROSreactive oxygen species
RTxradiotherapy
SCFAsynthesis of short chain fatty acids
SN-38 active metabolite of irinotecan
TLR toll-like receptors
TNF tumor necrosis factor
VS versus
VSL#3L. acidophilus, L. plantarum, L. casei, L. bulgaricus, B. breve, B. longum, B. infantis, S. thermophilus
WHOWorld Health Organization

References

  1. World Health Organization, International Agency for Research on Cancer (IARC), Global Cancer Observatory (GCO). Available online: https://gco.iarc.fr (accessed on 15 May 2021).
  2. Finlay A Macrae, “Macrae, F.A. Colorectal Cancer: Epidemiology, Risk Factors, and Protective Factors. Available online: https://www.uptodate.com/contents/colorectal-cancer-epidemiology-risk-factors-and-protective-factors (accessed on 17 May 2021).
  3. Ambalam, P.; Raman, M.; Purama, R.K.; Doble, M. Probiotics, prebiotics and colorectal cancer prevention. Best Pr. Res. Clin. Gastroenterol. 2016, 30, 119–131. [Google Scholar] [CrossRef]
  4. Jones-McLean, E.; Hu, J.; Greene-Finestone, L.S.; De Groh, M. A DASH dietary pattern and the risk of colorectal cancer in Canadian adults. Heal. Promot. Chronic Dis. Prev. Can. 2015, 35, 12–20. [Google Scholar] [CrossRef] [Green Version]
  5. Erdrich, J.; Zhang, X.; Giovannucci, E.; Willett, W. Proportion of colon cancer attributable to lifestyle in a cohort of US women. Cancer Causes Control. 2015, 26, 1271–1279. [Google Scholar] [CrossRef] [Green Version]
  6. de Almeida, C.V.; Camargo, M.; Russo, E.; Amedei, A. Role of diet and gut microbiota on colorectal cancer immunomodulation. World J. Gastroenterol. 2018, 25, 151–162. [Google Scholar] [CrossRef]
  7. Hofseth, L.J.; Hebert, J.R.; Chanda, A.; Chen, H.; Love, B.L.; Pena, M.M.; Murphy, E.A.; Sajish, M.; Sheth, A.; Buckhaults, P.J.; et al. Early-onset colorectal cancer: Initial clues and current views. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 352–364. [Google Scholar] [CrossRef] [PubMed]
  8. Scheer, A.; Auer, R.A.C. Surveillance after Curative Resection of Colorectal Cancer. Clin. Colon Rectal Surg. 2009, 22, 242–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Abdollahi, H.; Shiri, I.; Atashzar, M.; Sarebani, M.; Moloudi, K.; Samadian, H. Radiation protection and secondary cancer prevention using biological radioprotectors in radiotherapy. Int. J. Cancer Ther. Oncol. 2015, 3, 335. [Google Scholar] [CrossRef]
  10. Davila, M.; Bresalier, R. Gastrointestinal complications of oncologic therapy. Nat. Clin. Pr. Gastroenterol. Hepatol. 2008, 5, 682–696. [Google Scholar] [CrossRef] [PubMed]
  11. Smith, C.L.; Geier, M.S.; Yazbeck, R.; Torres, D.M.; Butler, R.N.; Howarth, G.S. Lactobacillus fermentumBR11 and Fructo-Oligosaccharide Partially Reduce Jejunal Inflammation in a Model of Intestinal Mucositis in Rats. Nutr. Cancer 2008, 60, 757–767. [Google Scholar] [CrossRef] [PubMed]
  12. Sonis, S.T. The pathobiology of mucositis. Nat. Rev. Cancer 2004, 4, 277–284. [Google Scholar] [CrossRef]
  13. Ciorba, M.A.; Hallemeier, C.L.; Stenson, W.F.; Parikh, P.J. Probiotics to prevent gastrointestinal toxicity from cancer therapy. Curr. Opin. Support. Palliat. Care 2015, 9, 157–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Lee, C.S.; Ryan, E.J.; Doherty, G.A. Gastro-intestinal toxicity of chemotherapeutics in colorectal cancer: The role of inflammation. World J. Gastroenterol. 2014, 20, 3751–3761. [Google Scholar] [CrossRef] [PubMed]
  15. Bowen, J.M.; Gibson, R.; Keefe, D.M.; Cummins, A.G. Cytotoxic chemotherapy upregulates pro-poptotic Bax and Bak in the small intestine of rats and humans. Pathology 2005, 37, 56–62. [Google Scholar] [CrossRef] [PubMed]
  16. Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The role of the microbiome in cancer development and therapy. CA A Cancer J. Clin. 2017, 67, 326–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Kato, K.; Mizuno, S.; Umesaki, Y.; Ishii, Y.; Sugitani, M.; Imaoka, A.; Otsuka, M.; Hasunuma, O.; Kurihara, R.; Iwasaki, A.; et al. Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis. Aliment. Pharmacol. Ther. 2004, 20, 1133–1141. [Google Scholar] [CrossRef] [PubMed]
  18. Girardin, M.; Seidman, E.G. Indications for the Use of Probiotics in Gastrointestinal Diseases. Dig. Dis. 2011, 29, 574–587. [Google Scholar] [CrossRef]
