PMMB 2023, 6, 1; a0000339. doi: 10.36877/pmmb.0000339 http://journals.hh-publisher.com/index.php/pmmb Review Article The Ameliorative Role of Probiotics in 5-fluorouracil Induced Intestinal Mucositis Alexandra Arcilla Xin Hui Sim1†, Jia Ying Cheam1†, Jodi Woan-Fei Law1,2, Vengadesh Letchumanan1,3, Yatinesh Kumari4, Satoshi Ogawa5, Sunny Hei Wong6, Kok-Gan Chan1,7, Loh Teng-Hern Tan1,8* Article History 1Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor Darul Ehsan, Malaysia. asim0007@student.monash.edu (AAXHS); jche0331@student.monash.edu (JYC); jodi.law1@monash.edu (JW-FL) vengadesh.letchumanan1@monash.edu (VL); kokgan@um.edu.my (K-GC) 2Next-Generation Precision Medicine and Therapeutics Research Group (NMeT), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia 3Pathogen Resistome Virulome and Diagnostic Research Group (PathRiD), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia 4Neurological Disorder and Aging Research Group (NDA), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor, Malaysia. yatinesh.kumari@monash.edu (YK) 5Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500, Selangor Darul Ehsan, Malaysia. satoshi.ogawa@monash.edu (SO) 6Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; sunny.wong@ntu.edu.sg (SHW) 7Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia 8Innovative Bioprospection Development Research Group (InBioD), Clinical School Johor Bahru, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Johor Bahru 80100, Malaysia † These authors contributed equally to this work. *Corresponding author: Loh Teng Hern Tan, Innovative Bioprospection Development Research Group (InBioD), Clinical School Johor Bahru, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Johor Bahru 80100, Malaysia; loh.teng.hern@monash.edu Received: 11 June 2023; Received in Revised Form: 11 July 2023; Accepted: 14 July 2022; Available Online: 16 July 2023 Abstract: Colorectal cancer (CRC) remains one of the top cancers in the world. Although early detection improves the survival rate to around 90%, late detection would mean the need to use chemotherapy or radiotherapy, especially if surgery is not feasible. 5-fluorouracil (5- FU) is one of the common anti-cancer drugs used in treating CRC. It is the drug that has the PMMB 2023, 6, 1; a0000339 2 of 34 greatest efficacy on CRC. Although it improves the survival rate, it has many detrimental side effects. One of these side effects is intestinal mucositis. It is described as having reduced villus height, villus atrophy, crypt fissure, abdominal discomfort, diarrhoea, and weight loss. Clinically, there is no conclusive treatment therapy for mucositis. This is possibly due to the complex mechanism of the pathobiology of intestinal mucositis that includes the production of pro-inflammatory cytokines and increased epithelial cell apoptosis. 5-FU itself is known to cause gut dysbiosis. Current studies revealed probiotics play a role in attenuating this inflammatory process of intestinal mucositis by reversing gut dysbiosis, reducing the expression of pro-inflammatory cytokines, and reducing intestinal damage. This review outlines the latest evidence supporting probiotic use in ameliorating 5-FU induced intestinal mucositis, thereby promoting good health and well-being in colorectal cancer patients receiving 5-FU chemotherapy. Keywords: Colorectal cancer; 5-fluorouracil; Intestinal mucositis; Gut dysbiosis; Probiotic; SDG 3 Good health and well-being 1. Introduction Colorectal cancer (CRC) is the second leading cause of death due to cancer, with 935,000 deaths worldwide[1–3]. The survival rate of CRC depends on the stages of cancer and when it was diagnosed[4]. Generally, 63% of CRC patients will live at least five years post- diagnosis. Early stages can have survival rates up to 90%, while for later stages, especially those diagnosed with metastatic CRC, the survival rates are only around or less than 20%[5,6]. When treating CRC, the goal is to achieve as much total removal of tumours as possible while preserving patients' quality of life and survival. Typically, like most cancers, conventional surgery or chemotherapy is the most common treatment option depending on the size, location, and extent of metastasis. Typically, most early stages CRC patients would undergo tumour resection, while those who cannot undergo surgery would undergo chemotherapy to hinder or kill cancer cells. 5-Fluorouracil (5-FU) is the most common CRC chemotherapy treatment. Currently, 5-FU is either administered as a single drug or in combination with other drugs[7]. Combination regimens available now are the FOLFOX regimen (5-FU and oxaliplatin), FOLFIRI regimen (5-FU, leucovorin, and irinotecan), FOLFOXIRI (5-Fu, oxaliplatin, and irinotecan) and others[7]. Chemotherapy prolongs survival, reduces cancer-related symptoms, and preserves general well-being in advanced CRC patients[8]. Survival rates increase by almost 20% in high-risk CRC patients that undergo chemotherapy[9,10]. Being the backbone of CRC chemotherapy, 5-FU improves overall survival and disease-free interval in stage 3 CRC[10]. 5-FU does this by inhibiting the normal function of DNA and RNA. 5-FU interferes with thymidylate synthesis and inhibits DNA synthesis during DNA replication and repair[11]. Eventually, 5-FU arrests the cell cycle and induces cell death[12]. Although 5-FU is a key drug to treat CRC, it comes with a cost. Since chemotherapy targets tissues with a high cell division rate, other normally dividing cells with the same rate of division are also affected[13]. PMMB 2023, 6, 1; a0000339 3 of 34 Repeated treatment with 5-FU, as frequently happens with chemotherapy, 5-FU causes changes to body weight, bowel movement and gut architecture. Weight loss starts around day five post-5-FU, along with diarrhoea two days before. There is around a 10% risk of developing grade three or four diarrhoea (with severe diarrhoea until requiring hospital admission or severe diarrhoea with life-threatening results) in patients that undergo the FOLFOX regimen, and it is doubled in patients receiving FOLFOXIRI[14]. Zooming into the gut's architecture, 5-FU causes villi shortening and reduced number of crypts and crypts cells. Inflammation occurs, with neutrophil infiltration (measured by myeloperoxidase activity), an increase in pro-inflammatory cytokines (PIC) and apoptosis. All these events can be categorised as intestinal mucositis (IM). A common finding is the increased crypt apoptosis and villus atrophy leading to poor nutrition, malnutrition and bacteraemia[15,16]. Having IM is correlated with the application of total parenteral nutrition, analgesics and antibiotics, which are used to decrease the side effects of the inflammation and inflammation. However, in the long run, these are coupled with side effects that impact patients' quality of life, leading to a longer hospital stay, hospital fees and increases in the risk of nosocomial infections[17]. Apart from that, IM often results in dose reduction of chemotherapeutic agents or deferral of treatment, which further increases the mortality rate[18]. All these affect patients' ability to tolerate chemotherapy, affecting their chances of survival[17]. Recognising these challenges, researchers are actively exploring natural resources to offer effective and less toxic alternatives to traditional chemotherapy[19–25], such as 5-FU in this case. The exploration of natural products, particularly those derived from microbes[26– 35], animals[36] and plants[37–40], holds promise in improving CRC treatment. While these natural products offer the potential for reduced toxicity and enhanced therapeutic effects, 5- Fu remains the mainstay of chemotherapy used when CRC is at the advanced stage or has a higher risk of recurrence[41]. Among the many therapeutic alternatives, probiotics emerge as the most promising adjuvant in complementing and enhancing the effects of chemotherapy drugs. Probiotics offer several mechanisms through the modulation of the gut microbiome, which plays a crucial role in overall health and immune function[42]. By promoting a healthy gut microbiome, probiotics may improve the body's response to chemotherapy and enhance treatment efficacy[43]. Additionally, probiotics can support gastrointestinal health, alleviate chemotherapy-induced side effects such as diarrhoea, and enhance nutrient absorption[44–46]. By leveraging the symbiotic relationship between the gut microbiome and the body's response to cancer treatment, these studies aim to enhance the effectiveness of chemotherapy while reducing its associated toxicity, ultimately improving patient outcomes and quality of life[44,47]. Gut microbiota influences the responses of a host to chemotherapeutic agents[48,49]. For instance, certain groups of gut microorganisms can enhance drug efficacy, while some promote chemoresistance and mediate chemotherapy-induced side effects[13]. One of the explanations is that chemotherapy causes ROS-mediated DNA and non-DNA damage, which then leads to bacterial translocation through the gut epithelium, hence causing an PMMB 2023, 6, 1; a0000339 4 of 34 inflammatory reaction that can lead to systemic infections and other complications[13]. Unsurprisingly, reports have shown the involvement of gut microbiota in the pathophysiology of IM. Specifically, the crucial role of GM in 5-Fu induced IM has recently been highlighted with the recent study on probiotics, showing that probiotics lessen side effects such as diarrhoea and weight loss, possibly because of restoration of altered gut microbiome composition caused by 5-FU[15]. Therefore, this review discusses the pathophysiology of IM induced by 5-FU and how GM can mediate the inflammatory process. Although most of the studies are performed in animal models, overlapping features are shown in the studies and current understanding of the human body. Key findings in these animal studies would serve as a good fundamental for future human studies. In short, GM can present as a target therapeutic approach in ameliorating chemotherapy-related side effects, especially in patients undergoing the 5-FU regimen. 2. The Role of Gut Microbiome and Their Involvement in The Pathophysiology of 5- FU Induced Intestinal Mucositis The microbiome is a huge and intricate microbial community comprising bacteria, viruses, archaea, fungi and parasites that inhabit the human body[50]. Among them, Actinobacteria, Firmicutes and Bacteroidetes are the most prevalent phyla of bacteria. There are more than 100 trillion microorganisms in the gastrointestinal tract, which serves as the site of primary communication between the microbiota, the host cells and the host immune system[51,52]. Thus, the gut microbiome (GM) is associated with many functions in the human immune, metabolic, and structural functions. However, the occurrence of gut dysbiosis, specifically the disruption of the balance of beneficial bacteria in the gut microbiome, has been consistently linked to a range of human diseases[53–62]. Recent evidence suggests that certain pathogenic gut microbiota play a role in carcinogenesis[63,64]. For example, sulfidogenic bacteria produce hydrogen sulphide which is capable of damaging DNA. It also inhibits DNA repair mechanisms, leading to extensive DNA damage[65]. These sulfidogenic bacteria include Fusobacterium, Desulfovibrio and Bilophila wadsworthia. Besides that, Gram-positive organisms such as Streptococcus bovis or currently named S. gallolyticus are positively correlated with the development of adenomas and carcinomas. Various studies show an estimated likelihood of a 6–71% chance of developing CRC in patients that had S. gallolyticus bacteremia. Infection with S. gallolyticus can occur via infectious endocarditis or the presence of S. gallolyticus in the blood[66,67]. IM is a complex process that involves many different cells, tissues and signalling molecules. The pathophysiology of IM is yet fully unravelled. Sonis[68] proposed a 5- step theory consisting of the i) initiation phase, ii) upregulation and message generating phase, iii) amplification and signalling phase, iv) ulcerative phase, and v) the healing phase. It is worth mentioning that the activators for every phase would be a good target for intervention. 5-FU induces dysbiosis in the gut, where it disturbs the delicate balance of homeostasis. Gut dysbiosis is worsened when given antibiotics. Antibiotics kill beneficial bacteria, such as those that secrete metabolites that protect the mucosa and those that influence the metabolism of 5-FU but spare pathogenic bacteria that contribute to decreasing the anti-tumour efficacy PMMB 2023, 6, 1; a0000339 5 of 34 of 5-FU[69]. With this being said, it could be thought that GM is important to humans and may play a role in preventing or treating IM. We will discuss Sonis[68] theory (summarised in Figure 1) with a focus on how the gut microbiome plays a role in this inflammatory process. GM also influences intestinal permeability (IP) and mucus layer, which are the gut's defence barrier. 