  19. Chang, C.-W.; Liu, C.-Y.; Lee, H.-C.; Huang, Y.-H.; Li, L.-H.; Chiau, J.-S.C.; Wang, T.-E.; Chu, C.-H.; Shih, S.-C.; Tsai, T.-H.; et al. Lactobacillus casei Variety rhamnosus Probiotic Preventively Attenuates 5-Fluorouracil/Oxaliplatin-Induced Intestinal Injury in a Syngeneic Colorectal Cancer Model. Front. Microbiol. 2018, 9, 983. [Google Scholar] [CrossRef] [PubMed]
  20. McConnell, E.L.; Liu, F.; Basit, A.W. Colonic treatments and targets: Issues and opportunities. J. Drug Target. 2009, 17, 335–363. [Google Scholar] [CrossRef]
  21. Sánchez, B.; Delgado, S.; Blanco-Míguez, A.; Lourenço, A.; Gueimonde, M.; Margolles, A. Probiotics, gut microbiota, and their influence on host health and disease. Mol. Nutr. Food Res. 2017, 61, 1600240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Kanmani, P.; Kumar, R.S.; Yuvaraj, N.; Paari, K.A.; Pattukumar, V.; Arul, V. Probiotics and Its Functionally Valuable Products—A Review. Crit. Rev. Food Sci. Nutr. 2013, 53, 641–658. [Google Scholar] [CrossRef] [PubMed]
  23. Laudanno, O.; Vasconcelos, L.; Catalana, J.; Cesolari, J. Anti-Inflammatory Effect of Bioflora Probiotic Administered Orally or Subcutaneously with Live or Dead Bacteria. Dig. Dis. Sci. 2006, 51, 2180–2183. [Google Scholar] [CrossRef]
  24. Shokryazdan, P.; Jahromi, M.F.; Liang, J.B.; Ho, Y.W. Probiotics: From Isolation to Application. J. Am. Coll. Nutr. 2017, 36, 666–676. [Google Scholar] [CrossRef]
  25. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert Consensus Document: The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  26. Hawrelak, J. Probiotics. In Textbook of Natural Medicine; Elsevier BV: Amsterdam, The Netherlands, 2013; pp. 979–994. [Google Scholar]
  27. Sharif, M.K.; Mahmood, S.; Ahsan, F. Role of Probiotics Toward the Improvement of Gut Health With Special Reference to Colorectal Cancer. In Diet, Microbiome and Health; Elsevier BV: Amsterdam, The Netherlands, 2018; pp. 35–50. [Google Scholar]
  28. Feldman, M.; Friedman, L.S.; Brandt, L.J. Probiotics and Fecal Microbiota Transplantation. In Sleisenger and Fordtran’s Gastrointestinal and Liver Disease, 10th ed.; Saunders Elsevier: Philadelphia, PA, USA, 2016; pp. 2339–2343. [Google Scholar]
  29. Hempel, S.; Newberry, S.; Ruelaz, A.; Wang, Z.; Miles, J.N.V.; Suttorp, M.J.; Johnsen, B.; Shanman, R.; Slusser, W.; Fu, N.; et al. Safety of probiotics used to reduce risk and prevent or treat disease. Évid. Rep. Assess. 2011, 1–645. [Google Scholar]
  30. Boyle, R.; Robins-Browne, R.; Tang, M.L.K. Probiotic use in clinical practice: What are the risks? Am. J. Clin. Nutr. 2006, 83, 1256–1264. [Google Scholar] [CrossRef] [PubMed]
  31. Besselink, M.G.; Van Santvoort, H.C.; Buskens, E.; Akkermans, L.M.; Gooszen, H.G. Probiotic prophylaxis in predicted severe acute pancreatitis – Authors’ reply. Lancet 2008, 372, 114. [Google Scholar] [CrossRef]
  32. Sanders, M.E.; Heimbach, J.T.; Pot, B.; Tancredi, D.J.; Lenoir-Wijnkoop, I.; Lähteenmäki-Uutela, A.; Gueimonde, M.; Bañares, S. Health claims substantiation for probiotic and prebiotic products. Gut Microbes 2011, 2, 127–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Stavropoulou, E.; Bezirtzoglou, E. Probiotics in Medicine: A Long Debate. Front. Immunol. 2020, 11, 11. [Google Scholar] [CrossRef] [PubMed]
  34. Ritchie, M.L.; Romanuk, T.N. A Meta-Analysis of Probiotic Efficacy for Gastrointestinal Diseases. PLoS ONE 2012, 7, e34938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Toumi, R.; Abdelouhab, K.; Rafa, H.; Soufli, I.; Raissi-Kerboua, D.; Djeraba, Z.; Touil-Boukoffa, C. Beneficial role of the probiotic mixture Ultrabiotique on maintaining the integrity of intestinal mucosal barrier in DSS-induced experimental colitis. Immunopharmacol. Immunotoxicol. 2013, 35, 403–409. [Google Scholar] [CrossRef] [PubMed]
  36. Bibiloni, R.; Fedorak, R.; Tannock, G.W.; Madsen, K.L.; Gionchetti, P.; Campieri, M.; De Simone, C.; Sartor, R.B. VSL#3 Probiotic-Mixture Induces Remission in Patients with Active Ulcerative Colitis. Am. J. Gastroenterol. 2005, 100, 1539–1546. [Google Scholar] [CrossRef] [PubMed]
  37. Shen, J.; Zuo, Z.; Mao, A.-P. Effect of Probiotics on Inducing Remission and Maintaining Therapy in Ulcerative Colitis, Crohnʼs Disease, and Pouchitis. Inflamm. Bowel Dis. 2014, 20, 21–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Kelesidis, T.; Pothoulakis, C. Efficacy and safety of the probiotic Saccharomyces boulardii for the prevention and therapy of gastrointestinal disorders. Ther. Adv. Gastroenterol. 2012, 5, 111–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Eslami, M.; Yousefi, B.; Kokhaei, P.; Hemati, M.; Nejad, Z.R.; Arabkari, V.; Namdar, A. Importance of probiotics in the prevention and treatment of colorectal cancer. J. Cell. Physiol. 2019, 234, 17127–17143. [Google Scholar] [CrossRef] [PubMed]
  40. Liong, M.-T. Roles of Probiotics and Prebiotics in Colon Cancer Prevention: Postulated Mechanisms and In-vivo Evidence. Int. J. Mol. Sci. 2008, 9, 854–863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Lee, J.W.; Shin, J.G.; Kim, E.H.; Kang, H.E.; Yim, I.B.; Kim, J.Y.; Joo, H.G.; Woo, H.J. Immunomodulatory and antitumor effects in vivo by the cytoplasmic fraction of Lactobacillus casei and Bifidobacterium longum. J. Veter-Sci. 2004, 5, 41–48. [Google Scholar] [CrossRef]
  42. Kim, Y.; Lee, D.; Kim, D.; Cho, J.; Yang, J.W.; Chung, M.; Kim, K.; Ha, N. Inhibition of proliferation in colon cancer cell lines and harmful enzyme activity of colon bacteria by Bifidobacterium adolescentis SPM0212. Arch. Pharmacal Res. 2008, 31, 468–473. [Google Scholar] [CrossRef]
  43. Govender, M.; Choonara, Y.; Kumar, P.; du Toit, L.; van Vuuren, S.; Pillay, V. A Review of the Advancements in Probiotic Delivery: Conventional vs. Non-conventional Formulations for Intestinal Flora Supplementation. AAPS PharmSciTech 2013, 15, 29–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Molska, M.; Reguła, J. Potential Mechanisms of Probiotics Action in the Prevention and Treatment of Colorectal Cancer. Nutrients 2019, 11, 2453. [Google Scholar] [CrossRef] [Green Version]
  45. Grover, S.; Rashmi, H.M.; Srivastava, A.K.; Batish, V.K. Probiotics for human health –new innovations and emerging trends. Gut Pathogens 2012, 4, 15. [Google Scholar] [CrossRef] [Green Version]
  46. Bajinka, O.; Tan, Y.; Abdelhalim, K.A.; Ozdemir, G.; Qiu, X. Extrinsic factors influencing gut microbes, the immediate consequences and restoring eubiosis. AMB Express 2020, 10, 1–11. [Google Scholar] [CrossRef]
  47. Jahani-Sherafat, S.; Alebouyeh, M.; Moghim, S.; Amoli, H.A.; Safaei, H.G. Role of gut microbiota in the pathogenesis of colorectal cancer; a review article. Gastroenterol. Hepatol. Bed bench 2018, 11, 101–109. [Google Scholar]
  48. Wang, Y.; Wu, Y.; Wang, Y.; Xu, H.; Mei, X.; Yu, D.; Wang, Y.; Li, W. Antioxidant Properties of Probiotic Bacteria. Nutrients 2017, 9, 521. [Google Scholar] [CrossRef] [PubMed]
  49. Bron, P.A.; Kleerebezem, M.; Brummer, R.-J.; Cani, P.D.; Mercenier, A.; Macdonald, T.T.; Garcia-Ródenas, C.L.; Wells, J.M. Can probiotics modulate human disease by impacting intestinal barrier function? Br. J. Nutr. 2017, 117, 93–107. [Google Scholar] [CrossRef] [PubMed]
  50. Melichar, B.; Zezulová, M. The significance of altered gastrointestinal permeability in cancer patients. Curr. Opin. Support. Palliat. Care 2011, 5, 47–54. [Google Scholar] [CrossRef] [PubMed]
  51. Touchefeu, Y.; Montassier, E.; Nieman, K.; Gastinne, T.; Potel, G.; Varannes, S.B.D.; Le Vacon, F.; De La Cochetière, M.F. Systematic review: The role of the gut microbiota in chemotherapy- or radiation-induced gastrointestinal mucositis - current evidence and potential clinical applications. Aliment. Pharmacol. Ther. 2014, 40, 409–421. [Google Scholar] [CrossRef]
  52. Jiang, M.; Dai, C.; Zhao, D.-H. VSL#3 probiotics regulate the intestinal epithelial barrier in�vivo and in vitro via the p38 and ERK signaling pathways. Int. J. Mol. Med. 2011, 29, 202–208. [Google Scholar] [CrossRef]
  53. Mennigen, R.; Nolte, K.; Rijcken, E.; Utech, M.; Loeffler, B.; Senninger, N.; Bruewer, M. Probiotic mixture VSL#3 protects the epithelial barrier by maintaining tight junction protein expression and preventing apoptosis in a murine model of colitis. Am. J. Physiol. Liver Physiol. 2009, 296, G1140–G1149. [Google Scholar] [CrossRef]
  54. Cinausero, M.; Aprile, G.; Ermacora, P.; Basile, D.; Vitale, M.G.; Fanotto, V.; Parisi, G.; Calvetti, L.; Sonis, S.T. New Frontiers in the Pathobiology and Treatment of Cancer Regimen-Related Mucosal Injury. Front. Pharmacol. 2017, 8, 354. [Google Scholar] [CrossRef] [Green Version]
  55. Venegas, D.P.; De La Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
  56. Prisciandaro, L.D.; Geier, M.S.; Butler, R.N.; Cummins, A.G.; Howarth, G.S. Evidence Supporting the use of Probiotics for the Prevention and Treatment of Chemotherapy-Induced Intestinal Mucositis. Crit. Rev. Food Sci. Nutr. 2011, 51, 239–247. [Google Scholar] [CrossRef] [PubMed]