2.1. Initiation Phase Based on clinical evidence, the onset of mucositis begins within 24 hours to 48 hours upon the initiation of the cancer treatment[18]. The first stage, which occurs seconds after exposure to chemotherapy, is marked by the production of reactive oxygen species (ROS), direct DNA and non-DNA damage, and immune system activation[13]. It turns out that the human gut is constantly in a steady state of low-grade inflammation. This condition is due to the Toll-like receptors (TLR) on epithelial cells and intestinal-associated immune cells. The conserved molecular motifs on bacteria are recognised by the TLRs outside the cell, activating several intracellular signalling cascades, such as the NF-κB signalling pathway[70]. TLR also maintains homeostasis by preserving the mucosal layer and preventing pathogenic bacteria from disrupting this homeostasis[70]. NF-κB is a transcription factor that upregulates PIC, chemokines and adhesion molecules such as TNF-α, IL-1β and IL-6[68,70]. NF-κB is activated by chemotherapy or radiotherapy and leads to the generation of reactive oxygen species (ROS). ROS are capable of causing tissue damage in the gut epithelium, connective tissue and even blood vessels, eventually resulting in DNA damage and cell death[68]. TLRs trigger a signalling cascade. The importance of TLR can be seen in TLR4 knocked-out mice exhibiting altered neutrophil recruitment and epithelial proliferation. It is thought that TLR4 is able to limit bacterial translocation and modulate intestinal response to injury[71]. This shows that intact TLR signalling might be crucial for the anti-inflammatory response. Whether TLR agonism confers direct protection to either the gut itself or the intestinal microbiome is still unknown. It serves as a potential target site for treating IM as TLRs are frequently exposed to bacteria parts, such as bacterial cell wall. Due to the constant exposure between TLRs and bacteria, there is always a basal activation of pro-inflammatory transcription factor NF-κB. However, commensals do not trigger an inflammatory cascade[72]. It is postulated that the epithelium sequesters the commensals, preventing TLR activation, while pathogenic bacteria, equipped with virulence factors, are able to bypass the epithelial barrier to be detected by TLR ligands on immune cells[72]. It is also shown that commensals can suppress NF-κB, such as in the case of Bacteroides thetaiotaomicron that targets active NF-κB subunit RelA complex and decrease its activation. B. thetaiotaomicron also removes NF-κB complexes formed when epithelial cells were induced with virulent Salmonella strains resulting in decreased PIC production[73]. The full mechanism of how TLRs are able to differentiate pathogenic and commensal bacteria serves as a gap in present evidence, prompting further research. PMMB 2023, 6, 1; a0000339 6 of 34 Figure 1. Pathogenic bacteria bind to Toll-like receptors (TLRs). TLRs activate IKK kinase. IKK phosphorylates NF-κB associated proteins, p50 and RelA. The phosphorylated complex is targeted for polyubiquitination, allowing degradation by proteasome, leaving only p50 and RelA. These free proteins translocate into DNA and activate pro-inflammatory gene expression. Commensal bacteria such as nonvirulent Salmonella can prevent IKK complex from being ubiquitinated, preventing degradation and incorporation into DNA. Other commensals act at the nuclear level, acting on RelA protein via transcription factor PPAR-γ. These two bind together and dampen NF-κB pro-inflammatory cascade. Besides B. thetaiotaomicron, NF-κB signalling can be suppressed directly by influencing other signalling pathways, as shown by Bifidobacterium. IL-10 is an anti- inflammatory, having NF-κB inhibitory properties. IL-10 can also block the transcription of IL-1β, IL-6, IL-8, and tumour necrosis factor (TNF-α)[74]. IL-10 maintains gut immune homeostasis by decreasing PIC concentrations at the tissue damage site[75]. In IL-10 deficient rats, Bifidobacterium infantis managed to prevent proinflammatory-associated rearrangement of tight junction (TJ) proteins and instead increase TER. The study also showed that long-term exposure to B. infantis reduces inflammation, maintains colon permeability to normal levels and reduces IFN-γ secretion[76]. IFN-γ is an important recruiter for macrophages, leading to immune cell infiltration[77]. Moreover, Faecalibacterium prausnitzii secretes an unknown molecule that promotes IL-10 production[78]. Short-chain fatty acids (SCFA) such as butyrates by F. prausnitzii can also inhibit NF-κB and IL-2[79,80]. Butyrates are the main products excreted by microbes during the fermentation of dietary fibre (also known as prebiotics). It is found that butyrate suppresses inflammation by reducing TNF-α, IFN-γ, and IL-12 and prevents carcinogenesis by strengthening the mucosal barrier and modulating oxidative stress[81,82]. Probiotic supernatants have immune-modulating properties as well. Supernatant of Bacillus coagulans directly increases IL-10 production, which leads to reduced ROS production[11]. Supernatant of Escherichia coli Nissle 1917 (EcN) was anti-inflammatory in rats with colitis, reducing TNF-α and increasing IL-10[83]. In short, probiotic is capable of downregulating tissue-damaging ROS by targeting upstream PMMB 2023, 6, 1; a0000339 7 of 34 factors, particularly NF-κB. The exact target to pinpoint in this pathway could be key in reducing IM. 2.2. Upregulation and Message-Generating Phase After the initiation phase, begins the upregulation and message-generating phase. DNA damage and ROS production activate NF-κB similar to the initiation phase. TNF-α is also activated, leading to epithelial cell apoptosis. NF-κB can transcribe up to 200 genes, most of which are toxic to the gut mucosa. For example, pro-apoptotic proteins are produced by the upregulation of Bcl2 genes and BAX genes. The overall upregulation of genes such as Bcl2 and BAX genes hastens mucosal damage. NF-κB also can increase PIC production in the mucosa. These cytokines damage the connective tissues and endothelium by initiating mesenchymal epithelial signaling and reducing oxygen concentrations in the epithelium resulting in epithelial cell death and injury[68]. This process causes thinning of the mucosa, making it erythematous. This condition can be ameliorated by supplementing with lactic acid bacteria (LAB) such as Bifidobacterium breve. LAB protects against inflammation by releasing antimicrobial peptides, IgA. Its presence itself competes with pathogens to adhere to the gut epithelium. LAB have a stronger adherence to epithelial cells by increasing the expression of binding proteins. Besides that, LABs are able to secrete antioxidant enzymes as well[84]. Another study showed Streptococcus thermophilus metabolites can cross the mucosa and suppress TNF-α[85]. 2.3. Amplification and Signalling Phase The third phase is the amplification and signalling phase. There is positive feedback by the mediators released in previous phases that magnify other biological factors. Chemotherapy such as 5-Fu can activate NF-κB (a transcription factor), such as found in the gut's epithelial, endothelial, and mesenchymal cells. NF-κB in macrophages can be activated as well. Activation of NF-κB leads to the production of PIC such as TNF-α and interleukin- 1β (IL-1β). They participate in positive feedback, further activating NF-κB. NF-κB upregulates transcription of mitogen-activated protein kinase (MAPK), cyclooxygenase 2 (COX2) and tyrosine-kinase signalling molecules. These biological factors gather in the mucosa and target the tissue. For example, increased TNF-α expression has positive feedback on NF-κB activity. TNF-α can activate caspase 3 (pro-apoptotic protein), which causes cell apoptosis. Upregulation of MAPK also can lead to activation of pro-apoptotic protein caspase 3. Additionally, upregulation of MAPK, COX2 and tyrosine-kinase leads to the activation of matrix metalloproteinases (MMPs) 1 and 3 present in epithelial cells and the lamina propria. MMPs ultimately cause damage to the gut mucosa[86]. Clinically, the gut mucosa may seem erythematous, and patients can manifest very few signs and symptoms while still in the early phases of inflammation[68]. PMMB 2023, 6, 1; a0000339 8 of 34 2.4. Ulcerative Phase The gut barrier consists of the mucus layer, TJ and epithelial cells. Disruption of any of these can impair the barrier. The mucus layer, secreted by goblet cells, provides a physical barrier around the mucosa lining. TJ acts as a seal between epithelial cells comprising a bunch of proteins such as occludin and junctional adhesion molecules. TJ interspersed between epithelial cells is a physical barrier to prevent unwanted substances such as pathogenic bacteria, toxins, or byproducts from penetrating the mucosa. Zonula occludins (ZO-1) tighten the TJ seal at the cell junction[87]. 5-FU decreases both TJ and ZO-1 expression[88]. Upstream inflammatory factors like TNF-α (through NF-κB activation) and IL-1β exacerbate this cycle by down-regulating ZO-1 proteins and altering the localisation of ZO-1 proteins[89,90]. Instead of the normal cohesive binding of ZO-1, it becomes gaplike and zig-zag in appearance. Disruption in both TJ and ZO-1 leads to a penetrable mucosal layer, leading to a more permeable intestine[89]. Besides the connection between epithelial cells, epithelial cells themselves are damaged. Cell apoptosis and villous atrophy occur, forming crypts. Crypts serve as a harbour for pathogenic bacteria to grow on. Crypts also leave nerve endings in the lamina propria exposed, making this phase painful for patients. Besides losing the mucus layer, the epithelial barrier itself is damaged. The gut mucosa is very permeable in this phase, allowing bacteria to translocate across this barrier. The 'leaky' gut mucosa puts patients at risk of developing bacteraemia and sepsis. Direct exposure of the gut to pathogenic bacteria causes the human body to recognise foreign bacteria and start another cascade of inflammatory processes. Mononuclear infiltrates by chemotaxis lead to more inflammation, possibly promoting pro- apoptotic gene expression and PIC, further potentiating further tissue injury (Figure 2)[68]. Figure 2. The gut mucosa is protected by one layer of epithelial cells with tight junction interspersed in between cells. This barrier is also reinforced with a mucus layer. Bacteria bind to Toll-like receptors (TLRs). The binding activates NF-κB. NF-κB causes an upregulation of PIC. Bacteria are also phagocytosed by dendritic cells. Dendritic cells present bacteria parts to immunoglobins, subsequently setting off an inflammatory response as well. PMMB 2023, 6, 1; a0000339 9 of 34 Regulating TJ and ZO-1 expression is vital to the progression of intestinal permeability. With the administration of Bifidobacteria, there was increased expression TJ forming proteins and decreased intestinal permeability[76]. Lactobacilli also has been shown to elevate TJ protein expression and restore intestinal permeability[91–94]. Probiotic strains such as E. coli Nissle 1917 and L. rhamnosus GG upregulates ZO-1, protecting the gut from being more permeable[95,96]. Upregulation of ZO-1 is important as it protects the epithelial cells from damage such as those caused by pathogenic bacteria like Escherichia coli O157:H7[96]. SCFA produced by bacteria is able to reduce IP and increase cell viability[81]. It also helps modulate the intracellular environment, maintaining the intestinal structure, function and integrity and promoting gene expression and proliferation of the epithelial cells[97]. Butyrate, propionate and acetate are usually produced due to the fermentation of undigested carbohydrates or prebiotics by B. breve and Bacteroides ovatus[98]. 