  57. Otte, J.-M.; Podolsky, D.K. Functional modulation of enterocytes by gram-positive and gram-negative microorganisms. Am. J. Physiol. Liver Physiol. 2004, 286, G613–G626. [Google Scholar] [CrossRef] [Green Version]
  58. Stringer, A.M. Interaction between Host Cells and Microbes in Chemotherapy-Induced Mucositis. Nutrients 2013, 5, 1488–1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Cario, E. BACTERIAL INTERACTIONS WITH CELLS OF THE INTESTINAL MUCOSA: TOLL-LIKE RECEPTORS AND NOD2. Gut 2005, 54, 1182–1193. [Google Scholar] [CrossRef] [PubMed]
  60. van Vliet, M.J.; Harmsen, H.J.M.; de Bont, E.S.J.M.; Tissing, W.J.E. The Role of Intestinal Microbiota in the Development and Severity of Chemotherapy-Induced Mucositis. PLoS Pathog. 2010, 6, e1000879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Blijlevens, N.; Sonis, S. Palifermin (recombinant keratinocyte growth factor-1): A pleiotropic growth factor with multiple biological activities in preventing chemotherapy- and radiotherapy-induced mucositis. Ann. Oncol. 2007, 18, 817–826. [Google Scholar] [CrossRef] [PubMed]
  62. Al-Ansari, S.; Zecha, J.A.E.M.; Barasch, A.; De Lange, J.; Rozema, F.R.; Raber-Durlacher, J.E. Oral Mucositis Induced By Anticancer Therapies. Curr. Oral Heal. Rep. 2015, 2, 202–211. [Google Scholar] [CrossRef] [Green Version]
  63. Ribeiro, R.A.; Wanderley, C.W.S.; Wong, D.V.T.; Mota, J.M.; Leite, C.A.V.G.; Souza, M.H.L.P.; Cunha, F.Q.; Lima-Júnior, R.C.P. Irinotecan- and 5-fluorouracil-induced intestinal mucositis: Insights into pathogenesis and therapeutic perspectives. Cancer Chemother. Pharmacol. 2016, 78, 881–893. [Google Scholar] [CrossRef]
  64. Al-Dasooqi, N.; For The Mucositis Study Group of the Multinational Association of Supportive Care in Cancer/International Society of Oral Oncology (MASCC/ISOO); Sonis, S.T.; Bowen, J.; Bateman, E.; Blijlevens, N.; Gibson, R.; Logan, R.M.; Nair, R.; Stringer, A.; et al. Emerging evidence on the pathobiology of mucositis. Support. Care Cancer 2013, 21, 2075–2083. [Google Scholar] [CrossRef] [PubMed]
  65. Chaveli-López, B. Oral toxicity produced by chemotherapy: A systematic review. J. Clin. Exp. Dent. 2014, 6, e81–e90. [Google Scholar] [CrossRef] [Green Version]
  66. Villa, A.; Sonis, S.T. Mucositis. Curr. Opin. Oncol. 2015, 27, 159–164. [Google Scholar] [CrossRef]
  67. Sukhotnik, I.; Geyer, T.; Pollak, Y.; Mogilner, J.G.; Coran, A.G.; Berkowitz, D. The Role of Wnt/β-Catenin Signaling in Enterocyte Turnover during Methotrexate-Induced Intestinal Mucositis in a Rat. PLoS ONE 2014, 9, e110675. [Google Scholar] [CrossRef] [PubMed]
  68. Bowen, J.; On behalf of the Mucositis Study Group of the Multinational Association of Supportive Care in Cancer/International Society of Oral Oncology (MASCC/ISOO); Al-Dasooqi, N.; Bossi, P.; Wardill, H.; Van Sebille, Y.; Al-Azri, A.; Bateman, E.; Correa, M.E.; Raber-Durlacher, J.; et al. The pathogenesis of mucositis: Updated perspectives and emerging targets. Support. Care Cancer 2019, 27, 4023–4033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Wongchana, W.; Palaga, T. Direct regulation of interleukin-6 expression by Notch signaling in macrophages. Cell. Mol. Immunol. 2011, 9, 155–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Lappas, M. NOD1 and NOD2 Regulate Proinflammatory and Prolabor Mediators in Human Fetal Membranes and Myometrium via Nuclear Factor-Kappa B1. Biol. Reprod. 2013, 89, 14. [Google Scholar] [CrossRef] [PubMed]
  71. Logan, R.M.; Gibson, R.; Sonis, S.T.; Keefe, D.M. Nuclear factor-κB (NF-κB) and cyclooxygenase-2 (COX-2) expression in the oral mucosa following cancer chemotherapy. Oral Oncol. 2007, 43, 395–401. [Google Scholar] [CrossRef]
  72. Sonis, S.T. The Biologic Role for Nuclear Factor-KappaB in Disease and its Potential Involvement in Mucosal Injury Associated with Anti-neoplastic Therapy. Crit. Rev. Oral Biol. Med. 2002, 13, 380–389. [Google Scholar] [CrossRef]
  73. Logan, R.M.; Stringer, A.; Bowen, J.; Gibson, R.; Sonis, S.T.; Keefe, D.M.K. Is the pathobiology of chemotherapy-induced alimentary tract mucositis influenced by the type of mucotoxic drug administered? Cancer Chemother. Pharmacol. 2009, 63, 239–251. [Google Scholar] [CrossRef]
  74. Lalla, R.V.; Peterson, D.E. Treatment of Mucositis, Including New Medications. Cancer J. 2006, 12, 348–354. [Google Scholar] [CrossRef]
  75. Yeung, C.-Y.; Chan, W.-T.; Jiang, C.-B.; Cheng, M.-L.; Liu, C.-Y.; Chang, S.-W.; Chiau, J.-S.C.; Lee, H.-C. Correction: Amelioration of Chemotherapy-Induced Intestinal Mucositis by Orally Administered Probiotics in a Mouse Model. PLoS ONE 2015, 10, e0141402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Logan, R.M.; Stringer, A.M.; Bowen, J.M.; Gibson, R.J.; Sonis, S.T.; Keefe, D.M.K. Serum levels of NF-κB and pro-inflammatory cytokines following administration of mucotoxic drugs. Cancer Biol. Ther. 2008, 7, 1139–1145. [Google Scholar] [CrossRef] [Green Version]
  77. Bowen, J.M.; Stringer, A.M.; Gibson, R.J.; Yeoh, A.S.J.; Hannam, S.; Keefe, D.M.K. VSL#3 probiotic treatment reduces chemotherapy-induced diarrhea and weight loss. Cancer Biol. Ther. 2007, 6, 1449–1454. [Google Scholar] [CrossRef] [PubMed]
  78. Krebs, B. Prebiotic and Synbiotic Treatment before Colorectal Surgery--Randomised Double Blind Trial. Coll. Antropol. 2016, 40, 35–40. [Google Scholar] [PubMed]
  79. Mego, M.; Chovanec, J.; Vochyanova-Andrezalova, I.; Konkolovsky, P.; Mikulova, M.; Reckova, M.; Miskovska, V.; Bystricky, B.; Beniak, J.; Medvecova, L.; et al. Prevention of irinotecan induced diarrhea by probiotics: A randomized double blind, placebo controlled pilot study. Complement. Ther. Med. 2015, 23, 356–362. [Google Scholar] [CrossRef] [PubMed]
  80. Bastos, R.; Pedroso, S.; Vieira, A.; Moreira, L.; França, C.; Cartelle, C.; Arantes, R.; Generoso, S.; Cardoso, V.; Neves, M.; et al. Saccharomyces cerevisiae UFMG A-905 treatment reduces intestinal damage in a murine model of irinotecan-induced mucositis. Benef. Microbes 2016, 7, 549–557. [Google Scholar] [CrossRef] [PubMed]
  81. Sezer, A.; Usta, U.; Cicin, I. The effect of Saccharomyces boulardii on reducing irinotecan-induced intestinal mucositis and diarrhea. Med Oncol. 2008, 26, 350–357. [Google Scholar] [CrossRef] [PubMed]
  82. Huang, L.; Chiau, J.-S.C.; Cheng, M.-L.; Chan, W.-T.; Jiang, C.-B.; Chang, S.-W.; Yeung, C.-Y.; Lee, H.-C. SCID/NOD mice model for 5-FU induced intestinal mucositis: Safety and effects of probiotics as therapy. Pediatr. Neonatol. 2019, 60, 252–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Kato, S.; Hamouda, N.; Kano, Y.; Oikawa, Y.; Tanaka, Y.; Matsumoto, K.; Amagase, K.; Shimakawa, M. Probiotic Bifidobacterium bifidum G9-1 attenuates 5-fluorouracil-induced intestinal mucositis in mice via suppression of dysbiosis-related secondary inflammatory responses. Clin. Exp. Pharmacol. Physiol. 2017, 44, 1017–1025. [Google Scholar] [CrossRef] [PubMed]
  84. Mi, H.; Dong, Y.; Zhang, B.; Wang, H.; Peter, C.C.; Gao, P.; Fu, H.; Gao, Y. Bifidobacterium Infantis Ameliorates Chemotherapy-Induced Intestinal Mucositis Via Regulating T Cell Immunity in Colorectal Cancer Rats. Cell. Physiol. Biochem. 2017, 42, 2330–2341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Wang, H.; Brook, C.L.; Whittaker, A.L.; Lawrence, A.; Yazbeck, R.; Howarth, G.S. Effects ofStreptococcus thermophilusTH-4 in a rat model of doxorubicin-induced mucositis. Scand. J. Gastroenterol. 2013, 48, 959–968. [Google Scholar] [CrossRef]
  86. Whitford, E.J.; Cummins, A.G.; Butler, R.N.; Prisciandaro, L.D.; Fauser, J.K.; Yazbeck, R.; Lawrence, A.; Cheah, K.Y.; Wright, T.H.; Lymn, K.A.; et al. Effects of Streptococcus thermophilus TH-4 on intestinal mucositis induced by the chemotherapeutic agent, 5-Fluorouracil (5-FU). Cancer Biol. Ther. 2009, 8, 505–511. [Google Scholar] [CrossRef] [Green Version]
  87. Yuan, K.-T.; Yu, H.-L.; Feng, W.-D.; Chong, P.; Yang, T.; Xue, C.-L.; Yu, M.; Shi, H.-P. Bifidobacterium infantis has a beneficial effect on 5-fluorouracil-induced intestinal mucositis in rats. Benef. Microbes 2015, 6, 113–118. [Google Scholar] [CrossRef] [PubMed]
  88. Tang, Y.; Wu, Y.; Huang, Z.; Dong, W.; Deng, Y.; Wang, F.; Li, M.; Yuan, J. Administration of probiotic mixture DM#1 ameliorated 5-fluorouracil–induced intestinal mucositis and dysbiosis in rats. Nutrition 2017, 33, 96–104. [Google Scholar] [CrossRef] [PubMed]
  89. Justino, P.F.C.; Melo, L.F.M.; Nogueira, A.F.; Morais, C.M.; Mendes, W.O.; Franco, A.X.; Souza, E.P.; Ribeiro, R.A.; Souza, M.H.L.P.; Soares, P.M.G. Regulatory role of Lactobacillus acidophilus on inflammation and gastric dysmotility in intestinal mucositis induced by 5-fluorouracil in mice. Cancer Chemother. Pharmacol. 2015, 75, 559–567. [Google Scholar] [CrossRef] [PubMed]
Table 1. Microorganisms considered as probiotics.