2.5. Healing Phase The last phase is the healing phase. Gut mucosa self-heals itself once the causative factor is removed. In this phase, the extracellular matrix (ECM) releases molecules which promote the healing of the ulcer. First, the molecules migrate from ECM to the ulcer border. Then, migration of the cells occurs to cover the ulcer. Then these cells begin to differentiate into epithelial cells. Normal architecture is then restored. Other molecules that promote hyperplasia are downregulated. Once the ulcer has healed, symptoms of IM begin to resolve[68]. It is known that gut dysbiosis with increased numbers of pathogenic bacteria on an exposed intestinal surface can activate more TLRs. Meanwhile, chemotherapy activates TLRs via NF-κB[70]. It turns out the steady-state inflammation by TLRs and commensal bacteria is proven to allow the repair of the epithelium. Commensal bacteria help to induce the recovery of the gut mucosa. For instance, in a study on chicks, migration of the cells is faster by more than half the time of those in germ-free chicks[99,100]. This allows faster healing of the gut epithelium. In this case, TLR is also significant because it can sense injured or dead cells as TLR recognises and binds to heat shock protein (HSP) produced by dying epithelial cells[70,71]. Cytoprotective effects of HSP are suppressed in TLR knockout mice. For example, a finding indicates that mice deficient in TLR (specifically TLR4) could not repair damaged cells. They experienced more severe damage in the epithelium, with decreased epithelial cell proliferation. TLR4 knockout mice show impaired TLR4 and HSP signalling, delaying response to dead or dying epithelial cells and leading to delayed epithelial healing[71]. As seen before, butyrate is a key player. Butyrate promotes the migration of epithelial cells, thus enhancing mucosal healing[81,101]. Other bacterial products, such as the peptides (p75 and p40) secreted by L. rhamnosus GG, prevent epithelial cell apoptosis and stimulate cell growth, hence stimulating the repair of the mucosal layer[102]. L. lactis NZ900 producing pancreatitis-associated protein I (PAP) also stimulates epithelial cell growth, including Paneth cells. PAP alters the expression of growth factors and proliferation-related molecules PMMB 2023, 6, 1; a0000339 10 of 34 along with antioxidant enzymes[103]. Figure 3 summarises the 5 phases of mucositis induced by 5-FU. Figure 3. The 5 phases of mucositis. The initiation phase begins with the exposure of gut mucosa to chemotherapy or radiotherapy. Here, we show exposure of the gut to 5-fluorouracil. Reactive oxygen species (ROS) are then formed, promoting DNA damage. ROS activates NF-κB, a transcription factor that subsequently activates PIC, such as interleukin 6 (IL-6), interleukin 8 (IL-8), IL-1β, TNF-α, and cyclooxygenase 2 (COX-2). DNA damage occurs and leads to apoptosis. In the third phase, there is a positive feedback loop in which PIC further reinforce stimulation to produce more PIC, causing further oxidative stress and worsening the injury. Eventually, a vicious cycle is created, and ulcers form due to intestinal injury. After stopping stimuli, the mucosa begins to heal by itself in a few days until the mucosa is restored. 2.6. Effects of 5-FU and Its Impact on Gut Microbiota Composition 5-FU causes a shift in gut microbiota profile by decreasing the abundance of intestine Firmicutes while increasing the abundance of Bacteroidetes and Verrucomicrobia[104]. Specific strains were shown to decrease in abundance, such as Clostridium spp., Lactobacillus spp. and Streptococcus spp., while some increased in abundance, like Escherichia spp.[105]. The gut microbiome is in a dynamic state, allowing external factors to influence it, such as antibiotics[106]. Studies have found antibiotics do not offer any clear protection against IM. This was demonstrated by mice that were treated with both 5-FU and a cocktail of antibiotics; pathogenic bacteria such as Escherichia, Shigella, and Enterobacter significantly increased in abundance while other commensals such as Firmicutes decreased in abundance. The combination of antibiotics and 5-FU downregulates genes involved in amino acid metabolism, replication and repair translation and nucleotide metabolism, which contributed to overall reduced antitumor efficacy. The effect of GM on 5-FU anti- PMMB 2023, 6, 1; a0000339 11 of 34 tumour efficacy is evident with the administration of antibiotics. Antibiotics use instead worsens patients' body weight in clinical trials. They also showed a higher tumour/body weight ratio in comparison to the 5-FU treatment alone[69]. Therefore, since antibiotics are not working in ameliorating IM, other strategies suitable for use along with 5-FU regimens to ameliorate IM need to be investigated. 3. Gut Microbiota Modulation-Based Strategies to Ameliorate 5-Fu-Induced Intestinal Mucositis The commensals residing in the human gut are crucial for sustaining a healthy human body. Healthy individuals usually contain bacterial species belonging to four phyla Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria[107,108]. GM helps hosts maintain immunity, protecting hosts from bacterial invasion and nutrient metabolism[47,109]. Shifts in this dynamic structure are present in inflammatory diseases such as inflammatory bowel disease (IBD), psoriasis and multiple sclerosis[107,110,111]. Neurological and psychiatric conditions, such as depression, obsessive compulsive disorder and myasthenia gravis, are also linked to gut microbiome changes[112,113]. Although the full mechanism of all of these interactions has yet to be unravelled, associations between GM and many common illnesses have been evidenced in numerous studies. For instance, gut dysbiosis is also evident in CRC patients. As mentioned earlier, 5-FU and antibiotics are also able to cause shifts in the gut microbial composition. Various strategies such as probiotics, prebiotics, dietary modifications, postbiotics, and fecal microbiota transplantation are utilized for gut microbiota modulation to promote health and manage diseases[114–118]. Among all of these strategies, probiotic use is one of the most common strategies, and it has been shown to prevent or ameliorate dysbiosis of the normal microbiota by maintaining a balanced gut microbiome by reducing potential pathogenic bacteria while increasing the number of beneficial bacteria[119–125]. With that, restoring the disturbed gut microbiota by supplementing probiotics can lessen the side effects of 5-FU, especially IM[126]. 3.1. Use of Probiotics in Intestinal Mucositis Probiotics are "live microorganisms when administered in adequate amounts that confer a health benefit on the host"[97]. They are non-invasive and safe when compared to pathogenic bacteria. Only viable bacteria can be considered probiotics, not including bacteria components that are not viable[18]. Some requirements that must be met to consider a bacterial strain as a probiotic include the ability to adhere to and colonise the gut mucosa to interact with the host to modulate the antagonism against pathogens and for immune system responses[44,127,128]. They colonise the gut temporarily and without releasing toxins or metabolites detrimental to the host. To ensure the viability of the bacteria for colonisation of the intestinal tract, they must be able to resist the environment of low pH and bile salt in the gastrointestinal tract[18]. They help to restore gut dysbiosis by cultivating good commensal bacteria. Probiotics currently are used as adjuvants for many gastrointestinal disorders such as diarrhoea due to infections (Rotavirus infection) or iatrogenic (antibiotic use) or due to other inflammatory diseases such as IBD[12,129]. It is thought that probiotics decrease PMMB 2023, 6, 1; a0000339 12 of 34 pathogenic bacteria that activate the immune system[107]. PIC and ROS have clearly been shown to be the main factors driving the inflammatory process of IM. As such, probiotics can come into play by being an anti-inflammatory agent. Since it has been shown that commensal organisms play a role in the inflammatory cascade, it would be a good target site to help ameliorate the manifestation of IM. Having said that, not only can probiotics suppress the inflammatory process in IM, but they also promote healing. Probiotics can improve host defence, such as mucus layer, TJ proteins and immune defence, subsequently ameliorating the side effects of 5-FU by reducing apoptosis and improving mucin production. Clinically, weight loss and diarrhoea were also attenuated with the administration of probiotics. Together, probiotics are capable of ameliorating 5-FU induced side effects via different mechanisms[105,130,131]. 3.1.1. Gut dysbiosis The composition of the gut microbiota is altered and modified through a process called dysbiosis due to undergoing chemotherapy, which is often related to the gastrointestinal tract's biochemistry and immunologic disorders[46]. Gut dysbiosis is shown to be common among CRC patients, indicating the possible relationship between the gut microbiome and the carcinogenesis of colorectal cancer[13,132]. A primary increase in Gram- negative bacteria was found following 5-Fu administration in rats, as shown in a preclinical study[13]. At the jejunal level, 5-FU decreases Clostridium sp., Lactobacillus sp. and Streptococcus sp., but increases Escherichia sp. In the colon, 5-FU caused decreases in Enterococcus sp., Lactobacillus sp. and Streptococcus sp. Faecal samples showed decreasing trends in Lactobacillus sp. and Bacteroides sp., and an increasing trend in E. coli and Staphylococcus sp.[133]. This was also validated in another study of mice administered with 5-Fu showing a shift away from good bacteria such as Actinobacteria and towards Bacteroidetes and Verrucomicrobia phyla with pro-inflammatory traits[13]. Hence, gut microbiota modulation-based strategies which can shift dysbiosis towards eubiosis or achieve gut microbiota homeostasis should be developed[46]. Probiotics have evidently been able to change this shift in organisms. Probiotic bacteria that can colonise the intestinal tract have been shown to restore eubiosis by releasing antimicrobial compounds such as bacteriocins and decreasing the pH to limit the development of other harmful bacteria[36]. Gut microbiota is brought back into balance when the amount of pathogenic bacteria reduces due to their inability to survive in the acidic environment while the amount of good bacteria which flourish in the acidic environment increases[18]. This was apparent when researchers used a probiotic mixture in mice and showed a decrease in opportunistic pathogens[69]. In male mice that received both 5-FU and Lactobacillus reuteri DSM 17938 (BG), it was found Bacteroides and Bacteroidaceae were most bountiful in 5-FU treated rats supplemented with the mixture, while those that received only saline solution showed that Muribaculaceae (S24_7) was most abundant[134]. Similarly, Lactobacillus plantarum 299v reduced 5-FU stimulated the growth of facultative anaerobes in the intestine[135]. Both probiotic strains, Lactobacillus casei PMMB 2023, 6, 1; a0000339 13 of 34 variety rhamnosus (Lcr35) and Bifidobacterium bifidum G9-1 (BBG9-1), also can significantly reverse the perturbed microbiota composition of Firmicutes and Bacteroidetes induced by FOLFOX. However, supplementation of Lcr35 itself did not affect faecal GM composition[12,131]. This shows that probiotics are strain- dependent. Hence, further research must be carried out in the future to find out more functional probiotic strains. Another strain, Rhodotorula mucilaginosa UFMGCB 18377, was also found to be able to reduce intestinal enterobacteria levels in 5-Fu treated mice with induced mucositis[136]. In vitro studies, non-recombinant L. lactis NZ9000 showed antagonistic activity against the invasive gut pathogen Listeria monocytogenes and its secreted compound called PAP had an inhibitory effect against opportunistic Enterococcus faecalis[137,138]. PAP is not the only property possessed by L. lactis NZ9000; it is thought that its antagonistic actions against pathogens could also be due to the secretion of lactic acid, bacteriocins or metabolites[137]. 3.1.2. Pro-inflammatory cytokines In the pathobiology of IM, TNF-α is a key PIC that causes IM. Other cytokines involved include interleukin (IL)-4, IL-6, IL-10 and IL-17. IL-17 is an inflammatory mediator. It acts together with IFN-y to synergistically enhance IL-8 secretion by human fetal intestinal epithelial cells[139]. When supplemented with probiotics Lcr35 and Bifidobacterium bifidum (LaBi, Infloran®), levels of PIC decreased[12,140,141]. The study shows Lcr35 has the ability to suppress NF-κB activity, also leading to suppression of inflammation and subsequently reduced IM in the intestine[12]. Other Lactobacillus strains, such as Lactobacillus delbrueckii CIDCA 133 (pExu:hsp65), a recombinant strain of L. delbrueckii is able to increase serum IL-10 in ileum tissue while reducing inflammatory infiltrate and cytokines such as TNF-α, IL-6, IL-12, and IL-1β[142]. Other strains S. thermophilus CRL 808 decreases IL-6 but increases IL-10. This vitamin-overproducing strain can reverse the rise of the anti-/proinflammatory cytokines ratio. It increases concentrations of IL-10 in the serum along with decreased IFN-γ levels[143]. In another experiment on a mouse model, it was found that the levels of TNF-α, IL-1β and IL-6 were higher in the 5-Fu induced mouse group than the control group. However, those concentrations markedly decrease in the use of S. thermophilus ST4 along with 5-Fu treatment[97]. B. infantis also successfully reduced high levels of PIC caused by 5-FU in CRC rats. As mentioned, IFN-γ recruits macrophages[77]. IL-2 is important for IFN-γ stimulation while IL-12 stimulates production of IFN-γ by naïve T cells[144] . In this case, it is found that Th1 cells and their cytokines, IL-2, IL-12 and IFN-γ, which are upregulated during chemotherapy, can be reversed by B. infantis[145]. PMMB 2023, 6, 1; a0000339 14 of 34 3.1.3. Intestinal damage As mentioned before, changes in the architecture of the intestinal mucosa are side effects of 5-Fu, including a decrease in villus high, an increase in crypt depth and intestinal permeability and a shortening of the intestinal length. Villus blunting and crypts fissure in the small intestine are mainly caused by an increase in apoptosis and a decrease in cell proliferation as a result of chemotherapy[97]. Restoration of the intestinal structure is very important as it influences the ability of nutrients to be absorbed by the body. A taller villus has more surface area for the absorption of nutrients, and it also lessens water loss and electrolyte loss[146]. Inefficient nutrient absorption can lead to loss of weight (LOW) in cancer patients[142]. Hence, probiotics have been proven to play a role in restoring intestinal damage. In an in vivo experiment, S. thermophilus ST4 was proven to be able to ameliorate the shortening of colon length of mice treated with 5-Fu. This is shown by a shorter average colon length in the mice treated only with 5-Fu when compared to the 5-Fu + S. thermophilus ST4 group. It was also found that the administration of S. thermophilus ST4 had also mitigated the decrease in the villus heights and crypt depths[97]. Another probiotic strain, R. mucilaginosa UFMGCB 18377, was also demonstrated to be able to decrease intestinal shortening of mice induced with mucositis by 5-Fu from around 17% shortening compared to the control group to 7.5% only. It is also found that this probiotic strain is also efficient in ameliorating the structure of the damaged intestinal mucosa, restoring its architecture and villus to crypt ratio, and reducing the increasing permeability of the intestine as a result of 5- Fu treatment[136]. Mucosal damage, such as crypt depths, has been shown to be significantly restored, even to levels similar to those only injected with saline, with Lcr35 and LaBi supplementation. Villus height to crypt depth ratio managed to return to normal[140], although another study by Huang et al.[141] has shown similar results without it being statistically significant. Other strains also showed similar results, with Saccharomyces boulardii and Lactobacillus fermentum BR11[130,147]. S. boulardii also induced the recovery of intestinal permeability by normalising the lactulose: mannitol ratio[130]. Effects of supernatant and live S. thermophilus TH-4 reduced ileal crypt fissure by at least half of the controls, although it unsuccessfully ameliorated 5-FU induced mucositis[148]. Lactobacillus plantarum ATCC 8014 showed a potential beneficial effect on ameliorating 5- Fu induced microscopic changes in the intestinal mucosa[149]. Lactobacillus delbrueckii (pExu:hsp65), L. delbrueckii subsp. Lactis CIDCA 133 strain, L. acidophilus or milk fermented with L. rhamnosus FLRH93 all were able to stop the shortening of the gut due to 5-FU. These probiotic strains also reduced 5-FU's adverse effects on IP, crypt depth, villus height, as well as villus/crypt ratio and goblet cells[142,146,150–152]. L. rhamnosus FLRH93 has also managed to prevent small intestine shortening, with an almost 5% difference compared to mice with 5-FU alone. This is consistent with the finding from the same study that the mice had less LOW[150]. PMMB 2023, 6, 1; a0000339 15 of 34 Two tested probiotic mixtures, such as Lactobacillus acidophilus and B. lactis, or a mix of four strains (Lactobacillus acidophilus, Lactobacillus paracasei, L. rhamnosus, and B. lactis) partially reversed histopathological changes induced by 5-FU[153]. BBG9-1 as mentioned before, prevented the increase in crypts quantity and reduction of small intestine length (although not statistically significant) but could not stop the induction of crypt cell apoptosis, as shown with B. infantis supplementation[145,154]. Supernatants from E. coli Nissle 1917 and F. prausnitzii enhanced barrier integrity by normalising crypt depth and prevented the change in barrier by 5-FU. S. thermophilus CRL 808 and S. thermophilus CRL 415 are strains that can produce folate, which was shown to induce less inflammation in the jejunum and preserved villus height/crypt depth ratio[143]. Improvement in the histopathological score was also apparent with P. freudenreichii WT[155]. Treatment with Saccharomyces cerevisiae UFMG A-905 successfully re-established a normal state in the intestine. Intestinal permeability returned to normal. S. cerevisiae UFMG A-905 has antioxidant and anti-inflammatory properties. Inflammatory markers were further reduced after giving selenium-enriched yeast[156]. 3.1.4. Immune cells infiltration The infiltration of inflammatory cells is broadly related to mucosal damage[136]. Neutrophil infiltration, associated with intestinal damage and its recruitment, can be measured quantitatively by myeloperoxidase activity (MPO). In a recent study, 5-FU can increase this by almost 500 times with an 83% decrease in sucrase[157]. On the other hand, eosinophil peroxidase (EPO) activity can be used to quantify the accumulation of another inflammatory cell called eosinophils. Eosinophils play an important role in inducing inflammation in the intestine by acting as antibacterials through the production of PIC and EPO activity. 5-Fu induced mucositis often results in the recruitment of neutrophil and eosinophil infiltrates or causing an increase in MPO and EPO levels, hence triggering inflammatory reactions in the tissues[136]. In a mucositis mouse model, MPO level has been shown to decrease with L. delbrueckii subsp. Lactis CIDCA 133 strain, S. boulardii, Lactococcus lactis NZ9000, Lactobacillus acidophilus A4 (A4), BBG9-1, R. mucilaginosa UFMGCB 18377 and probiotic mixtures[130,131,136,137,151,153,158,159]. R. mucilaginosa UFMGCB 18377 can also reduce EPO levels in the ileum and jejunum of mice with induced mucositis due to 5-Fu[136]. Lower MPO and EPO levels also indicate that there is lesser immune cell infiltration, which will reduce inflammatory responses. Besides, fewer polymorphonuclear cells in the lamina propria lead to less inflammatory activity by these cells, following fewer lesions occurring[137]. L. delbrueckii subsp. Lactis CIDCA 133 has also been shown to minimise 5-FU induced leukopenia[151]. Intestinal microbiota plays a part in the adaptive immune system by regulating IgA secretion. IgA is secreted by immune cells scattered across the mucosa[142]. IgA prevents bacteria colonisation and invasion into the mucosa. Toxic materials are blocked by IgA from penetrating the mucosa. IgA modulates the inflammatory response by inhibiting inflammatory response by activating regulatory T-cells[160]. Clearly, serum IgA has a dual PMMB 2023, 6, 1; a0000339 16 of 34 function in modulating microorganisms present in the gut. It protects the gut from pathogens but also regulates good bacteria. It seems that IgA has a different affinity for different microorganisms. There is a high affinity for pathogenic microorganisms but a low affinity for commensal bacteria[161]. IgA and commensals also regulate each other. Bifidobacteria is capable of modulating the expression of secretory IgA. Serum IgA was increased after 3 days of Bifidobacteria supplementation in rats that suffered burn injuries in the gut, but these levels were returned to normal by day 5. This self-modulating expression reduced incidences of bacteria translocated and reduced counts of E. coli and fungi[162]. Clearly, strains that increase IgA is a good probiotic to consider as well. L. delbrueckii CIDCA 133 (pExu:hsp65), a recombinant strain can also increase serum IgA[142]. 3.1.5. Apoptosis 5-FU is capable of inducing cell apoptosis by inducing the expression of IL-1β and TNF-α[15]. This is shown by its antagonist, interleukin-1 receptor (IL-1R) antagonist, in successfully reducing apoptosis after chemotherapy. Up-regulation of Bax and caspase 3 (pro-apoptotic proteins) along with downregulation of Bcl-2 and Bcl-xL (antiapoptotic proteins) occur during 5-FU induced intestinal apoptosis[163]. Caspases and Bcl-2 proteins are important in early apoptosis[164]. Bcl-2 can prevent cells from harsh environments such as radiation, heat and chemotherapy. Bcl-2 prevent these cells from undergoing apoptosis[163]. In vitro study shows that IL-1R antagonist inhibits apoptosis without affecting its anti-tumour efficacy. This shows that antagonising IL-1β activation can protect the gut from chemotherapy-induced IM[163]. Lcr35 has been shown to suppress apoptosis caused by FOLFOX administration. The reduction of the increased BAX/BCL-2 ratio induced by FOLFOX and a shift toward anti- apoptosis when Lcr35 is given[12]. Besides that, S. boulardii, a probiotic yeast, has been shown to reduce 5-FU induced intestinal cell apoptosis. 5-FU modifies the TLR response to activate PIC. When this cascade is activated, there is phosphorylation of ERK1/2, p38 and JNK. In particular, TNF-α and IL-1β activate these protein kinases in response to oxidative stress. The IL-1β induced MyD88 pathway and phosphorylation of protein kinases are inhibited by S. boulardii[165]. In another case, the probiotic Propionibacterium freudenreichii requires SlpB protein to alleviate mucositis induced by chemotherapy[130]. 3.1.6. Mucin production The human gut mucosa is protected by a mucus layer. This mucus layer comprises water, glycoproteins, trefoil factors, defensins and mucins. Mucin is an important component, it being the first-line defence in the gut. Mucins are produced by goblet cells that reside all over the small and large intestines and produce mucin[166]. Bacteria are able to adhere to mucin oligosaccharides. Commensals can compete with pathogens to bind and colonise the mucin, preventing pathogens from attaching to the mucosal surface. For example, the parasite Entamoeba histolytica requires contact with epithelial cells in order to invade it. Having a thick mucus layer prevents it from having contact with the epithelium. Once this mucus layer PMMB 2023, 6, 1; a0000339 17 of 34 is disrupted, E. histolytica can easily penetrate through the gut barrier leading to phagocytosis of epithelial cells[167]. Our gut epithelium is endlessly exposed to various noxious chemicals and physical insults such as digestive enzymes, faecal material, resident bacteria, and intestinal pathogens and their products. Mucin blocks the mucosa from bacterial enzymes by acting as a substrate for enzymes such as α-galactosidase, β-N-acetylgalactosaminidase, sialidase, β-glucuronidase, blood group degrading enzymes, and proteases[166]. Having a mucosal shield as a barrier between underlying epithelium and these substances is essential in preventing IM. The mucus layer is also thickened in response to the presence of pathogens. This quick response is key to eliminating pathogens. In other words, pathogens and their toxins have a positive stimulation on mucin production. Thick mucus allows easier excretion of pathogens but also carries a risk of diminished mucin[167,168]. Vibrio cholerae enterotoxin stimulated mucin production[169]. However, Helicobacter pylori downregulate mucin-producing genes MUC1 and MUC5AC resulting in reduced mucin synthesis[170]. Another important gene is MUC2. MUC2 is a key player in the formation of the mucus layer. It is stored in goblet cells and is key in determining goblet cell shape[171]. In MUC2 knockout mice, defective or depleted mucus defence layer showed bacteria colonisation and inflammation (with multiple crypts), subsequently leading to carcinogenesis[168,171]. For example, Lactobacillus plantarum 299v, L. rhamnosus strain GG and L acidophilus strain DDS-1 are shown to increase gene expression of MUC2 and MUC3 at the mRNA level[172,173]. This managed to prevent adherence of pathogenic E. coli onto gut epithelium[172]. Besides, non-pathogenic commensal E. coli Nissle 1917 regulates multiple genes. Besides MUC2 and MUC3, it also upregulates MUC5AC and MUC5A[174]. Butyrate-producing organisms also have shown abilities to increase MUC2 gene expression in in vitro studies. Human colon biopsies in ex vivo studies showed increased mucous secretion in the presence of butyrate-producing organisms. Trefoil factors, a contributor to the viscosity and elasticity of mucus, can reduce the chemotaxis of inflammatory cells and are thought to repair damaged mucosa. Like mucin, it is only produced by GC. Trefoil factors are reduced in rats with colitis, but with butyrate administration, these levels increased[175]. 5-FU treatment has been shown to influence the mucus layer and decrease goblet cells while increasing the number of crypts in the jejunum[176]. This is possibly due to the pro- inflammatory state 5-FU induces. It is found that Rhodotorula mucilaginosa UFMGCB 18377 decreases the loss of goblet cells in mice induced with mucositis by 5-Fu[136]. Other than that, treatment with L. delbrueckii rCIDCA 133:Hsp65 also increases goblet cells and expression of the MUC2 gene. A higher quantity of goblet cells and increased MUC2 gene expression can reinforce each other's positive effect in protecting the mucus layer[142]. 