Table 1. Microorganisms considered as probiotics.
Lactobacillus spp.Bifidobacterium spp.Other Lactic Acid BacteriaNon Lactic Acid Bacteria
L. acidophilusB. animalisStreptococcus thermophilusSaccharomyces cerevisiae
L. brevisB. adolescentisEnterococcus faeciumSaccharomyces boulardii
L. caseiB. bifidumPediococcus acidilactici
L. fermentumB. breveBacillus coagulans
L. johnsoniiB. infantis
L. lactisB. lactis
L. paracaseiB. longum
L. plantarumB. thermophilum
L. rhamnosus
L. bulgaricus
Table 2. Studies conducted on probiotic role in treating and prevention the side effect of CRC treatments in animal studies.
Table 2. Studies conducted on probiotic role in treating and prevention the side effect of CRC treatments in animal studies.
StudyMain ObjectiveNumber and Strain of AnimalsCTx RegimenProbioticsMajor Findings
Chun-Yan Yeung et al. [75]To investigate the effects and safety of probiotic
supplementation in ameliorating 5-FU-induced intestinal mucositis
72 Balb/c miceA 5-day repeated 30 mg/kg/day intraperitoneal dose of 5-FULactobacillus casei,
Lactobacillus rhamnosus, Lactobacillus acidophilus,
Bifidobacterium bifidum
(1 × 107 cfu/d) daily for 5 days
General condition: in 5-FU + probiotics group the decrease in BW was significantly less severe versus (vs) 5-FU + saline group.
Gut function: Diarrhea scores significantly lower after probiotics administration;
Histology: 5-FU + probiotics group was significantly increased jejunal villus length, restored crypts depth and increased number of goblet cells vs. 5-FU + saline;
Serum analysis: Proinflammatory cytokines TNF-α, IL-1β, IL-6 levels significantly decreased in 5-FU + probiotics group vs. 5-FU + saline;
Joanne M Bowen et al. [77]To investigate the probiotic mixture, VSL#3, for
amelioration of chemotherapy-induced diarrhea
48 female DA ratsA single intraperitoneal dose of 225 mg/kg irinotecan (CPT-11)VSL3# (3.0 × 108 cfu/d) daily for 21 days pre-treatment and 7 days post-treatmentGeneral condition: Probiotics reduced BW loss;
Gut function: Diarrhea scores significantly lower after probiotics administration;
Histology: increased crypt proliferation in irinotecan + VSL3# group combined with
an inhibition of apoptosis in both the small and large intestines
R.W.Bastos et al. [80]To evaluate the pre- or post-treatment with viable or inactivated Saccharomyces cerevisiae could prevent weight loss and intestinal lesions, and maintain integrity of the mucosal barrier in a mucositis model88 Swiss male miceA 3-day repeated 75 mg/kg/day intraperitoneal dose of irinotecanSaccharomyces cerevisiae UFMG A-905 (Sc-905) (1 × 109 cfu/d) daily, 10 days before, during and 2 days after CTxOnly post-treatment with viable Sc-905 was able to protect mice against the damage caused by CTx.
General condition: Saccharomyces cerevisiae after CTx reduced BW loss.
Gut function: yeast reduced intestinal permeability.