3.1.7. Weight loss Regarding weight loss, probiotic strains such as Lcr35 and LaBi have been shown to reduce body weight loss by 20%. Mice in the Lcr35 probiotic group showed a lesser degree of LOW compared to 5-FU and saline group[177]. Other Lactobacillus strains, such as L. PMMB 2023, 6, 1; a0000339 18 of 34 plantarum 299v, have been shown to improve food intake, resulting in less LOW, while others, such as L. delbrueckii subsp. Lactis CIDCA 133 strain and L. acidophilus could increase food and milk intake but still reduce LOW[135,142,151]. Riboflavin overproducing L. plantarum CRL2130 also showed similar results[152,178]. Besides these, L. fermentum BR11 and L. rhamnosus GG increase colon weight in conjunction with 5-FU treatment[157]. Lactobacillus plantarum 299v and BBG9‐1, at a concentration of 1 × 109 CFU/mL, improved both food consumption and body weight post 5-FU administration[131,147]. Interestingly, high doses resulted in LOW, while low doses resulted in body weight gain after one week of 5-FU treatment. On day 8 after the treatment, low doses increased body weight compared to high doses of the probiotic, which decreased food intake[147]. B. infantis also has shown weight-gaining properties[145,154]. In another in vivo experiment in mice, it was found that mice with 5-Fu induced IM showed a significant decrease of about 50% in food intake and about a 20% decrease in their body weight in relation to the initial weight on the 18th day. However, in the 5-Fu + S. thermophilus ST4 mice group, there was no big difference in body weight loss (only 1.85%) and food intake compared to the control group[97]. Another strain of probiotic, Rhodotorula mucilaginosa UFMGCB 18377 ameliorated the decrease in food consumption of mice induced with mucositis by 5-Fu hence reducing the weight loss in comparison to their initial weight from 20.3% to 12.4% in the probiotic-treated group as compared to the mucositis group[136]. A probiotic mixture (L. acidophilus, L. paracasei, L. rhamnosus, and B. lactis) reduced body weight loss[153]. However, the addition of Streptococcus genus, resulting in a mixture of B. breve DM8310 + L. acidophilus DM8302, L. casei DM8121 + S. thermophilus DM8309 showed no differences in faecal output, and food intake[158]. E. coli Nissle 1917 supernatant and F. prausnitzii supernatant prevented further weight loss from happening post 5-FU induction resulting in normal urine and faecal output[179]. These mixed results show that, clinically, some probiotic strains are better than others at preventing weight loss. None of the probiotics mentioned exacerbated weight loss, suggesting its importance in maintaining patients' weight. Further work should be done to understand the underlying mechanisms exhibited by some strains to preserve the body weight, potentially via improved feeding behaviour and the host's metabolism[180]. 3.1.8. Diarrhoea Severe diarrhoea is presumed to be closely related to the shortened colon, one of the side effects of the administration of 5-Fu[97]. Diarrhoea occurs when the colon's absorption capacity is surpassed by the rising fluid volume out of the small intestine[44]. Rat studies show that probiotics can ameliorate diarrhoea. The research was carried out on a mice model by assessing the severity of diarrhoea using Bowen's score system, in which diarrhoea is categorised into 4 grades based on the consistency of stool (0 — normal stool; 1 — mild diarrhoea, the stool is slightly soft and wet; 2 — moderate diarrhoea, stool are wet and unformed; 3 — severe diarrhoea, stool are watery). It is found that the 5-Fu + S. thermophilus PMMB 2023, 6, 1; a0000339 19 of 34 ST4 group shows a lower mean diarrhoea score that changed from 1.0 to 0.1 when compared to the group of mice induced with 5-Fu only that scored 2.5 and 2.5 respectively, on the most severe day 14th to 15th[97]. For example, Lcr35 group and LaBi group, L. delbrueckii CIDCA 133 (pExu:hsp65), L. plantarum CRL2130, BBG9-1 and also folate-producing S. thermophilus CRL 808 were shown to successfully reduce the incidence of diarrhoea[140,143,178]. Probiotics reduced the incidence of diarrhoea in human patients. In a systematic review, researchers found probiotics and fibre reduced the incidence of grade 3 or 4 abdominal discomfort (flatulence, borborygmi and abdominal distension) and the need for chemotherapy dose reduction, compared with placebo[14,181]. L. rhamnosus GG decreased the incidence of diarrhoea in grades 3 or 4 by around 15%. In patients with severe diarrhoea, metabolic and nutritional imbalances may occur, which may further worsen their condition. It is evident that probiotics show improvement in diarrhoea in animal models and humans[45]. In another clinical trial on patients treated with 5-Fu based chemotherapy and administered with probiotic Lactobacillus kefiri LKF01, it was found that the incidence of diarrhoea was reduced. Only 4.7% and 8.7% of patients treated with 5-Fu and FOLFOXIRI respectively developed G3-4 severe diarrhoea [CTCAE grade [G] 3-4]. In contrast, no incidence of high- grade diarrhoea was reported by patients treated with FOLFOX and FOLFIRI, respectively. Since their results also showed that the onset of diarrhoea mostly started at the early stages of chemotherapy, hence it is hypothesised that the intake of probiotics earlier before the treatment begins and not concomitantly as they did in their studies could possibly work as a preventive measure to reduce the rates of early-onset diarrhoea, but further studies are required to validate it[44]. Table 1. Probiotic genus, strains and mixtures that show beneficial effects toward IM. Different probiotics ameliorate IM in various aspects are summarised in this table. Probiotic Effects of probiotic on ameliorating intestinal mucositis Reference Lactobacillus acidophilus • Preserved villus and crypt length ratio • Reduced GSH levels • Reduced myeloperoxidase activity • Reduced nitrite levels • Reduced level of PIC, TNF-α, IL-1β • Reduced levels of chemokines CXCL-1 • Increased levels of anti-inflammatory cytokines IL-10 [152] Lactobacillus acidophilus A4 • Prevented attachment of Escherichia coli O157:H7 • Reduced level of PIC, IL-8, TNF-α, IL-1β • Induced the expression of MUC2 [182] Lactobacillus brevis 47 • Partially restored expression of Ki-67 epithelial proliferation cell marker [183] Lactobacillus plantarum CRL2130 • Reduced weight loss • Reduced diarrhoea scores • Maintained mucosal architecture • Reduced mucosal inflammation [178] PMMB 2023, 6, 1; a0000339 20 of 34 Probiotic Effects of probiotic on ameliorating intestinal mucositis Reference • Preserved villus and crypt length • Reduced level of PIC, IL-10 Lactobacillus plantarum supernatant • Increased sensitivity of colorectal cancer cells to 5- fluorouracil • Inhibited CD44 gene expression • Inhibited CD133 gene expression • Inhibited CD166 gene expression • Inhibited ALDH1 gene expression • Increased caspase 3 activity • Inhibited signalling pathway Wnt/β-catenin [43] Lactobacillus casei variety rhamnosus (L cr35) • Reduced diarrhoea scores • Reduced loss of weight • Normalised crypt depth • Preserved villus and crypt length ratio • Reduced villous inflammation • Diminished apoptosis • Reduced level of PIC, TNF-α and IL-6 • Reduced levels of NF-κB-, and BAX-activated cells • Restored composition of fecal gut microbiota • Decreased Firmicutes and Bacteroidetes ratio • Reduced CD3+/CD8+ count • Increased CD3+CD4+/CD3+CD8+ • Reduced CD44 the number of Ki67 proliferative cells [177] Lactobacillus delbrueckii CIDCA 133 (pExu:hsp65) • Reduced inflammatory infiltrate • Reduced intestinal permeability • Increased serum IgA in the intestinal fluid • Improved histological score • Prevented shortening of the intestine • Preserved villus and crypt length ratio • Reduced myeloperoxidase activity • Reduced level of PIC, TNF-α, IL-6, IL-12, IL-1β • Reduced level of Toll-like receptors • Increased level of anti-inflammatory cytokines IL-10 • Increased gene expression of MUC2 • Increased gene expression of claudin 1 • Increased the number of goblet cells [142] Lactococcus lactis NZ9000 • Prevented growth of Listeria monocytogenes • Pancreas-associated peptide secretion prevented growth of E. faecalis • Improved histological score • Reduced infiltration of neutrophils • Reduced infiltration of eosinophils • Reduced secretion of Immunoglobulin-A in the gut • Reduced gut inflammation [137] Lactobacillus sp. • Induced expression of MUC3 [173] Lactobacillus rhamnosus FLRH93 • Preserved villus and crypt length ratio • Lessen the decrease of goblet cells [150] PMMB 2023, 6, 1; a0000339 21 of 34 Probiotic Effects of probiotic on ameliorating intestinal mucositis Reference • Increased the expression Bcl-2 in the intestinal tract • Decreased expression of NLRP3 • Reduced level of PIC, TNF-α, IL-1β • Increased survival rate of mice Lactobacillus rhamnosus GG • Reduced diarrhea scores • Reduced incidence of abdominal discomfort [45] Streptococcus thermophilus CRL 808 • Enhanced chemotherapeutic effect of 5-FU • Reduced diarrhoea scores • Reduced jejunal inflammation • Increased histological score • Reduced level of PIC, IL-6 • Increased level of anti-inflammatory cytokines IL-10 [143] Streptococcus thermophilus ST4 • Increased food intake of mice • Decreased body weight loss • Reduced mean diarrhoea score • Reduced shortening of colon length • Restored villus heights and crypts depths • Reduced level of PIC, TNF-α, IL-1β and IL-6 [97] Bifidobacterium infantis • Mice showed higher body weight • Mice had taller villus • Reduced level of PIC, IL-6, IL-1β and TNF-α • Down-regulated levels of T-bet (Th transcription factor) • Increased CD4+ levels • Increased CD25+ levels • Increased Foxp3+ levels • Increased Tregs levels • Reduced CD4+ IL17A+ cells [145] Saccharomyces boulardii • Changed expression of Toll-like receptors, TLR2, TLR4 • Changed expression of MyD88 • Changed expression of NF-κB • Changed expression of ERK1 • Changed expression of phospho-p38 • Changed expression of phospho-JNK • Reduced level of PIC, TNF-α, IL-1β • Reduced level of chemokine CXCL-1 [165] Saccharomyces cerevisiae UFMG A-905 • Partially reduced intestinal permeability • Reduced myeloperoxidase activity • Reduced level of chemokine CXCL-1 • Reduced mucosal inflammation • Reduced oxidative stress [156] Propionibacterium freudenreichii • Reduced level of PIC, IL-12, IL-17a, IL-8, TNF- α • Changed expression of cld1 [155] PMMB 2023, 6, 1; a0000339 22 of 34 Probiotic Effects of probiotic on ameliorating intestinal mucositis Reference Wild strain of Propionibacterium freudenreichii • reduced weight loss • reduced gut inflammation • increased histopathological scores • induced production of Th17 cells • reduced level of IgA [155] Rhodotorula mucilaginosa UFMGCB 18377 • Ameliorated the decrease in food consumption • Reduced weight loss • Reduced shortening of intestinal length • Restored villus to crypt ratio • Reduced intestinal permeability • Reduced goblet cells loss • Reduced enterobacteria in intestine • Reduced MPO and EPO levels / Reduced neutrophils and eosinophils infiltration [136] Probiotic mixture : • Bifidobacterium breve DM8310 • Lactobacillus acidophilus DM8 302 • Lactobacillus casei DM8121 • Streptococcus thermophilus DM 8309 • Reduced level of PIC • Reduced infiltration of neutrophils • Reduced intestinal permeability • Changes of Toll-like receptors, TLR2, TLR4 signalling pathway [158] 4. Discussion and Future Direction Despite extensive evidence shown in this review, there is no solid guideline for treating intestinal mucositis due to chemotherapy. Current medications or other ways to combat chemotherapy-induced side effects are generally not completely effective, mostly fail to address possible long-term impacts, and may even cause additional side effects that merely exacerbate patients' sufferings[13]. Some of the interventions being used currently are limited to ice, analgesics, barrier protectors and topical antimicrobials. Hence, it is necessary to look for other possible alternatives to treat IM[136]. Probiotics have clearly shown to be a valuable adjuvant therapy in treating intestinal mucositis. Many strains successfully reduce levels of PIC, inhibit inflammatory pathways, and strengthen the gut's physical defence system. Probiotics also manage to prevent adherence of pathogenic bacteria and prevent gut cell apoptosis. When patients undergo cancer treatment, they are at risk of gut dysbiosis. They will be less protected from harmful external factors due to lack of commensal GM and also due to a weak immune system from cancer itself as well as the chemotherapy drug. PMMB 2023, 6, 1; a0000339 23 of 34 Supplementing with probiotics before, during and after chemotherapy can increase one more level of protection for cancer patients. Further research is required to investigate the integration of probiotic interventions into the clinical management of IM, effectively halting mucositis in its track. With the current understanding that the gut microbiome plays a crucial role in all stages of intestinal mucositis, including initiation, upregulation, message generation, amplification, signaling, ulceration, and healing, it is anticipated that this integration will be realised in the near future. There is also a risk of bacterial translocation, causing invasive infections. Immunocompromised cancer patients would be particularly vulnerable, especially the risk of bacteraemia or sepsis, worsening patients' survival and mortality rate. However, with all of these studies