Histology: Irinotecan + yeast group was significantly increased jejunal villus length, prevented the decrease of goblet cells and stimulated the replication of cells in the intestinal crypts vs. Irinotecan + saline;
Oxidative stress assessment: A significant reduce in lipid peroxidation was in Irinotecan + yeast group vs. Irinotecan + saline;
Sezer A et al. [81]To investigate the efficiency of Saccharomyces
boulardii on irinotecan-induced mucosal damage and
diarrhea in rats
50 male Sprague-Dawley ratsA 4-day repeated 60 mg/kg/day intravenously dose of irinotecanSaccharomyces boulardii (800 mg/kg) daily, 3 days before, during and 3 days after CTxGeneral condition: Probiotics reduced BW loss;
Gut function: Diarrhea scores significantly lower after probiotics administration;
Histology: Irinotecan + probiotics group was significantly increased jejunal villus length and reduced mucosal edema vs. Irinotecan group;
Ching-Wei Chang et al. [19]To evaluate the effect of Lactobacillus casei variety FOLFOX-induced mucosal injury rhamnosus (Lcr35) on
48 BALB/c miceA 5-day repeated 30 and 10 mg/kg intraperitoneal 5-FU and LV. Single dose of oxaliplatin 1 mg/kg i.p. on first day Lactobacillus casei variety rhamnosus Lcr35 (1 × 103−7 cfu/d) daily, 7 days before, during and 2 days after CTx
Gut function: Diarrhea scores significantly lower in FOLFOX + Lcr35 (1 × 107 CFU/daily) group;
Histology: FOLFOX + Lcr35 (1 × 105−7 CFU/daily) groups was significantly increased jejunal villus length and restored crypts depth vs. FOLFOX group; But FOLFOX + Lcr35 at the highest dose did not significantly reduce goblet cell damage;
Imunohistochemistry: FOLFOX + Lcr35 (1 × 107 CFU/daily) significantly reduced TUNEL-positive cells, number of p65-reactive cells and BAX-positive cells in the intestine; Lcr35 did not affect the proliferative activity and caspase-8 protein expression after FOLFOX;
Lcr35 (1 × 107 CFU/daily) significantly suppressed FOLFOX-induced IL-6, TNF-α in jejunum;
Lawrence Huang et al. [82]To evaluate the safety of probiotic supplementation
and to determine the probiotic effect in response to 5-FU intestinal mucositis
36 male SCID/NOD miceA 5-day repeated 30 mg/kg/day intraperitoneal dose of 5-FULactobacillus casei variety rhamnosus Lcr35;
Lactobacillus acidophilus;
Bifidobacterium bifidum LaBi
(1 × 107 cfu/d) daily for 5 days
General condition: in 5-FU + probiotics group the decrease in BW was significantly less severe vs. 5-FU + saline group. Lac35 had stronger protective effect vs. LaBi;
Gut function: Diarrhea scores significantly lower after probiotics administration;
Histology: 5-FU + probiotics groups was significantly increased jejunal villus length and restored crypts depth vs. 5-FU + saline;
Serum analysis: both Lcr35 and LaBi significantly inhibited serum cytokines TNF-α, IL-1β, IFNγ, IL-6, IL-4, IL-10, and IL-17;
Lcr35 and LaBi potentially safe
therapeutic option with no evidence of bacteremia;
Shinichi Kato et al. [83]To evaluate the effect of Bifidobacterium bifidum on 5-FU-induced
intestinal mucositis in mice
35 male miceA 6-day repeated 50 mg/kg/day intraperitoneal dose of 5-FUBifidobacterium bifidum G9-1 (BBG9-1) (1 × 107−9 cfu/d) daily for 9 days, begining 3 days before onset of 5-FUGeneral condition: BW loss was significantly lower in 5-FU + BBG9-1 (1 × 109 CFU/mouse) group;
Gut function: Diarrhea scores significantly lower after probiotics administration;
Histology: In 5-FU + BBG9-1 (1 × 109 CFU/mouse) group was significantly increased jejunal villus length and restored crypts depth vs. 5-FU group
Cytokine and enzyme assessment: MPO, TNF-α and IL-1β levels significantly decreased in 5-FU + BBG9-1 (1 × 109 CFU/mouse) group vs. 5-FU;
Hui Mi et al. [84]To investigate the effect of Bifidobacterium infantis in attenuating the severity of chemotherapy-induced intestinal mucositis in rats with colorectal cancer30 male Sprague-Dawley ratsDimethyl hydrazine injected subcutaneously weekly for 10 weeks, and then injected with SW480 cells in rectal mucosa to create a CRC model.
On the 8th day, a 3-day repeated 75 mg/kg i.p. of 5-FU and 8 mg/kg i.p. of oxaliplatin
Bifidobacterium infantis (1 × 109 cfu/d) daily for 11 days, beginning 8 days before CTxGeneral condition: Probiotics reduced BW loss;
Gut function: Diarrhea scores significantly lower after probiotics administration;
Histology: In 5-FU and oxaliplatin + B. infantis group was significantly increased jejunal villus length and restored crypts depth vs. 5-FU + saline group;
Serum analysis: cytokines TNF-α, IL-1β, L-6 levels were significantly reduced in 5-FU and oxaliplatin + B. infantis group; B. infantis effectively attenuated chemotherapy-induced intestinal mucositis by decreasing Th1 and Th17 response;
Hanru Wang et al. [85] To investigate the effects of Streptococcus thermophilus in a rat model of mucositis induced by the
anthracycline chemotherapy drug, doxorubicin
32 female Dark Agouti ratsA single intraperitoneal dose of 20 mg/kg doxorubicinStreptococcus thermophilus TH-4 (1 × 109 cfu/mL) daily for 9 days, on day
6 received CTx
General condition: TH-4 partially prevented the loss of BW induced by doxorubicin;
Histology: TH4 failed to reduce damage of jejunum and ileum tissue: to increase villus length and restore crypts depth after doxorubicin injection;