mentioned, there were no events of bacteraemia or sepsis. Probiotics are beneficial. They inhibit the invasion of pathogens and strengthen the immune system. It is also important to note that probiotics do not compromise 5-FU efficacy, supporting their further use[17]. As shown in this review, there is a large shortage of studies conducted on humans. Although some of the preclinical studies show promising results, they might not translate similarly to humans. Most of the studies are also done in mice induced for mucositis or induced with carcinoma, which could be different from humans with chemotherapy-induced mucositis with underlying CRC. Perhaps this is why there is currently no definitive proof supporting the use of probiotics in treating IM. Therefore, this review calls for future study of ethical, large, randomised, double-blind human trials. This is to further assess the capability of probiotics in real-life clinical settings. Studies should include isolated strains that can ameliorate all side effects of IM rather than just ameliorating one specific side effect. Other than that, the most suitable doses along with the most effective probiotic strains or combination of different probiotic strains, should also be further evaluated to maximise the beneficial effects of probiotics on the host. Also, human factors such as genetics[184,185], race, and environmental factors could affect patients' response to probiotics supplementation. Thus, knowing the correlation between these external factors and response to probiotics should be studied as well. By doing this, we might be able to predict which patients are specifically at risk of developing certain side effects. Perhaps we can tailor the probiotic supplementation to patients' needs, thus developing a personalised probiotic regimen for each patient to reach optimal treatment outcomes. Individual variability is impossible to predict; however, more studies on the association of certain external factors or internal factors, such as gene polymorphism, can be a starting point for having proper guidelines for IM therapy. PMMB 2023, 6, 1; a0000339 24 of 34 A challenge in using probiotics as a therapeutic method would be modulating its effects to target inflamed tissues while leaving normal tissues undisturbed and, most importantly, without disturbing the efficacy of 5-FU. Evidence showed probiotics do not affect the efficacy of 5-FU and its actions on tumour cells. However, similar to other studies, probiotics' influence on 5-FU's mode of action is only primarily known in rat studies. There has yet to be a large human trial to allow full justification. The challenge of maximising probiotic protection of mucosa while minimising the protection of tumour will need to be overcome. Perhaps, new techniques or technology should be used to overcome this challenge. Also, probiotics are required to remain in the body for a considerable amount of time. So far, it is uncertain how long the effects of probiotics will last and whether the effects will continue to last even after probiotics are no longer in the body[186]. This review also does not discuss the viability of probiotics after going through the digestive tract, such as stomach acid and digestive enzymes[146]. 5. Conclusion In conclusion, this review shows promising evidence that would encourage future studies on the use of probiotics in ameliorating the side effects of 5-FU or chemotherapy in general. More studies are important, as we have seen that IM is a complex process, and its association with gut microbes is even more complex. 5-FU continues to be used today, and yet there is only a superficial understanding of the relationship between GM and 5-FU. This review highlights that GM plays an important role in the host's response to 5-FU. Also, this review gives a framework on how probiotics may increase the efficacy and lessen the side effects of 5-FU; thus, future studies on this will be beneficial to many patients that use 5-FU. There has yet to be fully validated that probiotics could be the magic bullet, and for now, it serves better as an adjuvant targeted therapy. Nevertheless, this emerging field of research provides hope for finding safer and more effective treatment options for CRC and mitigating the adverse effects associated with traditional chemotherapy. These approaches not only combat cancer but also preserve patients' quality of life during treatment. Continued efforts in this direction may open new avenues for personalized and targeted approaches to CRC treatment, revolutionizing the way we combat this devastating disease. Author Contributions: AAX-HS and J-YC performed the literature search, critical data analysis as well as manuscript writing. J-YC, JW-FL, VL, YK, SO, SHW, and K-GC performed editing, revision and proofreading. JW-FL, VL, YK, SO, SHW, K-GC and LT-HT provided technical support and expertise. LT-TH conceptualised this review writing project. Acknowledgments: This work was inspired by the Jeffrey Cheah School of Medicine and Health Sciences (JCSMHS) "MED5101 Scholarly Intensive Placement (SIP)". The author also would like to acknowledge and sincerely thank the Young Scholar’s Program (YSP) conducted at JCSMHS, Monash University Malaysia. PMMB 2023, 6, 1; a0000339 25 of 34 Conflicts of Interest: The authors declare no conflict of interest. References 1. World Health Organization. Cancer. 2023 [Accessed 3 July 2023]; Available from: https://www.who.int/news- room/fact-sheets/detail/cancer. 2. Ishak M, Baharudin R, Tan LT-H, et al. Landscape of HOXA genes methylation in colorectal cancer. Prog Microbes Mol Biol 2020; 3(1). 3. Lye K-L, Tan LT-H, and Yap H-M. Insight of microRNA role in Colorectal Cancer. Prog Microbes Mol Biol 2020; 3(1). 4. Eng S-K, Tan LT-H, Goh B-H, et al KRAS as potential target in colorectal cancer therapy. Natural Bio-active Compounds: Volume 1: Production and Applications. 2019. 389–424. 5. Siegel RL, Miller KD, Goding Sauer A, et al. Colorectal cancer statistics, 2020. CA Cancer J Clin 2020; 70(3): 145–164. 6. Biller LH and Schrag D. Diagnosis and Treatment of Metastatic Colorectal Cancer: A Review. JAMA 2021; 325(7): 669–685. 7. Xie Y-H, Chen Y-X, and Fang J-Y. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther 2020; 5(1): 22. 8. Ragnhammar P, Hafström L, Nygren P, et al. A systematic overview of chemotherapy effects in colorectal cancer. Acta Oncol 2001; 40(2–3): 282–308. 9. Jee SH, Moon SM, Shin US, et al. Effectiveness of Adjuvant Chemotherapy with 5-FU/Leucovorin and Prognosis in Stage II Colon Cancer. J Korean Soc Coloproctol 2011; 27(6): 322–328. 10. International Multicentre Pooled Analysis of Colon Cancer Trials (IMPACT) investigators. Efficacy of adjuvant fluorouracil and folinic acid in colon cancer. Lancet 1995; 345(8955): 939–944. 11. Jensen GS, Benson KF, Carter SG, et al. GanedenBC30 cell wall and metabolites: anti-inflammatory and immune modulating effects in vitro. BMC Immunol 2010; 11: 15. 12. Chang C-W, Liu C-Y, Lee H-C, 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). 13. Kalasabail S, Engelman J, Zhang LY, et al. A perspective on the role of microbiome for colorectal cancer treatment. Cancers (Basel) 2021; 13(18): 4623. 14. Touchefeu Y, Montassier E, Nieman K, et al. 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(5): 409–421. 15. Hamouda N, Sano T, Oikawa Y, et al. Apoptosis, Dysbiosis and Expression of Inflammatory Cytokines are Sequential Events in the Development of 5-Fluorouracil-Induced Intestinal Mucositis in Mice. Basic Clin Pharmacol Toxicol 2017; 121(3): 159–168. 16. Chang C-T, Ho T-Y, Lin H, et al. 5-Fluorouracil induced intestinal mucositis via nuclear factor-κB activation by transcriptomic analysis and in vivo bioluminescence imaging. PLoS One 2012; 7(3): e31808. 17. van Vliet MJ, Harmsen HJ, de Bont ES, et al. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog 2010; 6(5): e1000879. 18. Miknevicius P, Zulpaite R, Leber B, et al. The impact of probiotics on intestinal mucositis during chemotherapy for colorectal cancer. A comprehensive review of animal studies. Int J Mol Sci 2021; 22(17): 9347. https://www.who.int/news-room/fact-sheets/detail/cancer https://www.who.int/news-room/fact-sheets/detail/cancer PMMB 2023, 6, 1; a0000339 26 of 34 19. Tan LTH, Chan KG, Pusparajah P, et al. Targeting Membrane Lipid a Potential Cancer Cure? Front Pharmacol 2017; 8: 12. 20. Tan LTH, Chan CK, Chan KG, et al. Streptomyces sp. MUM256: A Source for Apoptosis Inducing and Cell Cycle-Arresting Bioactive Compounds against Colon Cancer Cells. Cancers (Basel) 2019; 11(11): 1742. 21. Tan LT-H, Chan K-G, Pusparajah P, et al. Mangrove derived Streptomyces sp. MUM265 as a potential source of antioxidant and anticolon-cancer agents. BMC Microbiol 2019; 19: 1–16. 22. Ab Mutalib N-S, Wong SH, Ser H-L, et al. Bioprospecting of microbes for valuable compounds to mankind. Prog Microbes Mol Biol 2020; 3(1). 23. Elsalam RM, Goh KW, Mahadi M, et al. The Antibacterial Activities of Secondary Metabolites Derived from Streptomyces sp. Prog Microbes Mol Biol 2022; 5(1). 24. Law JW-F, Pusparajah P, Ab Mutalib N-S, et al. A review on mangrove actinobacterial diversity: the roles of Streptomyces and novel species discovery. Prog Microbes Mol Biol 2019; 2(1). 25. Ong YS and Tan LT-H. Cancer, natural products and nanodrug delivery systems. Prog Microbes Mol Biol 2020; 3(1). 26. Tan LTH, Ser HL, Yin WF, et al. Investigation of Antioxidative and Anticancer Potentials of Streptomyces sp. MUM256 Isolated from Malaysia Mangrove Soil. Front Microbiol 2015; 6: 1316. 27. Law JW, Law LN, Letchumanan V, et al. Anticancer Drug Discovery from Microbial Sources: The Unique Mangrove Streptomycetes. Molecules 2020; 25(22): 5365. 28. Hui ML, Tan LT, Letchumanan V, et al. The Extremophilic Actinobacteria: From Microbes to Medicine. Antibiotics (Basel) 2021; 10(6): 682. 29. Thye AY-K, Letchumanan V, Tan LT-H, et al. Malaysia’s Breakthrough in Modern Actinobacteria (MOD- ACTINO) Drug Discovery Research. Prog Microbes Mol Biol 2022; 5(1). 30. Law JWF, Tan LT-H, Letchumanan V, et al. Streptomyces griseiviridis sp. nov., A Novel “Modern Actinobacteria” isolated from Malaysia Mangrove Soil. Prog Microbes Mol Biol 2023; 6(1). 31. Pusparajah P, Law JW-F, Chan K-G, et al. Whole-Genome Sequence of Streptomyces pluripotens strain MUM 16J, a Potential Resource of Glycopeptide Antibiotic and Biocontrol Agent against Biofilm-forming Bacteria. Prog Microbes Mol Biol 2023; 6(1). 32. Goh YX, Chan KG, and Hong KW. Whole-Genome Sequence of Chelatococcus daeguensis Strain M38T9, Isolated from Ulu Slim Hot Spring in Malaysia. Prog Microbes Mol Biol 2022; 5(1). 33. Ser H-L, Tan LT-H, Tan W-S, et al. Whole-genome sequence of bioactive streptomycete derived from mangrove forest in Malaysia, Streptomyces sp. MUSC 14. Prog Microbes Mol Biol 2021; 4(1). 34. Kemung HM, Tan LT-H, Chan K-G, et al. Streptomyces sp. strain MUSC 5 from mangrove forest in Malaysia: Identification, antioxidant potential and chemical profiling of its methanolic extract. Prog Microbes Mol Biol 2020; 3(1). 35. Law JW-F, Letchumanan V, Tan LT-H, et al. The rising of “modern actinobacteria” era. Prog Microbes Mol Biol 2020; 3(1). 36. Chee PY, Mang M, Lau ES, et al. Epinecidin-1, an Antimicrobial Peptide Derived From Grouper (Epinephelus coioides): Pharmacological Activities and Applications. Front Microbiol 2019; 10: 2631. 37. Goh JXH, Tan LT, Goh JK, et al. Nobiletin and Derivatives: Functional Compounds from Citrus Fruit Peel for Colon Cancer Chemoprevention. Cancers (Basel) 2019; 11(6): 867. 38. Chan WK, Tan LT, Chan KG, et al. Nerolidol: A Sesquiterpene Alcohol with Multi-Faceted Pharmacological and Biological Activities. Molecules 2016; 21(5): 529. PMMB 2023, 6, 1; a0000339 27 of 34 39. Tang C, Hoo PC, Tan LTH, et al. Golden Needle Mushroom: A Culinary Medicine with Evidenced-Based Biological Activities and Health Promoting Properties. Front Pharmacol 2016; 7: 474. 40. Tan LKS, How CW, Foo JB, et al. Resveratrol as a potential broad-spectrum compound for cancer treatment. Prog Microbes Mol Biol 2020; 3(1). 41. Blondy S, David V, Verdier M, et al. 5-Fluorouracil resistance mechanisms in colorectal cancer: From classical pathways to promising processes. Cancer Sci 2020; 111(9): 3142–3154. 42. Tan LT-H, Chan K-G, Lee L-H, et al. Streptomyces bacteria as potential probiotics in aquaculture. Front Microbiol 2016; 7: 79. 43. An J and Ha EM. Combination Therapy of Lactobacillus plantarum Supernatant and 5-Fluouracil Increases Chemosensitivity in Colorectal Cancer Cells. J Microbiol Biotechnol 2016; 26(8): 1490–1503. 44. Ghidini M, Nicoletti M, Ratti M, et al. Lactobacillus kefiri LKF01 (Kefibios®) for prevention of diarrhoea in cancer patients treated with chemotherapy: A prospective study. Nutrients 2021; 13(2): 1–11. 45. Österlund P, Ruotsalainen T, Korpela R, et al. Lactobacillus supplementation for diarrhoea related to chemotherapy of colorectal cancer: a randomised study. Br J Cancer 2007; 97(8): 1028–1034. 46. Rodriguez-Arrastia M, Martinez-Ortigosa A, Rueda-Ruzafa L, et al. Probiotic supplements on oncology patients’ treatment-related side effects: A systematic review of randomized controlled trials. Int J Environ Res Public Health 2021; 18(8): 4265. 47. Prisciandaro LD, Geier MS, Butler RN, et al. Evidence Supporting the use of Probiotics for the Prevention and Treatment of Chemotherapy-Induced Intestinal Mucositis. Crit Rev Food Sci Nutr 2011; 51(3): 239–247. 48. Geller LT, Barzily-Rokni M, Danino T, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017; 357(6356): 1156–1160. 49. Li H, He J, and Jia W. The influence of gut microbiota on drug metabolism and toxicity. Expert Opin Drug Metab Toxicol 2016; 12(1): 31–40. 50. Joseph RJ, Ser H-L, Kuai Y-H, et al. Finding a balance in the vaginal microbiome: how do we treat and prevent the occurrence of bacterial vaginosis? Antibiotics 2021; 10(6): 719. 51. Lau AWY, Tan LT-H, Ab Mutalib N-S, et al. The chemistry of gut microbiome in health and diseases. Prog Microbes Mol Biol 2021; 4(1). 52. Lee JK, Tan LTH, Ramadas A, et al. Exploring the Role of Gut Bacteria in Health and Disease in Preterm Neonates. Int J Environ Res Public Health 2020; 17(19): 6963. 53. Ser H-L, Wong JYJ, Letchumanan V, et al. Moving beyond the gastrointestinal tract: the involvement of gut microbiome in endometriosis. in Gut. 2021. BMJ Publishing Group. 54. Tan LT-H, Letchumanan V, Law JW-F, et al. IDDF2022-ABS-0221 The roles of GUT microbiota in hand, foot and mouth disease. in Gut. 2022. 55. Loo K-Y, Law JW-F, Tan LT-H, et al. IDDF2022-ABS-0201 Vibrio: detecting you earlier, avoiding gastroenteritis and GUT microbiome dysbiosis. in Gut. 2022. 56. Learn-Han L, Ser H-L, Letchumanan V, et al. IDDF2020-ABS-0116 The role of gut microbiome in traditional Chinese medicine syndromes: focusing on the spleen deficiency syndrome. in Gut. 2020. 57. Lee L-H, Tan LT-H, Letchumanan V, et al. IDDF2020-ABS-0115 A moulding game: the role of gut microbiome in osteoporosis. in Gut. 2020. BMJ Publishing Group. 58. Learn-Han L, Law JW-F, Tan LT-H, et al. IDDF2020-ABS-0113 Budding association between gut microbiome in the development of Myasthenia Gravis. in Gut. 2020. PMMB 2023, 6, 1; a0000339 28 of 34 59. Lee L-H, Letchumanan V, Tan LT-H, et al. IDDF2020-ABS-0112 Gut-skin axis: decoding the link between the gut microbiome and hives. in Gut. 2020. BMJ Publishing Group. 60. Letchumanan V, Thye AY-K, Tan LT-H, et al. Gut feelings in depression: microbiota dysbiosis in response to antidepressants. in Gut. 2021. BMJ Publishing Group. 61. Lee L-H, Loo K-Y, Tan LT-H, et al. Exploring the gut microbiota variation in response to vibrio infection. in Gut. 2021. BMJ Publishing Group. 62. Law JW-F, Letchumanan V, Ser H-L, et al. Enterobacteriaceae–deciphering the culprit gut bacteria causing necrotizing enterocolitis in infants. in Gut. 2021. BMJ Publishing Group. 63. Chew SS, Tan LT, Law JW, et al. Targeting Gut Microbial Biofilms-A Key to Hinder Colon Carcinogenesis? Cancers (Basel) 2020; 12(8): 2272. 64. Ong IJ, Loo K-Y, Law LN-S, et al. Exploring the impact of Helicobacter pylori and potential gut microbiome modulation. Prog Microbes Mol Biol 2023; 6(1). 65. Attene-Ramos MS, Wagner ED, Plewa MJ, et al. Evidence that hydrogen sulfide is a genotoxic agent. Mol Cancer Res 2006; 4(1): 9–14. 66. Abdulamir AS, Hafidh RR, and Abu Bakar F. The association of Streptococcus bovis/gallolyticus with colorectal tumors: the nature and the underlying mechanisms of its etiological role. J Exp Clin Cancer Res 2011; 30(1): 11. 67. Gold JS, Bayar S, and Salem RR. Association of Streptococcus bovis bacteremia with colonic neoplasia and extracolonic malignancy. Arch Surg 2004; 139(7): 760–765. 68. Sonis ST. Pathobiology of mucositis. Semin Oncol Nurs 2004; 20(1): 11–15. 69. Yuan L, Zhang S, Li H, et al. The influence of gut microbiota dysbiosis to the efficacy of 5-Fluorouracil treatment on colorectal cancer. Biomed Pharmacother 2018; 108: 184–193. 70. Beg AA. ComPPARtmentalizing NF-κB in the gut. Nat Immunol 2004; 5(1): 14–16. 71. Fukata M, Michelsen KS, Eri R, et al. Toll-like receptor-4 is required for intestinal response to epithelial injury and limiting bacterial translocation in a murine model of acute colitis. Am J Physiol Gastrointest Liver Physiol 2005; 288(5): G1055–G1065. 72. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of Commensal Microflora by Toll-Like Receptors Is Required for Intestinal Homeostasis. Cell 2004; 118(2): 229–241. 73. Kelly D, Campbell JI, King TP, et al. Commensal anaerobic gut bacteria attenuate inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-γ and RelA. Nat Immunol 2004; 5(1): 104–112. 74. Wang P, Wu P, Siegel MI, et al. Interleukin (IL)-10 inhibits nuclear factor kappa B (NF kappa B) activation in human monocytes. IL-10 and IL-4 suppress cytokine synthesis by different mechanisms. J Biol Chem 1995; 270(16): 9558–9563. 75. Rubtsov YP, Rasmussen JP, Chi EY, et al. Regulatory T Cell-Derived Interleukin-10 Limits Inflammation at Environmental Interfaces. Immunity 2008; 28(4): 546–558. 76. Ewaschuk JB, Diaz H, Meddings L, et al. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am J Physiol Gastrointest Liver Physiol 2008; 295(5): G1025–G1034. 77. Suzuki Y, Orellana MA, Schreiber RD, et al. Interferon-gamma: the major mediator of resistance against Toxoplasma gondii. Science 1988; 240(4851): 516–518. 78. Sokol H, Pigneur B, Watterlot L, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A 2008; 105(43): 16731– 16736. PMMB 2023, 6, 1; a0000339 29 of 34 79. Louis P and Flint HJ. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol Lett 2009; 294(1): 1–8. 80. Duncan SH, Hold GL, Harmsen HJ, et al. Growth requirements and fermentation products of Fusobacterium prausnitzii, and a proposal to reclassify it as Faecalibacterium prausnitzii gen. nov., comb. nov. Int J Syst Evol Microbiol 2002; 52(6): 2141–2146. 81. Hamer HM, Jonkers D, Venema K, et al. Review article: the role of butyrate on colonic function. Aliment Pharmacol Ther 2008; 27(2): 104–119. 82. Nancey S, Bienvenu J, Coffin B, et al. Butyrate strongly inhibits in vitro stimulated release of cytokines in blood. Dig Dis Sci 2002; 47(4): 921–928. 83. Arribas B, Rodríguez-Cabezas ME, Camuesco D, et al. A probiotic strain of Escherichia coli, Nissle 1917, given orally exerts local and systemic anti-inflammatory effects in lipopolysaccharide-induced sepsis in mice. Br J Pharmacol 2009; 157(6): 1024–1033. 84. LeBlanc JG, Levit R, Savoy de Giori G, et al. Application of vitamin-producing lactic acid bacteria to treat intestinal inflammatory diseases. Appl Microbiol Biotechnol 2020; 104(8): 3331–3337. 85. Ménard S, Candalh C, Bambou JC, et al. Lactic acid bacteria secrete metabolites retaining anti-inflammatory properties after intestinal transport. Gut 2004; 53(6): 821–828. 86. Lin B, Ser HL, Wang L, et al. The Emerging Role of MMP12 in the Oral Environment. Int J Mol Sci 2023; 24(5): 4648. 87. Chang C-W, Lee H-C, Li L-H, et al. Fecal Microbiota Transplantation Prevents Intestinal Injury, Upregulation of Toll-Like Receptors, and 5-Fluorouracil/Oxaliplatin-Induced Toxicity in Colorectal Cancer. Int J Mol Sci 2020; 21(2): 386. 88. Carvalho PLA, Andrade MER, Trindade LM, et al. Prophylactic and therapeutic supplementation using fructo- oligosaccharide improves the intestinal homeostasis after mucositis induced by 5- fluorouracil. Biomed Pharmacother 2021; 133: 111012. 89. Ma TY, Iwamoto GK, Hoa NT, et al. TNF-α-induced increase in intestinal epithelial tight junction permeability requires NF-κB activation. Am J Physiol Gastrointest Liver Physiol 2004; 286(3): G367–G376. 90. Al-Sadi R, Ye D, Said HM, et al. IL-1β-Induced Increase in Intestinal Epithelial Tight Junction Permeability Is Mediated by MEKK-1 Activation of Canonical NF-κB Pathway. Am J Pathol 2010; 177(5): 2310–2322. 91. Liu Q, Nobaek S, Adawi D, et al. Administration of Lactobacillus plantarum 299v reduces side-effects of external radiation on colon anastomotic healing in an experimental model. Colorectal Dis 2001; 3(4): 245–252. 92. Qin HL, Shen TY, Gao ZG, et al. Effect of lactobacillus on the gut microflora and barrier function of the rats with abdominal infection. World J Gastroenterol 2005; 11(17): 2591–2596. 93. Moorthy G, Murali MR, and Devaraj SN. Lactobacilli facilitate maintenance of intestinal membrane integrity during Shigella dysenteriae 1 infection in rats. Nutrition 2009; 25(3): 350–358. 94. Stratiki Z, Costalos C, Sevastiadou S, et al. The effect of a bifidobacter supplemented bovine milk on intestinal permeability of preterm infants. Early Hum Dev 2007; 83(9): 575–579. 95. Ukena SN, Singh A, Dringenberg U, et al. Probiotic Escherichia coli Nissle 1917 inhibits leaky gut by enhancing mucosal integrity. PLoS One 2007; 2(12): e1308–e1308. 96. Johnson-Henry KC, Donato KA, Shen-Tu G, et al. Lactobacillus rhamnosus strain GG prevents enterohemorrhagic Escherichia coli O157:H7-induced changes in epithelial barrier function. Infect Immun 2008; 76(4): 1340–1348. PMMB 2023, 6, 1; a0000339 30 of 34 97. Shen S-R, Chen W-J, Chu H-F, et al. Amelioration of 5-fluorouracil-induced intestinal mucositis by Streptococcus thermophilus ST4 in a mouse model. PLoS One 2021; 16(7): e0253540–e0253540. 98. Macfarlane S and Macfarlane GT. Regulation of short-chain fatty acid production. Proc Nutr Soc 2003; 62(1): 67–72. 99. Savage DC, Siegel JE, Snellen JE, et al. Transit time of epithelial cells in the small intestines of germfree mice and ex-germfree mice associated with indigenous microorganisms. Appl Environ Microbiol 1981; 42(6): 996– 1001. 100. Rolls BA, Turvey A, and Coates ME. The influence of the gut microflora and of dietary fibre on epithelial cell migration in the chick intestine. Br J Nutr 1978; 39(1): 91–98. 101. Venkatraman A, Ramakrishna BS, and Pulimood AB. Butyrate hastens restoration of barrier function after thermal and detergent injury to rat distal colon in vitro. Scand J Gastroenterol 1999; 34(11): 1087–1092. 102. Yan F, Cao H, Cover TL, et al. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 2007; 132(2): 562–575. 103. Lv Y, Yang X, Huo Y, et al. Adenovirus-mediated Hepatocarcinoma-Intestine-Pancreas/Pancreatitis-Associated Protein Suppresses Dextran Sulfate Sodium-induced Acute Ulcerative Colitis in Rats. Inflamm Bowel Dis 2012; 18(10): 1950–1960. 104. Sougiannis AT, VanderVeen BN, Enos RT, et al. Impact of 5 fluorouracil chemotherapy on gut inflammation, functional parameters, and gut microbiota. Brain Behav Immun 2019; 80: 44–55. 105. Stringer AM, Gibson RJ, Logan RM, et al. Gastrointestinal Microflora and Mucins May Play a Critical Role in the Development of 5-Fluorouracil-Induced Gastrointestinal Mucositis. Exp Biol Med 2009; 234(4): 430–441. 106. Das B and Nair GB. Homeostasis and dysbiosis of the gut microbiome in health and disease. J Biosci 2019; 44(5): 117. 107. Sartor RB. Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology 2004; 126(6): 1620–1633. 108. Walter J and Ley R. The human gut microbiome: ecology and recent evolutionary changes. Annu Rev Microbiol 2011; 65: 411–429. 109. Lim WQ, Cheam JY, Law JW-F, et al. Role of Garlic in Chronic Diseases: Focusing on Gut Microbiota Modulation. Prog Microbes Mol Biol 2022; 5(1). 110. Wang Y and Kasper LH. The role of microbiome in central nervous system disorders. Brain Behav Immun 2014; 38: 1–12. 111. Thye AY-K, Bah Y-R, Law JW-F, et al. Gut–skin axis: Unravelling the connection between the gut microbiome and psoriasis. Biomedicines 2022; 10(5): 1037. 112. Kong GY-E, Letchumanan V, Tan LT-H, et al. Gut Microbiome in Obsessive Compulsive Disorder: Potential of Probiotics as an Adjuvant Therapy. Prog Microbes Mol Biol 2022; 5(1). 113. Thye AY, Law JW, Tan LTH, et al. Exploring the Gut Microbiome in Myasthenia Gravis. Nutrients 2022; 14(8): 1647. 114. Chong HY, Tan LT, Law JW, et al. Exploring the Potential of Human Milk and Formula Milk on Infants' Gut and Health. Nutrients 2022; 14(17): 3554. 115. Lim JM, Letchumanan V, Tan LT, et al. Ketogenic Diet: A Dietary Intervention via Gut Microbiome Modulation for the Treatment of Neurological and Nutritional Disorders (a Narrative Review). Nutrients 2022; 14(17): 3566. 116. Wang HJ, Battousse O, and Ramadas A. Modulation of gut microbiota by dietary macronutrients in type 2 diabetes: A review. Prog Microbes Mol Biol 2021; 4(1). PMMB 2023, 6, 1; a0000339 31 of 34 117. Durganaudu H, Kunasegaran T, and Ramadas A. Dietary glycaemic index and type 2 diabetes mellitus: Potential modulation of gut microbiota. Prog Microbes Mol Biol 2020; 3(1). 118. Tan LT-H, Letchumanan V, Law JW-F, et al. Exploring the effects of acupuncture therapy in restoring health via modulation of intestinal microbiota. in Gut. 2021. BMJ Publishing Group. 119. Letchumanan V, Vairavan KS, Law JW-F, et al. IDDF2023-ABS-0316 Exploring the covid-19 vaccination on autoimmune disease and probiotic administration in alleviating disease severity. in Gut. 2023. BMJ Publishing Group. 120. Law JW-F, Kong GY-E, Letchumanan V, et al. IDDF2023-ABS-0311 Looking into the gut microbiome of obsessive compulsive disorder: can probiotics help? in Gut. 2023. BMJ Publishing Group. 121. Learn-Han L, Letchumanan V, Law JW-F, et al. IDDF2022-ABS-0241 Exploring the potential role of probiotics in alleviating insomnia. in Gut. 2022. 122. Goh JX-H, Tan LT-H, Law JW-F, et al. IDDF2022-ABS-0231 The effect of probiotic supplementation in newly diagnosed type-1 diabetes mellitus patient: a systematic review of randomized controlled trials. in Gut. 2022. BMJ Publishing Group. 123. Letchumanan V, Low S-N, Tan LT-H, et al. IDDF2022-ABS-0212 Exploring the potential role of probiotics in substance use disorders. in Gut. 2022. BMJ Publishing Group. 124. Letchumanan V, Thye AY-K, Law JW-F, et al. IDDF2022-ABS-0207 The potential use of probiotics in alleviating psychological symptoms of long covid-19. in Gut. 2022. 125. Law JW-F, Thye AY-K, Letchumanan V, et al. IDDF2022-ABS-0200 Probiotics to improve preterm babies’ health outcomes: research in recent 10 years (2012–2022). in Gut. 2022. BMJ Publishing Group. 126. Picó-Monllor JA and Mingot-Ascencao JM. Search and Selection of Probiotics That Improve Mucositis Symptoms in Oncologic Patients. A Systematic Review. Nutrients 2019; 11(10): 2322. 127. Goh JXH, Tan LT-H, Law JW-F, et al. Probiotics: Comprehensive Exploration of the Growth Promotion Mechanisms in Shrimps. Prog Microbes Mol Biol 2023; 6(1). 128. Goh JXH, Tan LTH, Law JWF, et al. Harnessing the potentialities of probiotics, prebiotics, synbiotics, paraprobiotics, and postbiotics for shrimp farming. Reviews in Aquaculture 2022; 14(3): 1478–1557. 129. Selvaraj SM, Wong SH, Ser H-L, et al. Role of low FODMAP diet and probiotics on gut microbiome in irritable bowel syndrome (IBS). Prog Microbes Mol Biol 2020; 3(1). 130. Justino PFC, Melo LFM, Nogueira AF, et al. Treatment with Saccharomyces boulardii reduces the inflammation and dysfunction of the gastrointestinal tract in 5-fluorouracil-induced intestinal mucositis in mice. Br J Nutr 2014; 111(9): 1611–1621. 131. Kato S, Hamouda N, Kano Y, et al. 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(10): 1017–1025. 132. Chew S-S, Tan LT-H, Law JW-F, et al. Targeting Gut Microbial Biofilms—A Key to Hinder Colon Carcinogenesis? Cancers (Basel) 2020; 12(8): 2272. 133. Stringer AM, Al-Dasooqi N, Bowen JM, et al. Biomarkers of chemotherapy-induced diarrhoea: a clinical study of intestinal microbiome alterations, inflammation and circulating matrix metalloproteinases. Support Care Cancer 2013; 21(7): 1843–1852. 134. Yeung C-Y, Chiang Chiau J-S, Cheng M-L, et al. Modulations of probiotics on gut microbiota in a 5-fluorouracil- induced mouse model of mucositis. J Gastroenterol Hepatol 2020; 35(5): 806–814. PMMB 2023, 6, 1; a0000339 32 of 34 135. von Bültzingslöwen I, Adlerberth I, Wold AE, et al. Oral and intestinal microflora in 5-fluorouracil treated rats, translocation to cervical and mesenteric lymph nodes and effects of probiotic bacteria. Oral Microbiol Immunol 2003; 18(5): 278–284. 136. Coutinho JOPA, Quintanilha MF, Campos MRA, et al. Antarctic Strain of Rhodotorula mucilaginosa UFMGCB 18,377 Attenuates Mucositis Induced by 5-Fluorouracil in Mice. Probiotics Antimicrob Proteins 2021. 137. Carvalho RD, Breyner N, Menezes-Garcia Z, et al. Secretion of biologically active pancreatitis-associated protein I (PAP) by genetically modified dairy Lactococcus lactis NZ9000 in the prevention of intestinal mucositis. Microb Cell Factories 2017; 16(1): 27. 138. Carvalho R, Vaz A, Pereira FL, et al. Gut microbiome modulation during treatment of mucositis with the dairy bacterium Lactococcus lactis and recombinant strain secreting human antimicrobial PAP. Sci Rep 2018; 8(1): 15072. 139. Andoh A, Takaya H, Makino J, et al. Cooperation of interleukin-17 and interferon-gamma on chemokine secretion in human fetal intestinal epithelial cells. Clin Exp Immunol 2001; 125(1): 56–63. 140. Yeung C-Y, Chan W-T, Jiang C-B, et al. Amelioration of Chemotherapy-Induced Intestinal Mucositis by Orally Administered Probiotics in a Mouse Model. PLoS One 2015; 10(9): e0138746. 141. Huang L, Chiang Chiau J-S, Cheng M-L, et al. SCID/NOD mice model for 5-FU induced intestinal mucositis: Safety and effects of probiotics as therapy. Pediatr Neonatol 2019; 60(3): 252–260. 142. Barroso FAL, de Jesus LCL, de Castro CP, et al. Intake of Lactobacillus delbrueckii (pExu:hsp65) Prevents the Inflammation and the Disorganization of the Intestinal Mucosa in a Mouse Model of Mucositis. Microorganisms 2021; 9(1). 143. Levit R, Savoy de Giori G, de Moreno de LeBlanc A, et al. Folate-producing lactic acid bacteria reduce inflammation in mice with induced intestinal mucositis. J Appl Microbiol 2018; 125(5): 1494–1501. 144. Neurath MF, Fuss I, Kelsall BL, et al. Antibodies to interleukin 12 abrogate established experimental colitis in mice. J Exp Med 1995; 182(5): 1281–1290. 145. Mi H, Dong Y, Zhang B, et al. Bifidobacterium Infantis Ameliorates Chemotherapy-Induced Intestinal Mucositis Via Regulating T Cell Immunity in Colorectal Cancer Rats. Cell Physiol Biochem 2017; 42(6): 2330–2341. 146. Fernandes Cordeiro B, Rosa Oliveira E, Heloísa da Silva S, et al. Fermented milks, using Lactobacillus casei or Propionibacterium freudenreichii, prevent mucositis, a side effect of chemotherapy, in mice. in 22ème édition du colloque du Club des Bactéries Lactiques. 2019. Caen, France. 147. Smith CL, Geier MS, Yazbeck R, et al. Lactobacillus fermentum BR11 and Fructo-Oligosaccharide Partially Reduce Jejunal Inflammation in a Model of Intestinal Mucositis in Rats. Nutr Cancer 2008; 60(6): 757–767. 148. Whitford EJ, Cummins AG, Butler RN, 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(6): 505–511. 149. Ciobanu L, Tefas C, Oancea DM, et al. Effect of Lactobacillus plantarum ACTT 8014 on 5‑fluorouracil induced intestinal mucositis in Wistar rats. Exp Ther Med 2020; 20(6): 209. 150. Hu M, Wu X, Luo M, et al. Lactobacillus rhamnosus FLRH93 protects against intestinal damage in mice induced by 5-fluorouracil. J Dairy Sci 2020; 103(6): 5003–5018. 151. De Jesus LCL, Drumond MM, de Carvalho A, et al. Protective effect of Lactobacillus delbrueckii subsp. Lactis CIDCA 133 in a model of 5 Fluorouracil-Induced intestinal mucositis. J Funct Foods 2019; 53: 197–207. 152. Justino PF, Melo LF, Nogueira AF, et al. Regulatory role of Lactobacillus acidophilus on inflammation and gastric dysmotility in intestinal mucositis induced by 5-fluorouracil in mice. Cancer Chemother Pharmacol 2015; 75(3): 559–567. PMMB 2023, 6, 1; a0000339 33 of 34 153. Oh NS, Lee JY, Lee JM, et al. Mulberry leaf extract fermented with Lactobacillus acidophilus A4 ameliorates 5- fluorouracil-induced intestinal mucositis in rats. Lett Appl Microbiol 2017; 64(6): 459–468. 154. Yuan K-TY, 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(1): 113–118. 155. do Carmo FLR, Rabah H, Cordeiro BF, et al. Probiotic Propionibacterium freudenreichii requires SlpB protein to mitigate mucositis induced by chemotherapy. Oncotarget 2019; 10(68): 7198–7219. 156. Porto BAA, Monteiro CF, Souza ÉLS, et al. Treatment with selenium-enriched Saccharomyces cerevisiae UFMG A-905 partially ameliorates mucositis induced by 5-fluorouracil in mice. Cancer Chemother Pharmacol 2019; 84(1): 117–126. 157. Mauger CA, Butler RN, Geier MS, et al. Probiotic Effects on 5-Fluorouracil-Induced Mucositis Assessed by the Sucrose Breath Test in Rats. Dig Dis Sci 2007; 52(3): 612–619. 158. Tang Y, Wu Y, Huang Z, et al. Administration of probiotic mixture DM#1 ameliorated 5-fluorouracil–induced intestinal mucositis and dysbiosis in rats. Nutrition 2017; 33: 96–104. 159. Quaresma M, Damasceno S, Monteiro C, et al. Probiotic mixture containing Lactobacillus spp. and Bifidobacterium spp. attenuates 5-fluorouracil-induced intestinal mucositis in mice. Nutr Cancer 2020; 72(8): 1355–1365. 160. Strober W. Unraveling Gut Inflammation. Science 2006; 313(5790): 1052–1054. 161. Lycke NY and Bemark M. The regulation of gut mucosal IgA B-cell responses: recent developments. Mucosal Immunol 2017; 10(6): 1361–1374. 162. Wang Z, Xiao G, Yao Y, et al. The role of bifidobacteria in gut barrier function after thermal injury in rats. J Trauma 2006; 61(3): 650–657. 163. Wu ZQ, Han XD, Wang Y, et al. Interleukin-1 receptor antagonist reduced apoptosis and attenuated intestinal mucositis in a 5-fluorouracil chemotherapy model in mice. Cancer Chemother Pharmacol 2011; 68(1): 87–96. 164. Bowen JM, Gibson RJ, Cummins AG, et al. Intestinal mucositis: the role of the Bcl-2 family, p53 and caspases in chemotherapy-induced damage. Support Care Cancer 2006; 14(7): 713–731. 165. Justino PFC, Franco AX, Pontier-Bres R, et al. Modulation of 5-fluorouracil activation of toll-like/MyD88/NF- κB/MAPK pathway by Saccharomyces boulardii CNCM I-745 probiotic. Cytokine 2020; 125: 154791. 166. Specian RD and Oliver MG. Functional biology of intestinal goblet cells. Am J Physiol Cell Physiol 1991; 260(2): C183–C193. 167. Moncada DM, Kammanadiminti SJ, and Chadee K. Mucin and Toll-like receptors in host defense against intestinal parasites. Trends Parasitol 2003; 19(7): 305–311. 168. Johansson ME, Phillipson M, Petersson J, et al. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc Natl Acad Sci U S A 2008; 105(39): 15064–15069. 169. Lencer WI, Reinhart FD, and Neutra MR. Interaction of cholera toxin with cloned human goblet cells in monolayer culture. Am J Physiol 1990; 258(1 Pt 1): G96–G102. 170. Byrd JC, Yunker CK, Xu QS, et al. Inhibition of gastric mucin synthesis by Helicobacter pylori. Gastroenterology 2000; 118(6): 1072–1079. 171. Van der Sluis M, De Koning BA, De Bruijn AC, et al. Muc2-deficient mice spontaneously develop colitis, indicating that MUC2 is critical for colonic protection. Gastroenterology 2006; 131(1): 117–129. 172. Mack DR, Michail S, Wei S, et al. Probiotics inhibit enteropathogenic E. coli adherence in vitro by inducing intestinal mucin gene expression. Am J Physiol 1999; 276(4): G941–G950. PMMB 2023, 6, 1; a0000339 34 of 34 173. Mack DR, Ahrne S, Hyde L, et al. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut 2003; 52(6): 827–833. 174. Martín R, Chamignon C, Mhedbi-Hajri N, et al. The potential probiotic Lactobacillus rhamnosus CNCM I-3690 strain protects the intestinal barrier by stimulating both mucus production and cytoprotective response. Sci Rep 2019; 9(1): 5398. 175. Song M, Xia B, and Li J. Effects of topical treatment of sodium butyrate and 5-aminosalicylic acid on expression of trefoil factor 3, interleukin 1beta, and nuclear factor kappaB in trinitrobenzene sulphonic acid induced colitis in rats. Postgrad Med J 2006; 82(964): 130–135. 176. Saegusa Y, Ichikawa T, Iwai T, et al. Changes in the mucus barrier of the rat during 5-fluorouracil-induced gastrointestinal mucositis. Scand J Gastroenterol 2008; 43(1): 59–65. 177. Yeung C-Y, Chiau J-SC, Cheng M-L, et al. Immune Modulation Effects of Lactobacillus casei Variety rhamnosus on Enterocytes and Intestinal Stem Cells in a 5-FU-Induced Mucositis Mouse Model. Gastroenterol Res Pract 2021; 2021: 3068393. 178. Levit R, Savoy de Giori G, de Moreno de LeBlanc A, et al. Protective effect of the riboflavin-overproducing strain Lactobacillus plantarum CRL2130 on intestinal mucositis in mice. Nutrition 2018; 54: 165–172. 179. Wang H, Jatmiko YD, Bastian SEP, et al. Effects of Supernatants from Escherichia coli Nissle 1917 and Faecalibacterium prausnitzii on Intestinal Epithelial Cells and a Rat Model of 5-Fluorouracil-Induced Mucositis. Nutr Cancer 2017; 69(2): 307–318. 180. Ang W-S, Law JW-F, Letchumanan V, et al. A Keystone Gut Bacterium Christensenella minuta—A Potential Biotherapeutic Agent for Obesity and Associated Metabolic Diseases. Foods 2023; 12(13): 2485. 181. Vanlancker E, Vanhoecke B, Stringer A, et al. 5-Fluorouracil and irinotecan (SN-38) have limited impact on colon microbial functionality and composition in vitro. PeerJ 2017. 182. Kim, Younghoon, Sae-Hun K, et al. Inhibition of Escherichia coli O157:H7 Attachment by Interactions Between Lactic Acid Bacteria and Intestinal Epithelial Cells. J Microbiol Biotechnol 2008; 18(7): 1278–1285. 183. Marsova M, Odorskaya M, Novichkova M, et al. The Lactobacillus brevis 47 f Strain Protects the Murine Intestine from Enteropathy Induced by 5-Fluorouracil. Microorganisms 2020; 8(6). 184. Nasir SN, Ishak M, Ab Mutalib NS, et al. Circular RNA-EPHB4 As A Potential Biotarget In Colorectal Cancer: A Preliminary Analysis. Prog Microbes Mol Biol 2022; 5(1). 185. Yunos R-IM, Ab Mutalib N-S, Khor SS, et al. Whole exome sequencing identifies genomic alterations in proximal and distal colorectal cancer. Prog Microbes Mol Biol 2019; 2(1). 186. Bourlioux P, Koletzko B, Guarner F, et al. The intestine and its microflora are partners for the protection of the host: report on the Danone Symposium “The Intelligent Intestine,” held in Paris, June 14, 2002. Am J Clin Nutr 2003; 78(4): 675–683. Author(s) shall retain the copyright of their work and grant the Journal/Publisher right for the first publication with the work simultaneously licensed under: Creative Commons Attribution-NonCommercial 4.0 International (CC BY-NC 4.0). This license allows for the copying, distribution and transmission of the work, provided the correct attribution of the original creator is stated. Adaptation and remixing are also permitted.