Enzyme assessment: MPO levels significantly decreased in the jejunum in doxorubicin + TH4 group vs. doxorubicin;
Whitford et al. [86]To investigate S. thermophilus (TH-4) for their potential
to reduce the severity of 5-FU-induced
small intestinal damage in rats
45 female Dark Agouti ratsA single intraperitoneal dose of 150 mg/kg 5-FU Streptococcus thermophilus TH-4 (6 × 109 cfu/mL) live, supernatant and dead formulation daily for 6 days, on day 3 received CTx General condition: there were no significant differences in reducing BW loss after 5-FU injection + TH-4 live, supernatant or dead formulation;
Histology: 5-FU + live and supernatant TH4 significantly reduced crypt fission vs. 5-FU + skim milk; 5-FU + live TH-4 partially normalized mitotic count;
Enzyme assessment: no significant difference of MPO levels was in 5-FU + either live, dead or supernatant TH4 group vs. 5-FU + skim milk;
K.-T. Yuan et al. [87]To evaluate the beneficial effects of Bifidobacterium infantis in a rat model of intestinal mucositis induced by 5-fluorouracil30 male Sprague-Dawley ratsA single intraperitoneal dose of 150 mg/kg 5-FUBifidobacterium infantis (1 × 109 cfu/d) daily for 11 days, starting from 7 days before CTxGeneral condition: Probiotics significantly reduced BW loss;
Gut function: Diarrhea scores significantly lower after probiotics administration;
Histology: In 5-FU + B. Infantis group was significantly increased jejunal villus length vs. 5-FU group;
Imunohistochemistry: in 5-FU + B. infantis group significantly increased expression of proliferating cell nuclear antigen (PCNA), reduced expression of NF-κB vs. 5-FU group;
Cytokine and enzyme assessment: plasma cytokines TNF-α, IL-1β and MPO activity were significantly reduced in 5-FU + B. infantis vs. 5-FU group;
Yan Tang et al. [88]To evaluate the effects of a probiotic mixture, DM#1, on intestinal mucositis and dysbiosis of rats
treated with 5-fluorouracil
28 male Sprague-Dawley ratsA 5-day repeated 30 mg/kg/day intraperitoneal dose of 5-FUDM#1 (1 × 108−9) cfu/d) daily, during and 3 days after CTxGeneral condition: in 5-FU + probiotic group was significantly reduced BW loss vs. 5-FU group;
Histology: In 5-FU + DM#1 group was significantly increased ileal villus height and restored crypts depth vs. 5-FU group;
Cytokine and enzyme assessment: MPO activity, expression levels of TLR2 and TLR4 and pro-inflammatory cytokines TNF-α, IL-4, IL-6 were significantly reduced in 5-FU + DM#1 vs. 5-FU group; Increased intestinal permeability caused by 5-FU was normalized after administration of DM#1 mixture;
Justino PF et al. [89]To evaluate the effect of L. acidophilus on the inflammatory and functional outcomes of 5-FU-induced IM in mice24 male Swiss miceA single intraperitoneal dose of 450 mg/kg 5-FULactobacillus acidophilus (16 × 109 cfu/d) daily for 3 days after CTxGeneral condition: in 5-FU + probiotic group was significantly reduced BW loss vs. 5-FU group;
Gut function: slower GI transit, gastric retention and increased retention in the distal bowel segment caused by 5-FU was reversed by treatment with L. acidophilus;
Histology: In 5-FU + probiotic group was significantly increased ileal and jejunal villus height and restored crypts depth vs. 5-FU group;
Cytokine, oxidative stress and enzyme assessment: MPO activity and cytokine TNF-α, IL-1β, CXCL1 levels were significantly reduced in the jejunum and in the ileum in 5-FU + L. acidophilus vs. 5-FU group; glutatione (GSH) concentrations and anti-inflammatory cytokine IL-10 level in the jejunum and in the ileum caused by 5-FU was reduced after administration of L. acidophilus;
Smith CL et al. [11]To evaluate L. fermentum BR11 potential to decrease the severity
of 5-FU-induced small intestinal damage in rats
56 female dark agouti ratsA single intraperitoneal dose of 150 mg/kg 5-FUL. fermentum BR11 (1 × 106−9 cfu/d) daily for 9 days starting from 7 days before CTxGeneral condition: BR11 partially prevented the loss of BW induced by 5-FU;
Histology: In 5-FU + probiotic group was no significant differences in ileal and jejunal villus height and crypts depth vs. 5-FU group;
Enzyme assessment: MPO activity was significantly reduced in 5-FU + BR11 vs. 5-FU group;
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Miknevicius, P.; Zulpaite, R.; Leber, B.; Strupas, K.; Stiegler, P.; Schemmer, P. The Impact of Probiotics on Intestinal Mucositis during Chemotherapy for Colorectal Cancer: A Comprehensive Review of Animal Studies. Int. J. Mol. Sci. 2021, 22, 9347. https://doi.org/10.3390/ijms22179347

AMA Style

Miknevicius P, Zulpaite R, Leber B, Strupas K, Stiegler P, Schemmer P. The Impact of Probiotics on Intestinal Mucositis during Chemotherapy for Colorectal Cancer: A Comprehensive Review of Animal Studies. International Journal of Molecular Sciences. 2021; 22(17):9347. https://doi.org/10.3390/ijms22179347

Chicago/Turabian Style

Miknevicius, Povilas, Ruta Zulpaite, Bettina Leber, Kestutis Strupas, Philipp Stiegler, and Peter Schemmer. 2021. "The Impact of Probiotics on Intestinal Mucositis during Chemotherapy for Colorectal Cancer: A Comprehensive Review of Animal Studies" International Journal of Molecular Sciences 22, no. 17: 9347. https://doi.org/10.3390/ijms22179347

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop