Next Article in Journal
Peculiarities of Scattering of Ultrashort Laser Pulses on DNA and RNA Trinucleotides
Next Article in Special Issue
Characterization of Mucosal-Associated Invariant T Cells in Oral Lichen Planus
Previous Article in Journal
Top-Down Preparation of Nanoquartz for Toxicological Investigations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Experimental Chemotherapy-Induced Mucositis: A Scoping Review Guiding the Design of Suitable Preclinical Models

Melbourne Dental School, The University of Melbourne, Carlton, VIC 3053, Australia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(23), 15434; https://doi.org/10.3390/ijms232315434
Submission received: 11 October 2022 / Revised: 1 December 2022 / Accepted: 4 December 2022 / Published: 6 December 2022

Abstract

:
Mucositis is a common and most debilitating complication associated with the cytotoxicity of chemotherapy. The condition affects the entire alimentary canal from the mouth to the anus and has a significant clinical and economic impact. Although oral and intestinal mucositis can occur concurrently in the same individual, these conditions are often studied independently using organ-specific models that do not mimic human disease. Hence, the purpose of this scoping review was to provide a comprehensive yet systematic overview of the animal models that are utilised in the study of chemotherapy-induced mucositis. A search of PubMed/MEDLINE and Scopus databases was conducted to identify all relevant studies. Multiple phases of filtering were conducted, including deduplication, title/abstract screening, full-text screening, and data extraction. Studies were reported according to the updated Preferred Reporting Items for Systematic reviews and Meta-Analyses Extension for Scoping Reviews (PRISMA-ScR) guidelines. An inter-rater reliability test was conducted using Cohen’s Kappa score. After title, abstract, and full-text screening, 251 articles met the inclusion criteria. Seven articles investigated both chemotherapy-induced intestinal and oral mucositis, 198 articles investigated chemotherapy-induced intestinal mucositis, and 46 studies investigated chemotherapy-induced oral mucositis. Among a total of 205 articles on chemotherapy-induced intestinal mucositis, 103 utilised 5-fluorouracil, 34 irinotecan, 16 platinum-based drugs, 33 methotrexate, and 32 other chemotherapeutic agents. Thirteen articles reported the use of a combination of 5-fluorouracil, irinotecan, platinum-based drugs, or methotrexate to induce intestinal mucositis. Among a total of 53 articles on chemotherapy-induced oral mucositis, 50 utilised 5-fluorouracil, 2 irinotecan, 2 methotrexate, 1 topotecan and 1 with other chemotherapeutic drugs. Three articles used a combination of these drugs to induce oral mucositis. Various animal models such as mice, rats, hamsters, piglets, rabbits, and zebrafish were used. The chemotherapeutic agents were introduced at various dosages via three routes of administration. Animals were mainly mice and rats. Unlike intestinal mucositis, most oral mucositis models combined mechanical or chemical irritation with chemotherapy. In conclusion, this extensive assessment of the literature revealed that there was a large variation among studies that reproduce oral and intestinal mucositis in animals. To assist with the design of a suitable preclinical model of chemotherapy-induced alimentary tract mucositis, animal types, routes of administration, dosages, and types of drugs were reported in this study. Further research is required to define an optimal protocol that improves the translatability of findings to humans.

1. Introduction

Alimentary tract mucositis is a major adverse effect of patients receiving cancer chemotherapy treatments [1]. It is collectively referred to as chemotherapy-induced mucosal injury that affects the entire alimentary canal from the mouth to the anus, where oral mucositis (OM) and intestinal mucositis (IM) refer to painful inflammation and ulceration of the oral cavity and lower GI tract, respectively. Symptoms of this debilitating condition include ulceration, oral and abdominal pain, and can lead to a delay or even cessation of cancer treatment. Ancillary associated complications include anorexia, vomiting, and diarrhoea that remain a significant burden for 40–80% of patients undergoing chemotherapy treatment. Such obstructions to food and water intake may lead to weight loss [2].
There is currently no effective single intervention that can prevent or treat mucositis [3]. Being able to alleviate the symptoms of mucositis is important for several reasons. Firstly, this condition affects approximately 80% of cancer patients receiving chemotherapy and radiotherapy. Secondly, pain and decreased oral function often persist for a long period of time after therapy has concluded. Thirdly, mucositis increases mortality and morbidity which can contribute to rising healthcare costs [1]. However, despite mucositis’s significant clinical and economic impact, agents used in its management (anti-inflammatories, biologic response modifiers, cytoprotectants antimicrobials, antifungal) are generally only palliative, with few agents (palifermin and benzydamine) approved to date.
The exact pathogenic mechanisms of chemotherapy-induced alimentary tract mucositis are unknown, and this highlights that preclinical models are critical for the development of novel mechanism-based treatments. Mucositis involves multiple signalling pathways and processes that result in mucosal and luminal modifications to the intestine [4]. As such, many avenues have been explored to understand the multifactorial nature of mucositis but have not resulted in the development of effective mechanism-based management strategies.
We believe that the development of novel interventions for the benefit of patients with chemotherapy-associated mucosal injury are limited partly due to the use of inconsistent preclinical models. Furthermore, although alimentary tract mucositis occurs concurrently in the same individual, oral and intestinal mucositis are often studied separately using different, organ-specific models. Hence, this scoping review aims to synthesise the currently available evidence of mucositis models and interpret the data generated. In particular, we aim to assess the published literature describing animal models of mucositis to document suitable preclinical models that can be used to assess the efficacy of novel interventions for the prevention and/or treatment of chemotherapy-induced mucosal injury of the alimentary tract in vivo.

2. Results

2.1. Search Results

The search of Scopus and PubMed databases produced 478 results in total. Twelve duplicates were removed via Endnote. Automation tools were used in Scopus and PubMed to exclude non-English, nonhuman, and review articles and 323 articles remained. These publications’ title and abstract were screened in accordance with the eligibility criteria, and 276 articles remained and were sought for retrieval. A total of 9 articles were not retrievable, and 267 articles remained. After reading the full text of the 267 articles, 16 articles were further excluded. The remaining 251 articles were assessed and included in the qualitative synthesis. The study selection process is summarised in Figure 1.
Due to the large number of included articles, the data extraction sheet is provided in the Supplementary Materials (Table S1). Details include: reference number, authors and year, title, population, intervention, control/comparator, and outcomes (PICO), biomarker, methodology and sample type, assessment of mucosal damage, details of treatment or intervention, and major findings.
For clarity, the results reported below were stratified on the basis of the alimentary tract segment involved (oral or intestinal) and drug used.

2.2. Chemotherapy-Induced Intestinal Mucositis

2.2.1. Assessment of Intestinal Mucositis

Metabolic data including body weight and food/water intake and disease activity parameters including diarrhoea and rectal bleeding were included in most of the studies. Diarrhoea severity was measured by stool consistency, and body weight was expressed as weight loss over the experimental period. Stool consistency was commonly assessed via a scoring system, in which score 0 was normal stool and score 3 and 4 corresponded to severe diarrhoea and watery stool. Most studies reported that diarrhoea severity increased and body weight decreased during the course of intestinal mucositis.
The most common methodology used amongst the studies for histopathological and morphometric evaluation, documented in 188/205 (91.7%) of the studies, was haematoxylin–eosin staining (H&E) and subsequent examination by light microscopy. Villus height and crypt depth were measured to assess their histological changes in most studies. Villus height was measured from the baseline to the villus tip and crypt depth was measured from the baseline to the crypt bottom. The majority of studies found a decrease in villus height and either increase or decrease in crypt depth in the chemotherapy-treated group. In addition, various histopathological score systems were used in different studies to examine the severity of intestinal mucositis. Although each score system contains different criteria, the common pattern is that zero means normal or no damage and severity increases as the number gets larger. Histologic criteria described in the literature include: the morphology of the villi (blunting, atrophy), crypt architecture, vacuolization, crypt necrosis, an infiltration of inflammatory cells, a disruption of brush border and surface enterocytes, a dilation of lymphatics and capillaries, oedema, intestinal epithelium architecture, crypt damage (surviving crypts per millimetre and surviving cells per crypt), the presence of haemorrhagic areas, goblet cell numbers, crypt abscess formation, enterocytes mitotic figures, a thickening of the submucosal and muscularis externa layers, and the intensity of inflammation.
A macroscopic examination of the intestinal tissues was also employed in several studies and different score systems were used. The components described in the literature consist of inflammatory aspects such as erythema, haemorrhagic areas, epithelial ulcerations and abscesses, hyperaemia, and the size of the ulcerative area. Small intestine length, colon length, and mucosal thickness were also included in several studies.
Additionally, various biomarkers were investigated in the literature to help determine the severity of intestinal mucositis, with the most common being inflammatory biomarkers. The inflammatory markers investigated in the literature include IL-1β, IL-4, IL-5, IL-6, IL-10, IL-13, tumour necrosis factor (TNF-α), interferon-γ (IFN-γ), monocyte chemoattractant protein-1 (MCP-1), NF-κB, TGF-β, myeloperoxidase (MPO), CXCL1, CXCL3, and CXCL9. The most common methods to assess the change in inflammatory cytokines level such as TNF-α and IL-1β and anti-inflammatory cytokines such as IL-4 and IL-10 are real-time PCR and enzyme-linked immunosorbent assay (ELISA). Most studies found an increased level of inflammatory cytokines and a reduction in anti-inflammatory cytokines after chemotherapeutic treatment. Interestingly, one study reported an increase of IL-4 and IL-10, at 31.5% and 39.6%, respectively [5]. MPO activity was measured by a predetermined method in other studies. Briefly, the most common protocol used was adding a solution of o-dianisidine dihydrochloride or o-dianisidine hydrochloride and hydrogen peroxide into the intestinal tissue supernatant. The MPO activity was measured by the change in absorbance at 450 nm. The level of MPO activity increased overall during the course of intestinal mucositis. Apoptosis markers were also used to assess the level of apoptosis in the crypt cells. The terminal deoxyribonucleotide transferase (TdT)-mediated nick-end labelling (TUNEL) assay was the most common method to quantify the apoptotic cells in the intestinal crypt, calculated by dividing the number of apoptotic cells by the total number of cells in the selected villi or crypt. The most common method used to assess the apoptotic assay was using an in situ apoptosis detection kit followed by IHC. The number of apoptotic cells during the course of intestinal mucositis was found to be increased in the majority of studies. The expression of caspase-3 and the Bax/Bcl-2 ratio were also used to determine the enterocyte survival. The most common method used to assess the level of caspase-3 was immunohistochemistry. IHC, western blotting, and RT-PCR were documented for the assessment of Bax and Bcl-2 levels. The third category of biomarkers investigated in intestinal mucositis are intestinal oxidative stress biomarkers, which include a number of antioxidants: malonaldehyde (MDA), glutathione (GSH), catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx). The methods used to measure the level of oxidative stress markers were pre-established by other studies. Most studies reported a decrease in GSH, CAT, and SOD and an increase in MDA. However, one study reported a 3.81-fold increase in the level of GSH after chemotherapy [6]. Intestinal proliferation markers were also employed in some studies to assess the effect of mucositis on epithelial intestinal cells, including proliferating cell nuclear antigen (PCNA) and Ki-67. The most common method to measure PCNA and Ki-67 was via IHC. Most studies found a negative effect of chemotherapy on PCNA and Ki-67 and the reduction of PCNA positive cells. Two studies reported a significant increase in Ki-67 positive cells after FOLFOX chemotherapeutic regimen [7,8]. Sucrase activity was used by several studies as an indicator for small intestinal damage, which was measured by a 13C-sucrose breath test. Biomarkers were also used to assess the level of intestinal permeability as a result of mucositis; such biomarkers include the tight junction molecules ZO-1, occludin, junctional adhesion molecule-A, and Claudin-2, as well as other biomarkers such as diamine oxidase (DAO) and endotoxin. Some studies also suggested that DAO could be used to quantify intestinal mucosal injury [9,10]. The most common method to assess the level of ZO-1 and occludin was via IHC and RT-PCR and most studies found that chemotherapy induced negative changes to ZO-1 and occludin. ELISA was most commonly used to determine the level of DAO. A reduction of plasma DAO was reported in the majority of studies. It was also found that there was a dose–response relationship between DAO and the dosage of 5-Fluorouracil [11].

2.2.2. Animal Models of Intestinal Mucositis Related to Specific Chemotherapeutic Drugs

5-Fluorouracil (5-FU)

A total of 103 articles documented 5-FU-induced intestinal mucositis in animal models. A summary of findings is presented in Table 1. Of the 103 articles, 72 used mouse models, 30 studies used rats, and 1 used domestic pig models. Different dosages and routes of administration of the drug were employed. Most articles used intraperitoneal (i.p.) injection with a dosage range between 25 and 450 mg/kg. In fewer studies, mucositis was induced by 5-FU with a range of 20–50 mg/kg orally by gavage or dropsonde into the oesophagus of animal models, except pig models, who were fed by oral bolus. Only three articles used intravenous (i.v.) injection or infusion with a range of 50–200 mg/kg. In general, a similar dosage range of 5-FU was found among three types of animal models, although an outlier at 100 ng/kg was found in one paper reporting a combination of 5-FU with doxorubicin [8]. Moreover, the dosage of each injection chosen in these articles was influenced by the number of injections. In particular, most studies adopted a single dosage of 5-FU of 150 mg/kg and above to induce intestinal mucositis, whereas multiple injections were made with dosages of 100 mg/kg or less. A summary of the articles is presented in Table 1.

Irinotecan

A total of 34 articles produced drug-induced gastrointestinal mucositis in rat and mouse models with irinotecan. Among the 34 articles, 22 articles used mouse samples whilst the remaining 12 articles used rat samples. The concentration of irinotecan used varied among these studies, from a minimum of 10 mg/kg to a maximum of 270 mg/kg via i.p. on mouse models, and a minimum of 20 mg/kg to 200 mg/kg via intraperitoneal injection on rat models. The use of irinotecan was found to decrease the body weight and food intake of the animals in these experimental models. Diarrhoea was often seen as a result of dose-limiting side effects. A summary of the articles is shown in Table 2.

Platinum-Based Chemotherapy Drugs

Platinum-based drugs that were used included cisplatin, carboplatin, and oxaliplatin. A total of 251 in vivo articles were reviewed, and 16 articles reporting drug-induced gastrointestinal mucositis on rat and mouse models with platinum-based chemotherapy drugs were included. A summary of the articles is shown in Table 3. Among the 16 articles, 10 articles used mouse models whilst the remaining 6 articles used rats. For the mouse models, only one article used carboplatin (100 mg/kg intraperitoneal injection), five articles used cisplatin (2 mg/kg i.p. to 11 mg/kg i.p.), and four articles used oxaliplatin (1 mg/kg i.p. to 5 mg/kg i.p.). For the rat models, the five articles used cisplatin, from a minimum of 5 mg/kg i.p. to 7 mg/kg intraperitoneally. Aside from mucositis, other side effects such as diarrhoea, vomiting, and reduced body weight were often observed. A summary of the articles is shown in Table 3.

Methotrexate

Among the 251 included articles, 33 articles documented animal models of methotrexate-induced intestinal mucositis. Within the 33 articles, 6 articles reported the use of mouse models, and 27 articles reported the use of rat models. A summary of the articles is shown in Table 4.
In the six articles reporting murine models, four articles reported intraperitoneal (i.p.) injection of methotrexate (MTX) with a dosage ranging from 20 mg/kg to 500 mg/kg. The remaining two articles used subcutaneous (s.c.) injection with a dosage of 12.5 mg/kg.
In the 27 articles documenting a rat model, nine articles reported i.p. injection of MTX with a concentration ranging from 2.5 mg/kg to 90 mg/kg. Six articles used s.c. injection with a dosage ranging from 1.5 mg/kg to 3.5 mg/kg. Five articles documented the use of intramuscular (i.m.) injection of MTX with a dosage of 1.5 mg/kg. Five articles reported intravenous (i.v.) injection of MTX at a dosage from 20 mg/kg to 150 mg/kg. The remaining two articles reported oral intake of MTX at a dosage of 5 mg/kg.

Other Chemotherapeutic Agents

A total of 32 articles reported intestinal mucositis induced by other chemotherapeutic agents, including doxorubicin, capecitabine, afatinib, SN38, ailanthone, melphalan, busulfan, cyclophosphamide, paclitaxel, cytarabine (Ara-C), etoposide, ifosfamide, epirubicin, dioscrin, and S-1, a combination of 1-(2-tetrahydrofuryl)-5-fluorouracil (FT), 5-chloro-2,4 dihydroxypyridine (CDHP), and potassium oxonate (Oxo). A summary of the results is presented in Supplementary Table S2.
Doxorubicin: a mouse model was used in 10 publications [12,184,185,186,187,188,189,190,191,192], with a dosage ranging from 10 to 20 mg/kg intraperitoneally. Two articles documented the use of a rat model with a dosage of 20 mg/kg i.p. [132,193] and one article used a piglet model (100 mg/m2 surface area i.v.) [194].
Capecitabine: two articles reported a rat and murine model, respectively, both with a dosage of 500 mg/kg orally [195,196].
Afatinib and SN38: one article reported the use of both afatinib and SN38 in a zebrafish model, with a dosage ranging from 10 to 40 µg/g orally and 10 to 40 µg/g intraperitoneally, respectively [197].
Ailanthone: one article documented the use of ailanthone in a mouse model with a dosage of 2–39.8 mg/kg orally [198].
Melphalan: one publication reported a mouse model with a concentration from 85.5 to 95.7 mg/m2 i.p. [199].
Busulfan: a piglet model was used in one article with a dosage at 12.8 mg/kg i.v. [200]. A mouse model was documented in one article with a dosage at 40 mg/kg, but the route of administration was not specified [201].
Cyclophosphamide: five articles reported a mouse model with a dosage ranging from 50 to 550 mg/kg i.p. [127,202,203,204,205]. One article reported a rat model with a dosage at 120 mg/kg i.p. [132] and one article reported a piglet model with a dosage at 120 mg/kg i.v. [200].
Paclitaxel: mouse models were employed in one article, with a dosage ranging from 2 to 4 mg/kg i.v. [206].
Cytarabine (Ara-C): one article used a mouse model with a drug concentration at 3.6 mg/mouse i.p. [207] and one article reported a rat model with a dosage at 30 mg/kg s.c. [9].
Etoposide: a rat model was used in one publication with a dosage at 40 mg/kg i.p. [132].
Ifosfamide: one article reported the use of a rabbit model with a dosage ranging from 30 to 60 mg/kg i.v. [208].
Epirubicin: one study reported the use of epirubicin as a chemotherapeutic agent in a mouse model at a dosage of 12 mg/kg i.p. [209].
Dioscrin: dioscrin at a dosage of 60 mg/kg administered intragastrically was documented in one study using a rat model [148].
S-1: S-1, an oral fluorouracil comprising 1-(2-tetrahydrofuryl)-5-fluorouracil (FT), 5-chloro-2,4-dihydroxypyridine (CDHP), and potassium oxonate (Oxo) in a molecular ratio of 1:0.4:1, was reported to be used in a rat model at a dosage of 20 mg/kg orally [10].

2.3. Chemotherapy-Induced Oral Mucositis

2.3.1. Assessment of Oral Mucositis

The most common method used to evaluate histopathological features of oral mucositis was the H&E staining of the oral mucosa, tongue tissues, or cheek pouch, documented in 35/52 articles (67.3%). In the literature, various histological score systems were used to classify the severity of mucositis, -including the epithelial architecture, membrane integrity, intensity of inflammatory cell infiltration, connective tissue organisation, oedema of the submucosa, intensity of inflammation, vasodilation, haemorrhage, oedema, ulcers, and abscesses.
In addition, a macroscopic examination was used in a number of studies to assess mucosal injury. The macroscopic features used in the literature were similar to the criteria employed in the assessment of intestinal mucositis, which included inflammatory aspects such as erythema, hyperaemia, haemorrhagic areas, epithelial ulcerations, and abscesses, hyperaemia, and the dimension/area of the ulcerative area. Oral epithelial thickness and cheek pouch thickness were also used. Overall, a varying degree of reduction in epithelial thickness, architecture, and structure were detected. An increase in the abundance of inflammatory infiltrates was commonly reported.
The use of biomarkers was similar to the studies on intestinal mucositis. The categories of biomarkers documented included inflammatory cytokines (MPO, TGF-β, IL-1β, NF-κB, TNF-α, IL-1, IL-6), apoptosis markers (Bcl-2, caspase-3), proliferation markers (Ki-67, PCNA), and oxidative stress markers (MDA, GSH, SOD, CAT). The most common method to assess the change in inflammatory cytokines level such as TNF-α and IL-1β and anti-inflammatory cytokines such as IL-6 and IL-10 is an enzyme-linked immunosorbent assay (ELISA). Most studies found an increased level of inflammatory cytokines and a reduction in anti-inflammatory cytokines after chemotherapeutic treatment. MPO activity was measured with a similar protocol to MPO assays addressed in intestinal mucositis. The level of MPO activity increased overall during the course of oral mucositis. Metabolic data such as a reduction in body weight were also included in most of the studies.

2.3.2. Animal Models of Oral Mucositis Related to Specific Chemotherapeutic Drugs

5-Fluorouracil

A total of 50 studies documented oral mucositis induced by 5-FU in animal models. A summary of findings is presented in Table 5. Of the 50 articles, 13 used mice, 8 studies used rats and 29 used hamsters. Different dosages and routes of administration of the drug were employed, most articles reported using an intraperitoneal (i.p.) injection with a dosage range between 10 and 150 mg/kg body weight. One article reported i.v. administration of 5-fluorouracil [44], and one study did not specify the route of administration [210]. The total dosage of injections was dependent on the number of injections given to the model. Most studies adopted a double dosage of 5-FU at 40 mg/kg and then 60 mg/kg in subsequent days.

Irinotecan

A total of two articles reviewed utilised a drug-induced model of oral mucositis using irinotecan. Both these studies were on rats and the concentration of the drug used was 200 mg/kg body weight at a single dose. In both studies, irinotecan was administered intraperitoneally. One study utilised a multiple intervention model using irinotecan, methotrexate, and 5-FU [108]. The other study used irinotecan only as the intervention [254].

Topotecan

Of the oral mucositis models, only one used topotecan to induce mucositis. This article utilised rabbits as the animal model. The concentration of the drug used was 0.5 mg/kg of body weight and was administered via intravenous bolus [255].

Methotrexate

A total of two articles utilised methotrexate to induce a model of oral mucositis. Both of these studies were on rats and the concentration of the drug utilised was 1.5 mg/kg of body weight, administered intramuscularly [88,108].

Other Chemotherapeutic Agents

One article reported the use of a combination of drugs including busulfan and cyclophosphamide on pig models. Busulfan and cyclophosphamide were administered intravenously with a total dosage of 12.8 mg/kg and 120 mg/kg, respectively [200].

2.3.3. Induction of Oral Mucositis in Animal Models

Mechanical-, chemical-, or radiation-induced injury of the oral mucosal surface is additionally required in most existing chemotherapy-induced oral mucositis animal models when chemotherapy is intraperitonially administered. Of the 53 articles reporting oral mucositis, 33 articles introduced mechanical irritation to the cheek pouch such as superficial scratching with the tip of an 18-gauge needle. Eight articles reported the use of a chemical agent such as acetic acid for ulcer induction in animal models. Two articles report i.v. administration of 5-FU to induce oral mucositis in mice without any additional stimuli [44,142]. Another article also reported i.v. administration of topotecan to induce oral mucositis in rabbits without any additional stimuli [255]. Nine articles did not report any form of additional stimulating treatment other than the i.p. chemotherapeutic agents. However, when i.p. administration of 5-FU was used without additional intervention to induce oral mucositis, no visible ulcerations were recorded or observed in the oral cavity. A summary of the findings is presented in Table 6.

3. Discussion

Our scoping review includes data from 251 publications involving chemotherapy-induced intestinal and oral mucositis in animal models. Importantly, the vast majority of these models did not produce intestinal and oral mucositis together. Only seven articles [44,70,88,99,108,109,200] were chemotherapy-induced alimentary tract mucositis.
For chemotherapy-induced intestinal mucositis models, the review found that the most commonly used animals were mice, while the most commonly used drug was 5-FU. The most common route of administration was intraperitoneal (i.p.) injection (25–450 mg/kg).While this method is beneficial because it allows the absorption of large amounts of the intervention rapidly, the disadvantage is that the drug can have a large variability in effectiveness and misinjection [256]. Another problem associated with the i.p. mouse model is that the intestinal mucosa is more sensitive to chemotherapy compared to the upper alimentary tract such as the oral cavity. As a result, the intestinal function deteriorates quickly, and mice need to be euthanized. This usually occurs before any oral lesions develop, which makes it difficult to study oral mucositis [257]. For oral mucositis models, the most common animal models used were hamsters with i.p. injection of 5-FU (40–100 mg/kg). Most models (75%) applied additional mechanical or chemical irritation with chemotherapeutic drugs to induce oral mucositis. Hence, the majority of animal models did not reproduce a common clinical scenario in that they did not use the same route of administration of the antineoplastic agent as in patients and/or applied additional, nonphysiological stimuli.

3.1. Translatability of Models

Preclinical models evaluated in the literature mainly induced intestinal mucositis. There was no standardisation to the dosages administered, and the literature also reported that the dosage of each injection depended on the number of injections. In humans, the dosage for chemotherapy treatments is dependent on individual tolerances to the treatment. Thus, most dosages are calculated based on human surface area, while the dosage calculation for preclinical models is dependent on weight. As such, for the preclinical data to be translatable, a standardised conversion is required. For example, in the FOLFIRI regimen for colorectal cancer, the standard dosage of 5-FU is 2400 mg/m2, and it was calculated that a dosage of 400 mg/kg in rats approximated 2222 mg/m2 in humans [103]. However, the 400 mg/kg dosage sits in the high end of the dosage range reported in the literature, and most studies had doses lower than 400 mg/kg. Nevertheless, the equivalent dosage of human doses to other animal models still needs more research. Therefore, whether a lower dosage of chemotherapeutic drugs used in animal models can mimic human disease is questionable.
While 52 articles demonstrated the establishment of chemotherapy-induced oral mucositis, mechanical or chemical irritation was commonly used in the literature to induce oral mucositis in these animal models, in addition to the i.p. administration of chemotherapeutic agents. Only one study [44], where the chemotherapeutic agents were administered intravenously, produced ulceration lesions in the oral cavity and replicated oral and intestinal mucositis both macroscopically and histologically, without using any additional stimuli. It was suggested that intraperitoneal 5-FU injection in mouse models can induce intestinal mucositis reproducibly but did not affect the oral mucosa significantly, hence local mechanical trauma or chemical injury were included in animal oral mucositis models [257]. This may be likely attributed to the highly keratinized nature of oral epithelium in mice, thus rendering it less susceptible to mucosa breakage [258]. Furthermore, i.p. administration increases the concentration of chemotherapy in the peritoneal cavity and targets primarily organs in the peritoneal cavity, thus toxicity in these organs exceeds that at other sites [257]. It was supported by Yamaguchi et al. that chemotherapeutic drugs did not induce oral mucositis directly and an additional mechanical or chemical injury was needed [99]. However, with the addition of a mechanical or chemical injury, whether the direct effects of chemotherapeutic drugs on oral mucosa can be examined is questionable. The standardisation of such mechanical and chemical injury protocols is difficult [257]. Whether the additional mucosa manipulation mimics the course of oral mucositis in humans is also questionable. Bertolini et al., studied the possibility of inducing oral mucositis without additional stimuli [257]. It was found that a high daily dose of 5-FU i.p. was unable to induce oral mucositis, but the mouse model with 50 mg/kg 5-FU i.v. injected every 48 h for 13 days showed macroscopical and histological features and an inflammatory cytokine profile consistent with oral mucositis [257].

3.2. Pathophysiology

The pathophysiology of intestinal mucositis might involve a combination of villi length and crypt depth changes, oxidative stress, apoptosis, inflammatory reactions, a proliferative capacity of the intestinal cells, and the composition of the gut microbiome [8]. Sonis et al. proposed a five-stage model of the pathogenesis of mucositis induced by chemotherapeutic agents, which included initiation, upregulation and message generation, signalling and amplification, ulceration and inflammation, and the healing stage [259].
The production of reactive oxygen species and the increase in oxidative stress were suggested to contribute to the initiation phase, subsequently leading to cell death and DNA damage [260]. Rtibi et al., found that the antioxidant biomarkers SOD, CAT, and GPx decreased in 5-FU and CAP-induced intestinal mucositis in a rat model, confirming the role of oxidative stress in the onset of intestinal mucositis [195]. Consistently, the use of antioxidants has been found to reduce the severity of mucosal injury in vitro, in vivo, and in clinical studies [261].
The second phase is characterised by the involvement of inflammatory components, with NF-kB being the key molecule. The activation of proinflammatory cytokines can lead to tissue injury and cell apoptosis in both the intestinal crypt and oral mucosa [259]. Multiple studies examined the role of inflammatory cytokines in the pathophysiology of mucositis. Logan et al., reported an increase in NF-κB, TNF, IL-1β, and IL-6 serum concentration following administration of 5-FU, MTX, and irinotecan in an alimentary tract rat model, but the serum concentration changes followed histological changes in most instances. Both oral and intestinal histological changes were investigated in these studies [108]. It was also found that inflammasome activation following oxidative stress contributed to the pathogenesis of an irinotecan-induced intestinal mucositis mice model. Inflammasome activation was crucial to activate IL-1β and IL-18 [118].
The third stage is characterised by the amplification of inflammatory response by proinflammatory cytokines such as TNF-α [259]. In a 5-FU-induced oral mucositis hamster model, TNF-α inhibitors were found to reduce the severity of oral mucositis. TNF-α can modulate the expression of other cytokines such as IL-1β and IL-6 and affect the apoptosis and survival of other cells, hence it was postulated that TNF-α was important in the pathogenesis of oral mucositis [228].
The fourth ulcerative phase is considered to be the culmination of the mucositis process. The apoptosis of epithelial stem cells and tissue injury as a result of inflammation and oxidative stress result in the breakdown of mucosa and the loss of epithelial integrity observed in oral and intestinal mucositis. The breakdown of mucosa not only allows bacteria translocation, but the bacterial products can further enhance the inflammatory response by activating macrophages in the local environment.
The last healing phase is characterised by a restoration of the proliferation capacity and local microbiome [259]. In a 5-FU-induced intestinal mucositis rat model, bacterial translocation to the mesenteric lymph nodes was significant after 5-FU administration and was speculated to be the result of inflammatory cytokines release and mucosal injury [93]. Al-Azri et al. reported the involvement of matrix metalloproteinases MMP-3 in the initiation of inflammatory reactions and it may be associated with the production of NF-κB and inflammatory cytokines [254]. MMP-9 might be involved in the last healing phase in the 5-phase model of mucositis [254]. Sonis et al., summarised that the mechanisms of oral and gastrointestinal tract mucositis were likely to be similar, but further research was still required [259].

3.3. Optimal Dosage

From the publications that were included in the current scoping review, there were large differences between studies in terms of the dosage used and injection regime to induce mucositis in animal models, hence the total doses injected varied widely. Such variations were largest in intraperitoneal drug administration. One of the studies included in the present review by Zhang et al., investigated the optimal dosage range of a total of five dosages of 5-FU to induce intestinal mucositis in mouse models [11]. The study proposed the criteria of the optimal animal model as follows: it should closely mimic the mucosal injury in humans, and it should have an acceptable survival rate in order to perform any treatment and investigations on these animal models. The study suggested that five dosages of 50 mg/kg to 100 mg/kg i.p. per day fulfilled the above criteria, and a dosage lower or higher than this concentration could either result in insufficient mucosal injury or high mortality rate [11]. Another study conducted by Fijlstra et al., investigated the optimal dosage of a single dose of MTX in a rat model in a pilot experiment. The criteria were similar to Zhang et al., which included pronounced mucosal injury and low mortality rate. The experiment found that the severity of mucositis increased with increasing dosage, but the mortality rate increased at the same time [11]. They proposed a single dosage of 60 mg/kg of MTX i.v. was the optimal dosage to induce intestinal mucositis in a rat model [174]. The standardised optimal dosage of different chemotherapeutic drugs and the route of administration in different animal models require further research.

3.4. Limitations

Our study attempted to gain a clear understanding of the advancements made in animal models of mucositis, with the eventual aim to translate such results in humans. Our review assessed the current understanding of interventions against mucositis-induced preclinical models. However, our exclusion criteria, such as excluding non-English articles or search string (excluding radiotherapy or radiation in the title or abstract) could mean that we missed out on potential information that could provide us the complete bigger picture of this condition.
Within the articles themselves, there were some limitations. For example, many articles did not have enough statistical power as the sample size was generally low. Some studies reported n < 5 per experimental group. Furthermore, the risk of selection bias could not be ruled out among animal models. As the baseline characteristics such as the species, age, weight, and gender were not accounted for when determining drug concentrations. Future studies can iterate on these limitations by exploring the optimal dosage required, such as in terms of single or multiple doses over a period of time to mimic mucosal injury in humans while maintaining a high survival rate.

4. Methods

4.1. Protocol and Search Strategy

The results of the scoping review are reported in accordance with the updated Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines [5]. The following search strategy was used to search for related articles in Scopus and PubMed databases and was conducted in May 2022:
(((mucositis[Title/Abstract] OR mucosal injury[Title/Abstract]) AND (oral[Title/Abstract] OR intestinal[Title/Abstract] OR alimentary[Title/Abstract])) AND (chemotherap*[Title/Abstract] OR cancer treatment[Title/Abstract] OR cancer therapy[Title/Abstract] OR antineoplas*[Title/Abstract])) AND (mouse[Title/Abstract] OR mice[Title/Abstract] OR rat[Title/Abstract] OR hamster*[Title/Abstract] OR preclinical[Title/Abstract] OR pre-clinical[Title/Abstract] OR animal*[Title/Abstract]) NOT radiotherapy NOT radiation.

4.2. Eligibility Criteria

Articles were included in the review if they met the following criteria: (a) any peer-reviewed article presenting data such as original article, short communication, and research letter, (b) articles written in English; and (c) research using animals that were administered traditional chemotherapeutic agents. Articles with the following criteria were excluded: (a) articles not reporting original data such as reviews and systematic reviews; (b) articles not documenting animal models of chemotherapy-induced mucositis; (c) in vitro or human studies; (d) articles documenting immunotherapy-induced mucositis; and (e) articles not assessing mucosal damage in animal models.

4.3. Data Selection and Collection

Before title/abstract screening, automation tools in Scopus and PubMed were used to exclude review articles, nonhuman studies, and non-English articles. The articles were then deduplicated. A total of 5 reviewers were involved in the screening process. Each of the remaining articles were screened by 2 independent reviewers by reading the title and abstract. Articles were included or excluded according to the eligibility criteria. Cohen’s Kappa score was calculated to be 95.4, which showed a strong agreement between assessor pairs. Any discrepancy in the title/abstract screening between the two reviewers was resolved after discussion with the research supervisor (N. C.). A full-text screening for the included articles after title/abstract screening was then performed. Each article was screened and assessed against the eligibility criteria. Data extraction was performed and tabulated for the included articles. The extracted parameters were author, year, title, population studied, intervention used to induce mucositis, control, outcome and measurement, biomarkers, methodology and sample type, assessment of mucosal damage, details of treatment or intervention, and major findings.

5. Conclusions

The main purpose of this scoping review was to document and evaluate evidence from the entirety of the literature on chemotherapy-induced mucositis animal models. These in vivo models are used to elucidate the pathophysiology of mucositis and to develop potential interventions to alleviate or prevent mucositis. Our analysis showed that there were consistent data that may inform the development of a more standardised animal model that can help identify the pathophysiology of chemotherapy-induced mucositis. These models may further be used to detect, prevent, or ameliorate the mucosal toxicity of antineoplastic treatments. To help design a chemotherapy-induced alimentary tract mucositis animal model, all the studies reported here included animal types, routes of administration, dosages, and types of drugs. The results suggest that the use of a rodent model of chemotherapy reproducing the clinical and histologic features of both oral and intestinal mucositis without additional noxious stimuli (e.g., repeated cycles of 50 mg/kg 5-FU intravenously) may represent a useful in vivo preclinical model for studying chemotherapy-induced alimentary tract mucositis. However, there is still a large variation among studies and further research is required to define optimal experimental conditions for a reproducible animal model that is suitable for preclinical studies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms232315434/s1, Table S1: Data extraction table; Table S2: Summary of data collected from In Vivo animal studies involving intestinal mucositis induced by other chemotherapeutic agents.

Author Contributions

Conceptualization, methodology, supervision, and writing—review and editing, N.C.; formal analysis, data curation, writing—original draft preparation, J.H., A.Y.M.H., Y.J., B.K. and M.I.; writing—review and editing, A.I.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Full datasets are available upon reasonable request to the corresponding author.

Acknowledgments

The authors would like to acknowledge the support of the University of Melbourne for making the necessary bibliographic resources available.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rubenstein, E.B.; Peterson, D.E.; Schubert, M.; Keefe, D.; McGuire, D.; Epstein, J.; Elting, L.S.; Fox, P.C.; Cooksley, C.; Sonis, S.T.; et al. Clinical practice guidelines for the prevention and treatment of cancer therapy-induced oral and gastrointestinal mucositis. Cancer 2004, 100, 2026–2046. [Google Scholar] [CrossRef] [PubMed]
  2. Blijlevens, N.; Donnelly, J.; De Pauw, B. Mucosal barrier injury: Biology, pathology, clinical counterparts and consequences of intensive treatment for haematological malignancy: An overview. Bone Marrow Transplant. 2000, 25, 1269–1278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Knox, J.J.; Puodziunas, A.L.; Feld, R. Chemotherapy-induced oral mucositis. Drugs Aging 2000, 17, 257–267. [Google Scholar] [CrossRef] [PubMed]
  4. Chen, C.; Zhang, Q.; Yu, W.; Chang, B.; Le, A. Oral Mucositis: An Update on Innate Immunity and New Interventional Targets. J. Dent. Res. 2020, 99, 1122–1130. [Google Scholar] [CrossRef] [PubMed]
  5. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Int. J. Surg. 2021, 88, 105906. [Google Scholar] [CrossRef]
  6. Gautam, R.; Singh, M.; Gautam, S.; Rawat, J.K.; A Saraf, S.; Kaithwas, G. Rutin attenuates intestinal toxicity induced by Methotrexate linked with anti-oxidative and anti-inflammatory effects. BMC Complement. Altern. Med. 2016, 16, 99. [Google Scholar] [CrossRef] [Green Version]
  7. Chang, C.-W.; Lee, H.-C.; Li, L.-H.; Chiau, J.-S.C.; Wang, T.-E.; Chuang, W.-H.; Chen, M.-J.; Wang, H.-Y.; Shih, S.-C.; Liu, C.-Y.; 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, 386. [Google Scholar] [CrossRef] [Green Version]
  8. Chang, C.-W.; Liu, C.-Y.; Lee, H.-C.; Huang, Y.-H.; Li, L.-H.; Chiau, J.-S.C.; Wang, T.-E.; Chu, C.-H.; Shih, S.-C.; Tsai, T.-H.; et al. Lactobacillus casei Variety rhamnosus Probiotic Preventively Attenuates 5-Fluorouracil/Oxaliplatin-Induced Intestinal Injury in a Syngeneic Colorectal Cancer Model. Front. Microbiol. 2018, 9, 983. [Google Scholar] [CrossRef]
  9. Luk, G.D.; Vaughan, W.P.; Burke, P.J.; Baylin, S.B. Diamine oxidase as a plasma marker of rat intestinal mucosal injury and re-generation after administration of 1-β-D-arabinofuranosylcytosine. Cancer Res. 1981, 41, 2334–2337. [Google Scholar]
  10. Moriyama, K.; Kouchi, Y.; Morinaga, H.; Irimura, K.; Hayashi, T.; Ohuchida, A.; Goto, T.; Yoshizawa, Y. Diamine oxidase, a plasma biomarker in rats to GI tract toxicity of oral fluorouracil anti-cancer drugs. Toxicology 2006, 217, 233–239. [Google Scholar] [CrossRef]
  11. Zhang, S.; Liu, Y.; Xiang, D.; Yang, J.; Liu, D.; Ren, X.; Zhang, C. Assessment of dose-response relationship of 5-fluorouracil to murine intestinal injury. Biomed. Pharmacother. 2018, 106, 910–916. [Google Scholar] [CrossRef] [PubMed]
  12. Cardani, D.; Sardi, C.; La Ferla, B.; D’Orazio, G.; Sommariva, M.; Marcucci, F.; Olivero, D.; Tagliabue, E.; Koepsell, H.; Nicotra, F.; et al. Sodium glucose cotransporter 1 ligand BLF501 as a novel tool for management of gas-trointestinal mucositis. Mol. Cancer 2014, 13, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Zhan, Y.; Xu, C.; Liu, Z.; Yang, Y.; Tan, S.; Jiang, J.; Liu, H.; Chen, J.; Wu, B. β-Arrestin1 inhibits chemotherapy-induced intestinal stem cell apoptosis and mucositis. Cell Death Dis. 2016, 7, e2229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kato, S.; Hamouda, N.; Kano, Y.; Oikawa, Y.; Tanaka, Y.; Matsumoto, K.; Amagase, K.; Shimakawa, M. Probiotic Bifidobacterium bifidum G9-1 attenuates 5-fluorouracil-induced intestinal mucositis in mice via suppression of dysbiosis-related secondary inflammatory responses. Clin. Exp. Pharmacol. Physiol. 2017, 44, 1017–1025. [Google Scholar] [CrossRef]
  15. Soares, P.M.G.; Lima-Junior, R.C.P.; Mota, J.M.S.C.; Justino, P.F.C.; Brito, G.A.C.; Ribeiro, R.A.; Cunha, F.Q.; Souza, M.H.L.P. Role of plate-let-activating factor in the pathogenesis of 5-fluorouracil-induced intestinal mucositis in mice. Cancer Chemother Pharmacol. 2011, 68, 713–720. [Google Scholar] [CrossRef]
  16. Wu, Z.-Q.; Han, X.-D.; Wang, Y.; Yuan, K.-L.; Jin, Z.-M.; Di, J.-Z.; Yan, J.; Pan, Y.; Zhang, P.; Huang, X.-Y.; et al. Interleukin-1 receptor antagonist reduced apoptosis and attenuated intestinal mucositis in a 5-fluorouracil chemotherapy model in mice. Cancer Chemother. Pharmacol. 2010, 68, 87–96. [Google Scholar] [CrossRef]
  17. Sano, T.; Utsumi, D.; Amagase, K.; Matsumoto, K.; Tominaga, M.; Higuchi, K.; Takeuchi, T.; Kato, S. Lafutidine, a histamine H2 receptor antagonist with mucosal protective properties, attenuates 5-fluorouracil-induced intestinal mucositis in mice through activation of extrinsic primary afferent neurons. J. Physiol. Pharmacol. 2017, 68, 79–90. [Google Scholar]
  18. Yasuda, M.; Kato, S.; Yamanaka, N.; Iimori, M.; Utsumi, D.; Kitahara, Y.; Iwata, K.; Matsuno, K.; Amagase, K.; Yabe-Nishimura, C.; et al. Potential role of the NADPH oxidase NOX1 in the pathogenesis of 5-fluorouracil-induced intestinal mucositis in mice. Am. J. Physiol. Liver Physiol. 2012, 302, G1133–G1142. [Google Scholar] [CrossRef] [Green Version]
  19. Justino, P.F.C.; Melo, L.F.M.; Nogueira, A.F.; Morais, C.M.; Mendes, W.O.; Franco, A.X.; Souza, E.P.; Ribeiro, R.A.; Souza, M.H.L.P.; Soares, P.M.G. Regulatory role of Lactobacillus acidophilus on inflammation and gastric dysmotility in intestinal mucositis induced by 5-fluorouracil in mice. Cancer Chemother. Pharmacol. 2015, 75, 559–567. [Google Scholar] [CrossRef]
  20. Yasuda, M.; Kato, S.; Yamanaka, N.; Iimori, M.; Matsumoto, K.; Utsumi, D.; Kitahara, Y.; Amagase, K.; Horie, S.; Takeuchi, K. 5-HT3 receptor antagonists ameliorate 5-fluorouracil-induced intestinal mucositis by suppression of apoptosis in murine intestinal crypt cells. J. Cereb. Blood Flow Metab. 2012, 168, 1388–1400. [Google Scholar] [CrossRef] [Green Version]
  21. Gou, H.; Gu, L.; Shang, B.; Xiong, Y.; Wang, C. Protective effect of Bu-Zhong-Yi-Qi decoction, the water extract of Chinese traditional herbal medicine, on 5-fluorouracil-induced intestinal mucositis in mice. Hum. Exp. Toxicol. 2016, 35, 1243–1251. [Google Scholar] [CrossRef] [PubMed]
  22. Wu, Z.; Han, X.; Qin, S.; Zheng, Q.; Wang, Z.; Xiang, D.; Zhang, J.; Lu, H.; Wu, M.; Zhu, S.; et al. Interleukin 1 receptor antagonist reduces lethality and intestinal toxicity of 5-Fluorouracil in a mouse mucositis model. Biomed. Pharmacother. 2011, 65, 339–344. [Google Scholar] [CrossRef] [PubMed]
  23. Zhan, Y.S.; Tan, S.W.; Mao, W.; Jiang, J.; Liu, H.L.; Wu, B. Chemotherapy mediates intestinal injury via p53/p53 upregulated mod-ulator of apoptosis (PUMA) signaling pathway. J. Dig. Dis. 2014, 15, 425–434. [Google Scholar] [CrossRef] [PubMed]
  24. Jain, U.; Midgen, C.A.; Woodruff, T.M.; Schwaeble, W.J.; Stover, C.M.; Stadnyk, A.W. Properdin deficiency protects from 5-fluorouracil-induced small intestinal mucositis in a complement activation-independent, interleukin-10-dependent mechanism. Clin. Exp. Immunol. 2017, 188, 36–44. [Google Scholar] [CrossRef] [Green Version]
  25. Huang, T.-Y.; Chu, H.-C.; Lin, Y.-L.; Ho, W.-H.; Hou, H.-S.; Chao, Y.-C.; Liao, C.-L. Minocycline attenuates 5-fluorouracil-induced small intestinal mucositis in mouse model. Biochem. Biophys. Res. Commun. 2009, 389, 634–639. [Google Scholar] [CrossRef]
  26. Coutinho, J.O.P.A.; Quintanilha, M.F.; Campos, M.R.A.; Ferreira, E.; de Menezes, G.C.A.; Rosa, L.H.; Rosa, C.A.; Vital, K.D.; Fernandes, S.O.A.; Cardoso, V.N.; et al. Antarctic Strain of Rhodotorula mucilaginosa UFMGCB 18,377 At-tenuates Mucositis Induced by 5-Fluorouracil in Mice. Probiotics Antimicrob. Proteins 2022, 14, 486–500. [Google Scholar] [CrossRef]
  27. Gao, J.; Gao, J.; Qian, L.; Wang, X.; Wu, M.; Zhang, Y.; Ye, H.; Zhu, S.; Yu, Y.; Han, W. Activation of p38-MAPK by CXCL4/CXCR3 axis contributes to p53-dependent intestinal apoptosis initiated by 5-fluorouracil. Cancer Biol. Ther. 2014, 15, 982–991. [Google Scholar] [CrossRef] [Green Version]
  28. Azevedo, O.; Oliveira, R.A.C.; Oliveira, B.C.; Zaja-Milatovic, S.; Araújo, C.V.; Wong, D.V.T.; Costa, T.B.; Lucena, H.B.M.; Lima-Júnior, R.C.P.; A Ribeiro, R.; et al. Apolipoprotein E COG 133 mimetic peptide improves 5-fluorouracil-induced intestinal mucositis. BMC Gastroenterol. 2012, 12, 35. [Google Scholar] [CrossRef] [Green Version]
  29. Zeeshan, M.; Atiq, A.; Ain, Q.U.; Ali, J.; Khan, S.; Ali, H. Evaluating the mucoprotective effects of glycyrrhizic acid-loaded polymeric nanoparticles in a murine model of 5-fluorouracil-induced intestinal mucositis via suppression of inflammatory mediators and oxidative stress. Inflammopharmacology 2021, 29, 1539–1553. [Google Scholar] [CrossRef]
  30. Hamouda, N.; Sano, T.; Oikawa, Y.; Ozaki, T.; Shimakawa, M.; Matsumoto, K.; Amagase, K.; Higuchi, K.; Kato, S. 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, 159–168. [Google Scholar] [CrossRef] [Green Version]
  31. Matsumoto, K.; Nakajima, T.; Sakai, H.; Kato, S.; Sagara, A.; Arakawa, K.; Tashima, K.; Narita, M.; Horie, S. Increased Expression of 5-HT3 and NK1 Receptors in 5-Fluorouracil-Induced Mucositis in Mouse Jejunum. Am. J. Dig. Dis. 2013, 58, 3440–3451. [Google Scholar] [CrossRef] [PubMed]
  32. Yoneda, J.; Nishikawa, S.; Kurihara, S. Oral administration of cystine and theanine attenuates 5-fluorouracil-induced intestinal mucositis and diarrhea by suppressing both glutathione level decrease and ROS production in the small intestine of mu-cositis mouse model. BMC Cancer 2021, 21, 1343. [Google Scholar] [CrossRef] [PubMed]
  33. Maioli, T.U.; Silva, B.D.M.; Dias, M.N.; Paiva, N.C.; Cardoso, V.N.; Fernandes, S.O.; Carneiro, C.M.; Martins, F.D.S.; Generoso, S.D.V. Pretreatment with Saccharomyces boulardii does not prevent the experimental mucositis in Swiss mice. J. Negat. Results Biomed. 2014, 13, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Koizumi, R.; Azuma, K.; Izawa, H.; Morimoto, M.; Ochi, K.; Tsuka, T.; Imagawa, T.; Osaki, T.; Ito, N.; Okamoto, Y.; et al. Oral Administration of Surface-Deacetylated Chitin Nanofibers and Chitosan Inhibit 5-Fluorouracil-Induced Intestinal Mucositis in Mice. Int. J. Mol. Sci. 2017, 18, 279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Magalhães, T.A.F.M.; de Souza, M.O.; Gomes, S.V.; e Silva, R.M.; Martins, F.D.S.; de Freitas, R.N.; Amaral, J.F.D. Açaí (Euterpe oleracea Martius) Promotes Jejunal Tissue Regeneration by Enhancing Antioxidant Response in 5-Fluorouracil-Induced Mucositis. Nutr. Cancer 2021, 73, 523–533. [Google Scholar] [CrossRef] [PubMed]
  36. Sumiyoshi, M.; Suzuki, T.; Kimura, Y. Protective effects of water-soluble low-molecular-weight beta-(1,3-1,6)d-glucan purified from Aureobasidium pullulans GM-NH-1A1 against UFT toxicity in mice. J. Pharm. Pharmacol. 2009, 61, 795–800. [Google Scholar]
  37. Huang, F.S.; Kemp, C.J.; Williams, J.L.; Erwin, C.R.; Warner, B.W. Role of epidermal growth factor and its receptor in chemothera-py-induced intestinal injury. Am. J. Physiol. Gastrointest. Liver Physiol. 2002, 282, G432–G442. [Google Scholar] [CrossRef]
  38. Kato, S.; Hayashi, S.; Kitahara, Y.; Nagasawa, K.; Aono, H.; Shibata, J.; Utsumi, D.; Amagase, K.; Kadowaki, M. Saireito (TJ-114), a Japanese traditional herbal medicine, reduces 5-fluorouracil-induced intestinal mucositis in mice by inhibiting cyto-kine-mediated apoptosis in intestinal crypt cells. PLoS ONE 2015, 10, e0116213. [Google Scholar] [CrossRef]
  39. Tucker, J.M.; Davis, C.; E Kitchens, M.; A Bunni, M.; Priest, D.G.; Spencer, H.; Berger, F.G. Response to 5-fluorouracil chemotherapy is modified by dietary folic acid deficiency in ApcMin/+ mice. Cancer Lett. 2002, 187, 153–162. [Google Scholar] [CrossRef]
  40. Chen, K.-J.; Huang, Y.-L.; Kuo, L.-M.; Chen, Y.-T.; Hung, C.-F.; Hsieh, P.-W. Protective role of casuarinin from Melastoma malabath-ricum against a mouse model of 5-fluorouracil-induced intestinal mucositis: Impact on inflammation and gut microbiota dysbiosis. Phytomed. Int. J. Phytother. Phytopharm. 2022, 101, 154092. [Google Scholar]
  41. Boushey, R.P.; Yusta, B.; Drucker, D.J. Glucagon-like peptide (GLP)-2 reduces chemotherapy-associated mortality and enhances cell survival in cells expressing a transfected GLP-2 receptor. Cancer Res. 2001, 61, 687–693. [Google Scholar] [PubMed]
  42. Yue, X.; Wen, S.; Long-Kun, D.; Man, Y.; Chang, S.; Min, Z.; Shuang-Yu, L.; Xin, Q.; Jie, M.; Liang, W. Three important short-chain fatty acids (SCFAs) attenuate the inflammatory response induced by 5-FU and maintain the integrity of intestinal mucosal tight junction. BMC Immunol. 2022, 23, 19. [Google Scholar] [CrossRef] [PubMed]
  43. Kim, K.-A.; Kakitani, M.; Zhao, J.; Oshima, T.; Tang, T.; Binnerts, M.; Liu, Y.; Boyle, B.; Park, E.; Emtage, P.; et al. Mitogenic Influence of Human R-Spondin1 on the Intestinal Epithelium. Science 2005, 309, 1256–1259. [Google Scholar] [CrossRef] [PubMed]
  44. Bertolini, M.; Ranjan, A.; Thompson, A.; Diaz, P.I.; Sobue, T.; Maas, K.; Dongari-Bagtzoglou, A. Candida albicans induces mucosal bacterial dysbiosis that promotes invasive infection. PLoS Pathog. 2019, 15, e1007717. [Google Scholar] [CrossRef] [PubMed]
  45. Oliveira, M.M.B.; de Araújo, A.A.; Ribeiro, S.B.; Mota, P.C.M.d.S.; Marques, V.B.; Rebouças, C.d.S.M.; Figueiredo, J.G.; Barra, P.B.; Brito, G.A.D.C.; Leitão, R.F.D.C.; et al. Losartan improves intestinal mucositis induced by 5-fluorouracil in mice. Sci. Rep. 2021, 11, 23241. [Google Scholar] [CrossRef]
  46. Wang, L.; Wang, R.; Wei, G.-Y.; Wang, S.-M.; Du, G.-H. Dihydrotanshinone attenuates chemotherapy-induced intestinal mucositis and alters fecal microbiota in mice. Biomed. Pharmacother. 2020, 128, 110262. [Google Scholar] [CrossRef]
  47. Wang, L.; Song, B.; Hu, Y.; Chen, J.; Zhang, S.; Chen, D.; Wang, J. Puerarin Ameliorates 5-Fluorouracil–Induced Intestinal Mucositis in Mice by Inhibiting JAKs. J. Pharmacol. Exp. Ther. 2021, 379, 147–155. [Google Scholar] [CrossRef]
  48. Cai, B.; Pan, J.; Chen, H.; Chen, X.; Ye, Z.; Yuan, H.; Sun, H.; Wan, P. Oyster polysaccharides ameliorate intestinal mucositis and improve metabolism in 5-fluorouracil-treated S180 tumour-bearing mice. Carbohydr. Polym. 2021, 256, 117545. [Google Scholar] [CrossRef]
  49. Yeung, C.; Chiau, J.C.; Cheng, M.; Chan, W.; Chang, S.; Chang, Y.; Jiang, C.; Lee, H. Modulations of probiotics on gut microbiota in a 5-fluorouracil-induced mouse model of mucositis. J. Gastroenterol. Hepatol. 2020, 35, 806–814. [Google Scholar] [CrossRef]
  50. Wenqin, D.; Yaodong, Z.; Wanji, S.; FengLi, Z.; Li, S.; Haili, J.; Ping, L.; Mei, Z. Armillariella Oral Solution Ameliorates Small Intestinal Damage in a Mouse Model of Chemotherapy-Induced Mucositis. Nutr. Cancer 2019, 71, 1142–1152. [Google Scholar] [CrossRef]
  51. Huang, L.; Chiau, J.-S.C.; Cheng, M.-L.; Chan, W.-T.; Jiang, C.-B.; Chang, S.-W.; Yeung, C.-Y.; Lee, H.-C. SCID/NOD mice model for 5-FU induced intestinal mucositis: Safety and effects of probiotics as therapy. Pediatr. Neonatol. 2019, 60, 252–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Yan, X.-X.; Li, H.-L.; Zhang, Y.-T.; Wu, S.-Y.; Lu, H.-L.; Yu, X.-L.; Meng, F.-G.; Sun, J.-H.; Gong, L.-K. A new recombinant MS-superoxide dismutase alleviates 5-fluorouracil-induced intestinal mucositis in mice. Acta Pharmacol. Sin. 2020, 41, 348–357. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, Z.; Xie, W.; Li, M.; Teng, N.; Liang, X.; Zhang, Z.; Yang, Z.; Wang, X. Oral Administration of Polaprezinc Attenuates Fluoroura-cil-induced Intestinal Mucositis in a Mouse Model. Basic Clin. Pharmacol. Toxicol. 2017, 121, 480–486. [Google Scholar] [CrossRef] [PubMed]
  54. Chartier, L.C.; Howarth, G.S.; Mashtoub, S. Chemotherapy-induced mucositis development in a murine model of coli-tis-associated colorectal cancer. Scand. J. Gastroenterol. 2020, 55, 47–54. [Google Scholar] [CrossRef] [PubMed]
  55. Ferreira, T.M.; Leonel, A.J.; Melo, M.A.; Santos, R.R.G.; Cara, D.C.; Cardoso, V.N.; Correia, M.I.T.D.; Alvarez-Leite, J.I. Oral Supplementation of Butyrate Reduces Mucositis and Intestinal Permeability Associated with 5-Fluorouracil Administration. Lipids 2012, 47, 669–678. [Google Scholar] [CrossRef] [PubMed]
  56. Bastos, R.; Pedroso, S.; Vieira, A.; Moreira, L.; França, C.; Cartelle, C.; Arantes, R.; Generoso, S.; Cardoso, V.; Neves, M.; et al. Saccharomyces cerevisiae UFMG A-905 treatment reduces intestinal damage in a murine model of irinotecan-induced mucositis. Benef. Microbes 2016, 7, 549–557. [Google Scholar] [CrossRef]
  57. Shen, S.-R.; Chen, W.-J.; Chu, H.-F.; Wu, S.-H.; Wang, Y.-R.; Shen, T.-L. Amelioration of 5-fluorouracil-induced intestinal mucositis by Streptococcus thermophilus ST4 in a mouse model. PLoS ONE 2021, 16, e0253540. [Google Scholar] [CrossRef]
  58. Wang, X.; Zhu, S.; Qian, L.; Gao, J.; Wu, M.; Zhang, Y.; Chan, G.L.; Yu, Y.; Han, W. IL-1Ra selectively protects intestinal crypt epithelial cells, but not tumor cells, from chemotoxicity via p53-mediated upregulation of p21WAF1 and p27KIP1. Pharmacol. Res. 2014, 82, 21–33. [Google Scholar] [CrossRef]
  59. Zhao, G.; Williams, J.; Washington, M.K.; Yang, Y.; Long, J.; Townsend, S.D.; Yan, F. 2′-Fucosyllactose Ameliorates Chemothera-py-Induced Intestinal Mucositis by Protecting Intestinal Epithelial Cells Against Apoptosis. Cell. Mol. Gastroenterol. Hepatol. 2022, 13, 441–457. [Google Scholar] [CrossRef]
  60. Carvalho, R.D.; Breyner, N.; Menezes-Garcia, Z.; Rodrigues, N.M.; Lemos, L.; Maioli, T.U.; Souza, D.D.G.; Carmona, D.; de Faria, A.M.C.; Langella, P.; 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, 27. [Google Scholar] [CrossRef] [Green Version]
  61. Ali, J.; Khan, A.U.; Shah, F.A.; Ali, H.; Islam, S.U.; Kim, Y.S.; Khan, S. Mucoprotective effects of Saikosaponin-A in 5-fluorouracil-induced intestinal mucositis in mice model. Life Sci. 2019, 239, 116888. [Google Scholar] [CrossRef] [PubMed]
  62. Levit, R.; De Giori, G.S.; Leblanc, A.D.M.D.; Leblanc, J.G. Folate-producing lactic acid bacteria reduce inflammation in mice with induced intestinal mucositis. J. Appl. Microbiol. 2018, 125, 1494–1501. [Google Scholar] [CrossRef] [PubMed]
  63. Levit, R.; de Giori, G.S.; LeBlanc, A.D.M.D.; LeBlanc, J.G. Protective effect of the riboflavin-overproducing strain Lactobacillus plantarum CRL2130 on intestinal mucositis in mice. Nutrition 2018, 54, 165–172. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, C.; Yang, S.; Gao, L.; Wang, L.; Cao, L. Carboxymethyl pachyman (CMP) reduces intestinal mucositis and regulates the intestinal microflora in 5-fluorouracil-treated CT26 tumour-bearing mice. Food Funct. 2018, 9, 2695–2704. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, X.; Gao, J.; Qian, L.; Zhu, S.; Wu, M.; Zhang, Y.; Guan, W.; Ye, H.; Yu, Y.; Han, W. Exogenous IL-1Ra attenuates intestinal mucositis induced by oxaliplatin and 5-fluorouracil through suppression of p53-dependent apoptosis. Anti-Cancer Drugs 2015, 26, 35–45. [Google Scholar] [CrossRef] [PubMed]
  66. Fideles, L.d.S.; de Miranda, J.A.L.; Martins, C.d.S.; Barbosa, M.L.L.; Pimenta, H.B.; Pimentel, P.V.d.S.; Teixeira, C.S.; Scafuri, M.A.S.; Façanha, S.d.O.; Barreto, J.E.F.; et al. Role of Rutin in 5-Fluorouracil-Induced Intestinal Mucositis: Prevention of Histological Damage and Reduction of Inflammation and Oxidative Stress. Molecules 2020, 25, 2786. [Google Scholar] [CrossRef]
  67. Leocádio, P.C.L.; Antunes, M.M.; Teixeira, L.G.; Leonel, A.J.; Alvarez-Leite, J.I.; Machado, D.C.C.; Generoso, S.V.; Cardoso, V.N.; Correia, M.I.T.D. L-Arginine Pretreatment Reduces Intestinal Mucositis as Induced by 5-FU in Mice. Nutr. Cancer 2015, 67, 486–493. [Google Scholar] [CrossRef]
  68. Levit, R.; de Giori, G.S.; LeBlanc, A.D.M.D.; LeBlanc, J.G. Evaluation of vitamin-producing and immunomodulatory lactic acid bacteria as a potential co-adjuvant for cancer therapy in a mouse model. J. Appl. Microbiol. 2021, 130, 2063–2074. [Google Scholar] [CrossRef]
  69. Chen, Y.; Zheng, H.; Zhang, J.; Wang, L.; Jin, Z.; Gao, W. Protective effect and potential mechanisms of Wei-Chang-An pill on high-dose 5-fluorouracil-induced intestinal mucositis in mice. J. Ethnopharmacol. 2016, 190, 200–211. [Google Scholar] [CrossRef]
  70. Li, C.-H.; Ko, J.-L.; Ou, C.-C.; Lin, W.-L.; Yen, C.-C.; Hsu, C.-T.; Hsiao, Y.-P. The Protective Role of GMI, an Immunomodulatory Protein from Ganoderma microsporum, on 5-Fluorouracil-Induced Oral and Intestinal Mucositis. Integr. Cancer Ther. 2019, 18, 1534735419833795. [Google Scholar] [CrossRef] [Green Version]
  71. Zhang, L.; Jin, Y.; Peng, J.; Chen, W.; Lisha, L.; Lin, J. Qingjie Fuzheng Granule attenuates 5-fluorouracil-induced intestinal mucosal damage. Biomed. Pharmacother. 2019, 118, 109223. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, K.-J.; Chen, Y.-L.; Ueng, S.-H.; Hwang, T.-L.; Kuo, L.-M.; Hsieh, P.-W. Neutrophil elastase inhibitor (MPH-966) improves intes-tinal mucosal damage and gut microbiota in a mouse model of 5-fluorouracil-induced intestinal mucositis. Biomed. Pharmacother. 2021, 134, 111152. [Google Scholar] [CrossRef] [PubMed]
  73. Lee, J.M.; Yoo, I.K.; Lee, J.M.; Kim, S.H.; Choi, H.S.; Kim, E.S.; Keum, B.; Seo, Y.S.; Jeen, Y.T.; Chun, H.J.; et al. Dipeptidyl-peptidase-4 (DPP-4) inhibitor ameliorates 5-flurouracil induced intestinal mucositis. BMC Cancer 2019, 19, 1016. [Google Scholar] [CrossRef] [PubMed]
  74. Hytting-Andreasen, R.; Balk-Møller, E.; Hartmann, B.; Pedersen, J.; Windeløv, J.A.; Holst, J.J.; Kissow, H. Endogenous glucagon-like peptide- 1 and 2 are essential for regeneration after acute intestinal injury in mice. PLoS ONE 2018, 13, e0198046. [Google Scholar] [CrossRef] [PubMed]
  75. Chen, G.; Zeng, H.; Li, X.; Liu, J.; Li, Z.; Xu, R.; Ma, Y.; Liu, C.; Xue, B. Activation of G protein coupled estrogen receptor prevents chemotherapy-induced intestinal mucositis by inhibiting the DNA damage in crypt cell in an extracellular signal-regulated kinase 1- and 2- dependent manner. Cell Death Dis. 2021, 12, 1034. [Google Scholar] [CrossRef] [PubMed]
  76. Kim, H.J.; Kim, J.H.; Moon, W.; Park, J.; Park, S.J.; Song, G.A.; Han, S.H.; Lee, J.H. Rebamipide Attenuates 5-Fluorouracil-Induced Small Intestinal Mucositis in a Mouse Model. Biol. Pharm. Bull. 2015, 38, 179–183. [Google Scholar] [CrossRef] [Green Version]
  77. Xiang, D.-C.; Yang, J.-Y.; Xu, Y.-J.; Zhang, S.; Li, M.; Zhu, C.; Zhang, C.-L.; Liu, D. Protective effect of Andrographolide on 5-Fu induced intestinal mucositis by regulating p38 MAPK signaling pathway. Life Sci. 2020, 252, 117612. [Google Scholar] [CrossRef]
  78. Han, X.; Wu, Z.; Di, J.; Pan, Y.; Zhang, H.; Du, Y.; Cheng, Z.; Jin, Z.; Wang, Z.; Zheng, Q.; et al. CXCL9 attenuated chemotherapy-induced intestinal mucositis by inhibiting proliferation and reducing apoptosis. Biomed. Pharmacother. 2011, 65, 547–554. [Google Scholar] [CrossRef]
  79. Yeung, C.-Y.; Chan, W.-T.; Jiang, C.-B.; Cheng, M.-L.; Liu, C.-Y.; Chang, S.-W.; Chiang Chiau, J.-S.; Lee, H.-C. Amelioration of chemo-therapy-induced intestinal mucositis by orally administered probiotics in a mouse model. PLoS ONE 2015, 10, e0138746. [Google Scholar]
  80. Lu, H.; Liu, H.; Wang, J.; Shen, J.; Weng, S.; Han, L.; Sun, T.; Qian, L.; Wu, M.; Zhu, S.; et al. The chemokine CXCL9 exacerbates chemotherapy-induced acute intestinal damage through inhibition of mucosal restitution. J. Cancer Res. Clin. Oncol. 2015, 141, 983–992. [Google Scholar] [CrossRef]
  81. Torres, D.M.; Tooley, K.L.; Butler, R.N.; Smith, C.L.; Geier, M.S.; Howarth, G.S. Lyprinol™ only partially improves indicators of small intestinal integrity in a rat model of 5-fluorouracil-induced mucositis. Cancer Biol. Ther. 2008, 7, 295–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Mashtoub, S.; Tran, C.D.; Howarth, G.S. Emu oil expedites small intestinal repair following 5-fluorouracil-induced mucositis in rats. Exp. Biol. Med. 2013, 238, 1305–1317. [Google Scholar] [CrossRef] [PubMed]
  83. Lindsay, R.J.; Geier, M.S.; Yazbeck, R.; Butler, R.N.; Howarth, G.S. Orally administered emu oil decreases acute inflammation and alters selected small intestinal parameters in a rat model of mucositis. Br. J. Nutr. 2010, 104, 513–519. [Google Scholar] [CrossRef] [Green Version]
  84. Kissow, H.; Viby, N.-E.; Hartmann, B.; Holst, J.J.; Timm, M.; Thim, L.; Poulsen, S.S. Exogenous glucagon-like peptide-2 (GLP-2) pre-vents chemotherapy-induced mucositis in rat small intestine. Cancer Chemother. Pharmacol. 2012, 70, 39–48. [Google Scholar] [CrossRef] [PubMed]
  85. Prisciandaro, L.D.; Geier, M.S.; Butler, R.N.; Cummins, A.G.; Howarth, G.S. Probiotic factors partially improve parameters of 5-fluorouracil-induced intestinal mucositis in rats. Cancer Biol. Ther. 2011, 11, 671–677. [Google Scholar] [CrossRef] [PubMed]
  86. Machida, M.; Shiga, S.; Machida, T.; Ohno, M.; Iizuka, K.; Hirafuji, M. Potentiation of Glucagon-Like Peptide-2 Dynamics by Methotrexate Administration in Rat Small Intestine. Biol. Pharm. Bull. 2019, 42, 1733–1740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Al-Asmari, A.K.; Khan, A.Q.; Al-Asmari, S.A.; Al-Rawi, A.; Al-Omani, S. Alleviation of 5-fluorouracil-induced intestinal mucositis in rats by vitamin E via targeting oxidative stress and inflammatory markers. J. Complement. Integr. Med. 2016, 13, 377–385. [Google Scholar] [CrossRef] [PubMed]
  88. Logan, R.M.; Stringer, A.M.; Bowen, J.M.; Gibson, R.J.; Sonis, S.T.; Keefe, D.M.K. Is the pathobiology of chemotherapy-induced ali-mentary tract mucositis influenced by the type of mucotoxic drug administered? Cancer Chemother. Pharmacol. 2009, 63, 239–251. [Google Scholar] [CrossRef]
  89. George, R.P.; Barker, T.; Lymn, K.A.; Bigatton, D.A.; Howarth, G.S.; Whittaker, A.L. A Judgement Bias Test to Assess Affective State and Potential Therapeutics in a Rat Model of Chemotherapy-Induced Mucositis. Sci. Rep. 2018, 8, 8193. [Google Scholar] [CrossRef] [Green Version]
  90. Whittaker, A.L.; Lymn, K.A.; Wallace, G.L.; Howarth, G.S. Differential Effectiveness of Clinically-Relevant Analgesics in a Rat Model of Chemotherapy-Induced Mucositis. PLoS ONE 2016, 11, e0158851. [Google Scholar] [CrossRef] [Green Version]
  91. Saegusa, Y.; Ichikawa, T.; Iwai, T.; Goso, Y.; Okayasu, I.; Ikezawa, T.; Shikama, N.; Saigenji, K.; Ishihara, K. Changes in the mucus barrier of the rat during 5-fluorouracil-induced gastrointestinal mucositis. Scand. J. Gastroenterol. 2008, 43, 59–65. [Google Scholar] [CrossRef]
  92. Kissow, H.; Hartmann, B.; Holst, J.J.; Poulsen, S.S. Glucagon-like peptide-1 as a treatment for chemotherapy-induced mucositis. Gut 2013, 62, 1724–1733. [Google Scholar] [CrossRef] [PubMed]
  93. Tsuji, E.; Hiki, N.; Nomura, S.; Fukushima, R.; Kojima, J.-I.; Ogawa, T.; Mafune, K.-I.; Mimura, Y.; Kaminishi, M. Simultaneous onset of acute inflammatory response, sepsis-like symptoms and intestinal mucosal injury after cancer chemotherapy. Int. J. Cancer 2003, 107, 303–308. [Google Scholar] [CrossRef] [PubMed]
  94. Cheah, K.Y.; Howarth, G.S.; Bastian, S.E.P. Grape Seed Extract Dose-Responsively Decreases Disease Severity in a Rat Model of Mucositis; Concomitantly Enhancing Chemotherapeutic Effectiveness in Colon Cancer Cells. PLoS ONE 2014, 9, e85184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Yazbeck, R.; Howarth, G.S.; Borges, L.; Geier, M.S.; Smith, C.L.; Davidson, G.P.; Butler, R.N. Non-invasive detection of a palifer-min-mediated adaptive response following chemotherapy-induced damage to the distal small intestine of rats. Cancer Biol. Ther. 2011, 12, 399–406. [Google Scholar] [CrossRef] [PubMed]
  96. Bajic, J.E.; Howarth, G.S.; Mashtoub, S.; Whittaker, A.L.; Bobrovskaya, L.; Hutchinson, M.R. Neuroimmunological complications arising from chemotherapy-induced gut toxicity and opioid exposure in female dark agouti rats. J. Neurosci. Res. 2022, 100, 237–250. [Google Scholar] [CrossRef]
  97. Takechi, H.; Mawatari, K.; Harada, N.; Nakaya, Y.; Asakura, M.; Aihara, M.; Takizawa, H.; Goto, M.; Nishino, T.; Minato, T.; et al. Glutamine protects the small intestinal mucosa in anticancer drug-induced rat enteritis model. J. Med. Investig. 2014, 61, 59–64. [Google Scholar] [CrossRef] [Green Version]
  98. Renck, D.; Santos, A.A.J.; Machado, P.; Petersen, G.O.; Lopes, T.G.; Santos, D.S.; Campos, M.M.; Basso, L.A. Human uridine phosphor-ylase-1 inhibitors: A new approach to ameliorate 5-fluorouracil-induced intestinal mucositis. Investig. New Drugs 2014, 32, 1301–1307. [Google Scholar] [CrossRef]
  99. Yamaguchi, K.; Ono, K.; Hitomi, S.; Ito, M.; Nodai, T.; Goto, T.; Harano, N.; Watanabe, S.; Inoue, H.; Miyano, K.; et al. Distinct TRPV1-and TRPA1-based mechanisms underlying enhancement of oral ul-cerative mucositis-induced pain by 5-fluorouracil. Pain 2016, 157, 1004–1020. [Google Scholar] [CrossRef]
  100. Zheng, H.; Gao, J.; Man, S.; Zhang, J.; Jin, Z.; Gao, W. The protective effects of Aquilariae Lignum Resinatum extract on 5-Fuorouracil-induced intestinal mucositis in mice. Phytomedicine 2019, 54, 308–317. [Google Scholar] [CrossRef]
  101. Medeiros, A.D.C.; Azevedo, Í.M.; Lima, M.L.; Araújo Filho, I.; Moreira, M.D. Effects of simvastatin on 5-fluorouracil-induced gastrointestinal mucositis in rats. Rev. Col. Bras. Cir. 2018, 45, e1968. [Google Scholar] [CrossRef] [PubMed]
  102. Li, B.-R.; Shao, S.-Y.; Yuan, L.; Jia, R.; Sun, J.; Ji, Q.; Sui, H.; Zhou, L.-H.; Zhang, Y.; Liu, H.; et al. Effects of mild moxibustion on intestinal microbiome and NLRP3 inflammasome in rats with 5-fluorouracil-induced intestinal mucositis. J. Integr. Med. 2021, 19, 144–157. [Google Scholar] [CrossRef] [PubMed]
  103. Abalo, R.; Uranga, J.; Pérez-García, I.; De Andrés, R.; Girón, R.; Vera, G.; López-Pérez, A.; Martín-Fontelles, M. May cannabinoids prevent the development of chemotherapy-induced diarrhea and intestinal mucositis? Experimental study in the rat. Neurogastroenterol. Motil. 2017, 29, e12952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Chen, H.; Zhang, F.; Li, R.; Liu, Y.; Wang, X.; Zhang, X.; Xu, C.; Li, Y.; Guo, Y.; Yao, Q. Berberine regulates fecal metabolites to ameliorate 5-fluorouracil induced intestinal mucositis through modulating gut microbiota. Biomed. Pharmacother. 2020, 124, 109829. [Google Scholar] [CrossRef]
  105. Mashtoub, S.; Lampton, L.S.; Eden, G.L.; Cheah, K.Y.; Lymn, K.A.; Bajic, J.E.; Howarth, G.S. Emu Oil Combined with LyprinolTM Re-duces Small Intestinal Damage in a Rat Model of Chemotherapy-Induced Mucositis. Nutr. Cancer 2016, 68, 1171–1180. [Google Scholar] [CrossRef]
  106. Al-Asmari, A.K.; Al-Zahrani, A.M.; Khan, A.Q.; Al-Shahrani, H.M.; Ali Al Amri, M. Taurine ameliorates 5-flourouracil-induced intestinal mucositis, hepatorenal and reproductive organ damage in Wistar rats: A biochemical and histological study. Hum. Exp. Toxicol. 2016, 35, 10–20. [Google Scholar] [CrossRef]
  107. Whitford, E.J.; Cummins, A.G.; Butler, R.N.; Prisciandaro, L.D.; Fauser, J.K.; Yazbeck, R.; Lawrence, A.; Cheah, K.Y.; Wright, T.H.; Lymn, K.A.; et al. Effects of Streptococcus thermophilus TH-4 on intestinal mucositis induced by the chemotherapeutic agent, 5-Fluorouracil (5-FU). Cancer Biol. Ther. 2009, 8, 505–511. [Google Scholar] [CrossRef] [Green Version]
  108. M Logan, R.; MStringer, A.; MBowen, J.; JGibson, R.; TSonis, S.; MKKeefe, D. Serum levels of NF-κB and pro-inflammatory cytokines following administration of mucotoxic drugs. Cancer Biol. Ther. 2008, 7, 1139–1145. [Google Scholar] [CrossRef] [Green Version]
  109. Gerhard, D.; Sousa, F.J.d.S.S.d.; Andraus, R.A.C.; Pardo, P.E.; Nai, G.A.; Neto, H.B.; Messora, M.R.; Maia, L.P. Probiotic therapy reduces inflammation and improves intestinal morphology in rats with induced oral mucositis. Braz. Oral Res. 2017, 31, e71. [Google Scholar] [CrossRef]
  110. Manzano, M.; Bueno, P.; Rueda, R.; Ramirez-Tortosa, C.; Prieto, P.; Lopez-Pedrosa, J. Intestinal Toxicity Induced by 5-Fluorouracil in Pigs: A New Preclinical Model. Chemotherapy 2007, 53, 344–355. [Google Scholar] [CrossRef]
  111. Wong, D.V.T.; Holanda, R.B.F.; Cajado, A.G.; Bandeira, A.M.; Pereira, J.F.B.; Amorim, J.O.; Torres, C.S.; Ferreira, L.M.M.; Lopes, M.H.S.; Oliveira, R.T.G.; et al. TLR4 deficiency upregulates TLR9 expression and enhances irinotecan-related intestinal mucositis and late-onset diarrhoea. Br. J. Pharmacol. 2021, 178, 4193–4209. [Google Scholar] [CrossRef] [PubMed]
  112. Lian, Q.; Xu, J.; Yan, S.; Huang, M.; Ding, H.; Sun, X.; Bi, A.; Ding, J.; Sun, B.; Geng, M. Chemotherapy-induced intestinal inflammatory responses are mediated by exosome secretion of double-strand DNA via AIM2 inflammasome activation. Cell Res. 2017, 27, 784–800. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, S.; Yueh, M.-F.; Bigo, C.; Barbier, O.; Wang, K.; Karin, M.; Nguyen, N.; Tukey, R.H. Intestinal glucuronidation protects against chemotherapy-induced toxicity by irinotecan (CPT-11). Proc. Natl. Acad. Sci. USA 2013, 110, 19143–19148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Wessner, B.; Strasser, E.-M.; Koitz, N.; Schmuckenschlager, C.; Unger-Manhart, N.; Roth, E. Green Tea Polyphenol Administration Partly Ameliorates Chemotherapy-Induced Side Effects in the Small Intestine of Mice. J. Nutr. 2007, 137, 634–640. [Google Scholar] [CrossRef] [Green Version]
  115. de Alencar, N.M.N.; Bitencourt, F.D.S.; de Figueiredo, I.S.T.; Luz, P.B.; Lima-Júnior, R.C.P.; Aragão, K.S.; Magalhães, P.J.C.; Brito, G.A.D.C.; Ribeiro, R.A.; de Freitas, A.P.F.; et al. Side-Effects of Irinotecan (CPT-11), the Clinically Used Drug for Colon Cancer Therapy, Are Eliminated in Experimental Animals Treated with Latex Proteins from Calotropis procera (Apocynaceae). Phytother. Res. PTR 2017, 31, 312–320. [Google Scholar] [CrossRef]
  116. Arifa, R.D.N.; Brito, C.B.; de Paula, T.P.; Lima, R.L.; Menezes-Garcia, Z.; Cassini-Vieira, P.; Boas, F.A.V.; Queiroz-Junior, C.M.; da Silva, J.M.; da Silva, T.A.; et al. Eosinophil plays a crucial role in intestinal mucositis induced by antineoplastic chemotherapy. Immunology 2022, 165, 355–368. [Google Scholar] [CrossRef]
  117. Pedroso, S.H.S.P.; Vieira, A.T.; Bastos, R.W.; Oliveira, J.S.; Cartelle, C.T.; Arantes, R.M.E.; Soares, P.M.G.; Generoso, S.V.; Cardoso, V.N.; Teixeira, M.M.; et al. Evaluation of mucositis induced by irinotecan after microbial colonization in germ-free mice. Microbiology 2015, 161, 1950–1960. [Google Scholar] [CrossRef]
  118. Arifa, R.D.; Madeira, M.F.; de Paula, T.P.; Lima, R.L.; Tavares, L.D.; Menezes-Garcia, Z.; Fagundes, C.T.; Rachid, M.A.; Ryffel, B.; Zamboni, D.S.; et al. Inflammasome Activation Is Reactive Oxygen Species Dependent and Mediates Irinotecan-Induced Mucositis through IL-1β and IL-18 in Mice. Am. J. Pathol. 2014, 184, 2023–2034. [Google Scholar] [CrossRef]
  119. Lima-Júnior, R.C.P.; Freitas, H.C.; Wong, D.V.T.; Wanderley, C.W.S.; Nunes, L.G.; Leite, L.L.; Miranda, S.P.; Souza, M.H.L.P.; Brito, G.; Magalhaes, P.; et al. Targeted inhibition of IL-18 attenuates irinotecan-induced intestinal mu-cositis in mice. Br. J. Pharmacol. 2014, 171, 2335–2350. [Google Scholar] [CrossRef]
  120. Lima-Júnior, R.C.P.; Figueiredo, A.A.; Freitas, H.C.; Melo, M.L.P.; Wong, D.V.T.; Leite, C.A.V.G.; Medeiros, R.P.; Marques-Neto, R.D.; Vale, M.L.; Brito, G.A.C.; et al. Involvement of nitric oxide on the pathogenesis of irinotecan-induced intestinal mucositis: Role of cytokines on inducible nitric oxide synthase activation. Cancer Chemother. Pharmacol. 2012, 69, 931–942. [Google Scholar] [CrossRef]
  121. Secombe, K.R.; Crame, E.E.; Tam, J.S.Y.; Wardill, H.R.; Gibson, R.J.; Coller, J.K.; Bowen, J.M. Intestinal toll-like receptor 4 knockout alters the functional capacity of the gut microbiome following irinotecan treatment. Cancer Chemother. Pharmacol. 2022, 89, 275–281. [Google Scholar] [CrossRef] [PubMed]
  122. Boeing, T.; de Souza, P.; Speca, S.; Somensi, L.B.; Mariano, L.N.B.; Cury, B.J.; dos Anjos, M.F.; Quintão, N.L.M.; Dubuqoy, L.; Desreumax, P.; et al. Luteolin prevents irinotecan-induced intestinal mucositis in mice through antioxidant and anti-inflammatory properties. J. Cereb. Blood Flow Metab. 2020, 177, 2393–2408. [Google Scholar] [CrossRef] [PubMed]
  123. Boeing, T.; Gois, M.B.; de Souza, P.; Somensi, L.B.; Sant Ana, D.D.M.G.; da Silva, L.M. Irinotecan-induced intestinal mucositis in mice: A histopathological study. Cancer Chemother. Pharmacol. 2021, 87, 327–336. [Google Scholar] [CrossRef] [PubMed]
  124. Gallotti, B.; Galvao, I.; Leles, G.; Quintanilha, M.F.; Souza, R.O.; Miranda, V.C.; Rocha, V.M.; Trindade, L.M.; Jesus, L.C.L.; Mendes, V.; et al. Effects of dietary fibre intake in chemotherapy-induced mucositis in murine model. Br. J. Nutr. 2021, 126, 853–864. [Google Scholar] [CrossRef] [PubMed]
  125. Arifa, R.D.N.; de Paula, T.P.; Madeira, M.F.M.; Lima, R.L.; Garcia, Z.M.; Ÿvila, T.V.; Pinho, V.; Barcelos, L.S.; Pinheiro, M.V.B.; Ladeira, L.O.; et al. The reduction of oxidative stress by nanocomposite Fullerol decreases mu-cositis severity and reverts leukopenia induced by Irinotecan. Pharmacol. Res. 2016, 107, 102–110. [Google Scholar] [CrossRef] [PubMed]
  126. Guabiraba, R.; Besnard, A.; Menezes, G.B.; Secher, T.; Jabir, M.; Amaral, S.S.; Braun, H.; Lima-Junior, R.C.P.; A Ribeiro, R.; Cunha, F.Q.; et al. IL-33 targeting attenuates intestinal mucositis and enhances effective tumor chemotherapy in mice. Mucosal Immunol. 2014, 7, 1079–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Fernandes, C.; Wanderley, C.W.S.; Silva, C.M.S.; Muniz, H.A.; Teixeira, M.A.; Souza, N.R.P.; Cândido, A.G.F.; Falcão, R.B.; Souza, M.H.L.P.; Almeida, P.R.C.; et al. Role of regulatory T cells in irinotecan-induced intestinal mucositis. Eur. J. Pharm. Sci. 2018, 115, 158–166. [Google Scholar] [CrossRef] [PubMed]
  128. Ouyang, M.; Luo, Z.; Zhang, W.; Zhu, D.; Lu, Y.; Wu, J.; Yao, X. Protective effect of curcumin against irinotecan induced intestinal mucosal injury via attenuation of NF κB activation, oxidative stress and endoplasmic reticulum stress. Int. J. Oncol. 2019, 54, 1376–1386. [Google Scholar] [CrossRef] [PubMed]
  129. Cechinel-Zanchett, C.C.; Boeing, T.; Somensi, L.B.; Steimbach, V.M.B.; Campos, A.; Krueger, C.D.M.A.; Schultz, C.; Sant’Ana, D.D.M.G.; Cechinel-Filho, V.; da Silva, L.M.; et al. Flavonoid-rich fraction of Bauhinia forficata Link leaves prevents the intestinal toxic effects of irinotecan chemotherapy in IEC-6 cells and in mice. Phytother. Res. 2019, 33, 90–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Wardill, H.R.; Gibson, R.J.; Van Sebille, Y.Z.; Secombe, K.R.; Coller, J.K.; White, I.A.; Manavis, J.; Hutchinson, M.R.; Staikopoulos, V.; Logan, R.M.; et al. Irinotecan-Induced Gastrointestinal Dysfunction and Pain Are Mediated by Common TLR4-Dependent Mechanisms. Mol. Cancer Ther. 2016, 15, 1376–1386. [Google Scholar] [CrossRef] [Green Version]
  131. Quintanilha, M.F.; Miranda, V.C.; Souza, R.O.; Gallotti, B.; Cruz, C.; Santos, E.A.; Alvarez-Leite, J.I.; Jesus, L.C.; Azevedo, V.; Trindade, L.M.; et al. Bifidobacterium longum subsp. longum 5(1A) attenuates intestinal injury against irinotecan-induced mucositis in mice. Life Sci. 2022, 289, 120243. [Google Scholar] [CrossRef] [PubMed]
  132. Howarth, G.S.; Tooley, K.L.; Davidson, G.P.; Butler, R.N. A non-invasive method for detection of intestinal mucositis induced by different classes of chemotherapy drugs in the rat. Cancer Biol. Ther. 2006, 5, 1189–1195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Bowen, J.M.; Gibson, R.J.; Stringer, A.M.; Chan, T.W.; Prabowo, A.S.; Cummins, A.G.; Keefe, D.M. Role of p53 in irinotecan-induced intestinal cell death and mucosal damage. Anti-Cancer Drugs 2007, 18, 197–210. [Google Scholar] [CrossRef] [PubMed]
  134. Al-Dasooqi, N.; Bowen, J.M.; Gibson, R.J.; Logan, R.M.; Stringer, A.M.; Keefe, D.M. Selection of housekeeping genes for gene ex-pression studies in a rat model of irinotecan-induced mucositis. Chemotherapy 2011, 57, 43–53. [Google Scholar] [CrossRef] [PubMed]
  135. Bateman, E.; Weaver, E.; Klein, G.; Wignall, A.; Wozniak, B.; Plews, E.; Mayo, B.; White, I.; Keefe, D. Serum-derived bovine immuno-globulin/protein isolate in the alleviation of chemotherapy-induced mucositis. Support. Care Cancer Off. 2016, 24, 377–385. [Google Scholar]
  136. Al-Dasooqi, N.; Bowen, J.M.; Gibson, R.J.; Logan, R.M.; Stringer, A.M.; Keefe, D.M. Irinotecan-induced alterations in intestinal cell kinetics and extracellular matrix component expression in the dark agouti rat. Int. J. Exp. Pathol. 2011, 92, 357–365. [Google Scholar] [CrossRef]
  137. Al-Dasooqi, N.; Gibson, R.; Bowen, J.M.; Logan, R.M.; Stringer, A.; Keefe, D.M. Matrix metalloproteinases are possible mediators for the development of alimentary tract mucositis in the dark agouti rat. Exp. Biol. Med. 2010, 235, 1244–1256. [Google Scholar] [CrossRef]
  138. Gibson, R.J.; Bowen, J.M.; Keefe, D.M. Palifermin reduces diarrhea and increases survival following irinotecan treatment in tumor-bearing DA rats. Int. J. Cancer 2005, 116, 464–470. [Google Scholar] [CrossRef]
  139. Tentori, L.; Leonetti, C.; Scarsella, M.; Muzi, A.; Mazzon, E.; Vergati, M.; Forini, O.; Lapidus, R.; Xu, W.; Dorio, A.S.; et al. Inhibition of poly(ADP-ribose) polymerase prevents irinotecan-induced intestinal damage and enhances irinotecan/temozolomide efficacy against colon carcinoma. FASEB J. 2006, 20, 1709–1711. [Google Scholar] [CrossRef] [Green Version]
  140. Thorpe, D.; Butler, R.; Sultani, M.; Vanhoecke, B.; Stringer, A. Irinotecan-induced mucositis is associated with goblet cell dysreg-ulation and neural cell damage in a tumour bearing DA rat model. Pathol. Oncol. Res. 2020, 26, 955–965. [Google Scholar] [CrossRef]
  141. Thorpe, D.; Sultani, M.; Stringer, A. Irinotecan induces enterocyte cell death and changes to muc2 and muc4 composition during mucositis in a tumour-bearing DA rat model. Cancer Chemother. Pharmacol. 2019, 83, 893–904. [Google Scholar] [CrossRef] [PubMed]
  142. Fakiha, K.; Coller, J.K.; Logan, R.M.; Gibson, R.J.; Bowen, J.M. Amitriptyline prevents CPT-11-induced early-onset diarrhea and colonic apoptosis without reducing overall gastrointestinal damage in a rat model of mucositis. Support. Care Cancer 2019, 27, 2313–2320. [Google Scholar] [CrossRef] [PubMed]
  143. Yu, C.; Zhou, B.; Xia, X.; Chen, S.; Deng, Y.; Wang, Y.; Wu, L.; Tian, Y.; Zhao, B.; Xu, H.; et al. Prevotella copri is associated with carboplatin-induced gut toxicity. Cell Death Dis. 2019, 10, 714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Deng, L.; Zeng, H.; Hu, X.; Xiao, M.; He, D.; Zhang, Y.; Jin, Y.; Hu, Y.; Zhu, Y.; Gong, L.; et al. Se@Albumin nanoparticles ameliorate intestinal mucositis caused by cisplatin via gut microbi-ota-targeted regulation. Nanoscale 2021, 13, 11250–11261. [Google Scholar] [CrossRef] [PubMed]
  145. Deng, L.; Zhou, X.; Lan, Z.; Tang, K.; Zhu, X.; Mo, X.; Zhao, Z.; Zhao, Z.; Wu, A.M. Simotang Alleviates the Gastrointestinal Side Effects of Chemotherapy by Altering Gut Microbiota. J. Microbiol. Biotechnol. 2022, 32, 405–418. [Google Scholar] [CrossRef]
  146. Araújo, R.S.; Silveira, A.L.M.; Souza, L.D.S.E.; Freire, R.H.; de Souza, C.M.; Reis, D.C.; Costa, B.R.C.; Sugimoto, M.A.; Silveira, J.N.; Martins, F.D.S.; et al. Intestinal toxicity evaluation of long-circulating and pH-sensitive lipo-somes loaded with cisplatin. Eur. J. Pharm. Sci. 2017, 106, 142–151. [Google Scholar]
  147. Liao, P.-L.; Huang, S.-H.; Hung, C.-H.; Huang, W.-K.; Tsai, C.-H.; Kang, J.-J.; Wang, H.-P.; Cheng, Y.-W. Efficacy of Azatyrosine-Phenylbutyric Hydroxamides, a Histone Deacetylase Inhibitor, on Chemotherapy-Induced Gastrointestinal Mucositis. Int. J. Mol. Sci. 2019, 20, 249. [Google Scholar] [CrossRef]
  148. Jin, S.; Guan, T.; Wang, S.; Hu, M.; Liu, X.; Huang, S.; Liu, Y. Dioscin Alleviates Cisplatin-Induced Mucositis in Rats by Modulating Gut Microbiota, Enhancing Intestinal Barrier Function and Attenuating TLR4/NF-κB Signaling Cascade. Int. J. Mol. Sci. 2022, 23, 4431. [Google Scholar] [CrossRef]
  149. Bilg, A.O.; Topal, I.; Akbul, U.E.; Cimen, O.; Kolkiran, A.; Akturan, S.; Cim, F.K.; Cankaya, M.; Eden, A.O.; Suleyman, Z. Effect of Rutin on Cisplatin-induced Small Intestine (Jejunum) Damage in Rats. Int. J. Pharmacol. 2018, 14, 1136–1144. [Google Scholar] [CrossRef] [Green Version]
  150. Wu, Y.; Wu, J.; Lin, Z.; Wang, Q.; Li, Y.; Wang, A.; Shan, X.; Liu, J. Administration of a Probiotic Mixture Ameliorates Cispla-tin-Induced Mucositis and Pica by Regulating 5-HT in Rats. J. Immunol. Res. 2021, 2021, 9321196. [Google Scholar] [CrossRef]
  151. Tazuke, Y.; Maeda, K.; Wasa, M.; Satoko, N.; Fukuzawa, M. Protective mechanism of glutamine on the expression of proliferating cell nuclear antigen after cisplatin-induced intestinal mucosal injury. Pediatr. Surg. Int. 2011, 27, 151–158. [Google Scholar] [CrossRef] [PubMed]
  152. Nose, S.; Wasa, M.; Tazuke, Y.; Owari, M.; Fukuzawa, M. Cisplatin upregulates glutamine transport in human intestinal epithelial cells: The protective mechanism of glutamine on intestinal mucosa after chemotherapy. JPEN J. Parenter. Enter. Nutr. 2010, 34, 530–537. [Google Scholar] [CrossRef] [PubMed]
  153. Zenitani, M.; Sasaki, T.; Oue, T. Kampo medicines Rikkunshito and Hangeshashinto prevent cisplatin-induced intestinal mu-cosal injury in rats. J. Pediatr. Surg. 2021, 56, 1211–1218. [Google Scholar] [CrossRef] [PubMed]
  154. Donald, E.L.; Stojanovska, L.; Apostolopoulos, V.; Nurgali, K. Resveratrol alleviates oxidative damage in enteric neurons and associated gastrointestinal dysfunction caused by chemotherapeutic agent oxaliplatin. Maturitas 2017, 105, 100–106. [Google Scholar] [CrossRef] [PubMed]
  155. Xia, T.; Zhang, J.; Han, L.; Jin, Z.; Wang, J.; Li, X.; Man, S.; Liu, C.; Gao, W. Protective effect of magnolol on oxaliplatin-induced intes-tinal injury in mice. Phytother. Res. PTR 2019, 33, 1161–1172. [Google Scholar] [CrossRef] [PubMed]
  156. Gao, Y.; Sun, Q.; Yang, X.; Lu, W.; Zhao, Y.; Ge, W.; Yang, Y.; Xu, X.; Zhang, J. Orally administered salecan ameliorates methotrexate-induced intestinal mucositis in mice. Cancer Chemother. Pharmacol. 2019, 84, 105–116. [Google Scholar] [CrossRef]
  157. Tran, C.D.; Sundar, S.; Howarth, G.S. Dietary zinc supplementation and methotrexate-induced small intestinal mucositis in metallothionein-knockout and wild-type mice. Cancer Biol. Ther. 2009, 8, 1662–1667. [Google Scholar] [CrossRef]
  158. Frank, M.; Hennenberg, E.M.; Eyking, A.; Rünzi, M.; Gerken, G.; Scott, P.; Parkhill, J.; Walker, A.W.; Cario, E. TLR Signaling Modulates Side Effects of Anticancer Therapy in the Small Intestine. J. Immunol. 2015, 194, 1983–1995. [Google Scholar] [CrossRef] [Green Version]
  159. Musa, N.S.E.; Howarth, G.S.; Tran, C.D. Zinc Supplementation Alone Is Effective for Partial Amelioration of Methotrexate-induced Intestinal Damage. Altern. Ther. Health Med. 2015, 21 (Suppl. S2), 22–31. [Google Scholar]
  160. De Koning, B.A.E.; Sluis, M.v.d.; Lindenbergh-Kortleve, D.J.; Velcich, A.; Pieters, R.; Büller, H.A.; Einerhand, A.W.C.; Renes, I.B. Methotrexate-induced mucositis in mucin 2-deficient mice. J. Cell Physiol. 2007, 210, 144–152. [Google Scholar] [CrossRef]
  161. Yilmaz, E.; Azizoglu, Z.B.; Aslan, K.; Erdem, S.; Haliloglu, Y.; Suna, P.A.; Yay, A.H.; Deniz, K.; Tasdemir, A.; Per, S.; et al. Therapeutic effects of vitamin D and IL-22 on methotrexate-induced mucositis in mice. Anti-Cancer Drugs 2022, 33, 11–18. [Google Scholar] [CrossRef] [PubMed]
  162. Shi, C.-J.; Wen, X.-S.; Gao, H.-F.; Liu, Z.-H.; Xu, X.-K.; Li, L.-F.; Shen, T.; Xian, C.J. Steamed root of Rehmannia glutinosa Libosch (Plantaginaceae) alleviates methotrexate-induced intestinal mucositis in rats. J. Ethnopharmacol. 2016, 183, 143–150. [Google Scholar] [CrossRef] [PubMed]
  163. Yamamoto, A.; Itoh, T.; Nasu, R.; Kajiwara, E.; Nishida, R. Sodium alginate inhibits methotrexate-induced gastrointestinal mu-cositis in rats. Biol. Pharm. Bull. 2013, 36, 1528–1534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Shiga, S.; Machida, T.; Yanada, T.; Machida, M.; Hirafuji, M.; Iizuka, K. The role of nitric oxide in small intestine differs between a single and a consecutive administration of methotrexate to rats. J. Pharmacol. Sci. 2020, 143, 30–38. [Google Scholar] [CrossRef] [PubMed]
  165. Kesik, V.; Uysal, B.; Kurt, B.; Kismet, E.; Koseoglu, V. Ozone ameliorates methotrexate-induced intestinal injury in rats. Cancer Biol. Ther. 2009, 8, 1623–1628. [Google Scholar] [CrossRef] [Green Version]
  166. Clarke, J.M.; Pelton, N.C.; Bajka, B.H.; Howarth, G.S.; Read, L.C.; Butler, R.N. Use of the 13C-sucrose breath test to assess chemo-therapy-induced small intestinal mucositis in the rat. Cancer Biol. Ther. 2006, 5, 34–38. [Google Scholar] [CrossRef] [Green Version]
  167. Gibson, R.J.; Keefe, D.M.; Clarke, J.M.; Regester, G.O.; Thompson, F.M.; Goland, G.J.; Edwards, B.G.; Cummins, A.G. The effect of keratinocyte growth factor on tumour growth and small intestinal mucositis after chemotherapy in the rat with breast cancer. Cancer Chemother. Pharmacol. 2002, 50, 53–58. [Google Scholar] [CrossRef]
  168. Harsha, W.T.F.; Kalandarova, E.; McNutt, P.; Irwin, R.; Noel, J. Nutritional Supplementation with Transforming Growth Factor-β, Glutamine, and Short Chain Fatty Acids Minimizes Methotrexate-Induced Injury. J. Pediatr. Gastroenterol. Nutr. 2006, 42, 53–58. [Google Scholar] [CrossRef]
  169. Alamir, I.; Boukhettala, N.; Aziz, M.; Breuillé, D.; Déchelotte, P.; Coëffier, M. Beneficial effects of cathepsin inhibition to prevent chemotherapy-induced intestinal mucositis. Clin. Exp. Immunol. 2010, 162, 298–305. [Google Scholar] [CrossRef]
  170. Kolli, V.K.; Abraham, P.; Isaac, B.; Kasthuri, N. Preclinical Efficacy of Melatonin to Reduce Methotrexate-Induced Oxidative Stress and Small Intestinal Damage in Rats. Dig. Dis. Sci. 2013, 58, 959–969. [Google Scholar] [CrossRef]
  171. Chen, B.; Dragomir, M.P.; Fabris, L.; Bayraktar, R.; Knutsen, E.; Liu, X.; Tang, C.; Li, Y.; Shimura, T.; Ivkovic, T.C.; et al. The Long Noncoding RNA CCAT2 Induces Chromosomal Instability Through BOP1-AURKB Signaling. Gastroenterology 2020, 159, 2146–2162.e33. [Google Scholar] [CrossRef] [PubMed]
  172. Sukhotnik, I.; Shteinberg, D.; Ben Lulu, S.; Bashenko, Y.; Mogilner, J.G.; Ure, B.M.; Shaoul, R.; Shamian, B.; Coran, A.G. Transforming growth factor-alpha stimulates enterocyte proliferation and accelerates intestinal recovery following methotrexate-induced intestinal mucositis in a rat and a cell culture model. Pediatr. Surg. Int. 2008, 24, 1303–1311. [Google Scholar] [CrossRef] [PubMed]
  173. Van’t Land, B.; Van Beek, N.; Van Den Berg, J.J.; M’Rabet, L. Lactoferrin reduces methotrexate-induced small intestinal damage, possibly through inhibition of GLP-2-mediated epithelial cell proliferation. Dig. Dis. Sci. 2004, 49, 425–433. [Google Scholar] [CrossRef] [PubMed]
  174. Fijlstra, M.; Rings, E.H.H.M.; Verkade, H.J.; van Dijk, T.H.; Kamps, W.A.; Tissing, W.J.E. Lactose maldigestion during methotrexate-induced gastrointestinal mucositis in a rat model. Am. J. Physiol. Liver Physiol. 2011, 300, G283–G291. [Google Scholar] [CrossRef] [Green Version]
  175. Ferreira, A.R.D.S.; Wardill, H.R.; Havinga, R.; Tissing, W.J.E.; Harmsen, H.J.M. Prophylactic Treatment with Vitamins C and B2 for Methotrexate-Induced Gastrointestinal Mucositis. Biomolecules 2020, 11, 34. [Google Scholar] [CrossRef]
  176. Ozcicek, F.; Kara, A.V.; Akbas, E.M.; Kurt, N.; Yazici, G.N.; Cankaya, M.; Mammadov, R.; Ozcicek, A.; Suleyman, H. Effects of anakinra on the small intestine mucositis induced by methotrexate in rats. Exp. Anim. 2020, 69, 144–152. [Google Scholar] [CrossRef] [Green Version]
  177. Tooley, K.L.; Howarth, G.S.; Lymn, K.A.; Butler, R.N. Optimization of the non-invasive 13C-sucrose breath test in a rat model of methotrexate-induced mucositis. Cancer Chemother. Pharmacol. 2010, 65, 913–921. [Google Scholar] [CrossRef]
  178. Fijlstra, M.; Tissing, W.J.E.; Stellaard, F.; Verkade, H.J.; Rings, E.H.H.M. Reduced absorption of long-chain fatty acids during metho-trexate-induced gastrointestinal mucositis in the rat. Clin. Nutr. Edinb. Scotl. 2013, 32, 452–459. [Google Scholar] [CrossRef]
  179. Koppelmann, T.; Pollak, Y.; Ben-Shahar, Y.; Gorelik, G.; Sukhotnik, I. The Mechanisms of the Anti-Inflammatory and An-ti-Apoptotic Effects of Omega-3 Polyunsaturated Fatty Acids during Methotrexate-Induced Intestinal Damage in Cell Line and in a Rat Model. Nutrients 2021, 13, 888. [Google Scholar] [CrossRef]
  180. Tooley, K.L.; Howarth, G.S.; Lymn, K.A.; Lawrence, A.; Butler, R.N. Oral ingestion of Streptococcus thermophilus does not affect mucositis severity or tumor progression in the tumor-bearing rat. Cancer Biol. Ther. 2011, 12, 131–138. [Google Scholar] [CrossRef] [Green Version]
  181. Arslan, A.; Ozcicek, A.; Suleyman, B.; Coban, T.A.; Cimen, F.K.; Nalkiran, H.S.; Kuzucu, M.; Altuner, D.; Cetin, N.; Suleyman, H. Effects of nimesulide on the small intestine mucositis induced by methotrexate in rats. Exp. Anim. 2016, 65, 329–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Carneiro-Filho, B.A.; Lima, I.P.F.; Araujo, D.H.; Cavalcante, M.C.; Carvalho, G.H.P.; Brito, G.; Lima, V.; Monteiro, S.M.N.; Santos, F.N.; Ribeiro, R.A.; et al. Intestinal Barrier Function and Secretion in Methotrexate-Induced Rat Intestinal Mucositis. Dig. Dis. Sci. 2004, 49, 65–72. [Google Scholar] [CrossRef] [PubMed]
  183. Sukhotnik, I.; Geyer, T.; Pollak, Y.; Mogilner, J.G.; Coran, A.G.; Berkowitz, D. The Role of Wnt/β-Catenin Signaling in Enterocyte Turnover during Methotrexate-Induced Intestinal Mucositis in a Rat. PLoS ONE 2014, 9, e110675. [Google Scholar] [CrossRef]
  184. Pinto, C.; Horta, L.; Soares, A.; Carvalho, B.; Ferreira, E.; Lages, E.; Ferreira, L.; Faraco, A.; Santiago, H.; Goulart, G. Nanoencapsulated Doxorubicin Prevents Mucositis Development in Mice. Pharmaceutics 2021, 13, 1021. [Google Scholar] [CrossRef] [PubMed]
  185. Kimura, Y.; Sawai, N.; Okuda, H. Antitumour activity and adverse reactions of combined treatment with chitosan and doxo-rubicin in tumour-bearing mice. J. Pharm. Pharmacol. 2001, 53, 1373–1378. [Google Scholar] [CrossRef]
  186. Sheahan, B.J.; Theriot, C.M.; Cortes, J.E.; Dekaney, C.M. Prolonged oral antimicrobial administration prevents doxorubicin-induced loss of active intestinal stem cells. Gut Microbes 2022, 14, 2018898. [Google Scholar] [CrossRef]
  187. Nexoe, A.B.; Pedersen, A.A.; von Huth, S.; Sorensen, G.L.; Holmskov, U.; Jiang, P.-P.; Detlefsen, S.; Husby, S.; Rathe, M. No effect of deleted in malignant brain tumors 1 deficiency on chemotherapy induced murine intestinal mucositis. Sci. Rep. 2021, 11, 14687. [Google Scholar] [CrossRef] [PubMed]
  188. Anderson, C.J.; Medina, C.B.; Barron, B.J.; Karvelyte, L.; Aaes, T.L.; Lambertz, I.; Perry, J.S.A.; Mehrotra, P.; Gonçalves, A.; Lemeire, K.; et al. Microbes exploit death-induced nutrient release by gut epithelial cells. Nature 2021, 596, 262–267. [Google Scholar] [CrossRef]
  189. Rigby, R.J.; Carr, J.; Orgel, K.; King, S.L.; Lund, P.K.; Dekaney, C.M. Intestinal bacteria are necessary for doxorubicin-induced intes-tinal damage but not for doxorubicin-induced apoptosis. Gut Microbes 2016, 7, 414–423. [Google Scholar] [CrossRef] [Green Version]
  190. Andersen, M.C.E.; Johansen, M.W.; Nissen, T.; Nexoe, A.B.; Madsen, G.I.; Sorensen, G.L.; Holmskov, U.; Schlosser, A.; Moeller, J.B.; Husby, S.; et al. FIBCD1 ameliorates weight loss in chemotherapy-induced murine mucositis. Support. Care Cancer 2021, 29, 2415–2421. [Google Scholar] [CrossRef]
  191. Beukema, M.; Jermendi, É.; Koster, T.; Kitaguchi, K.; de Haan, B.J.; van den Berg, M.A.; Faas, M.M.; Schols, H.A.; de Vos, P. Attenuation of Doxorubicin-Induced Small Intestinal Mucositis by Pectins is Dependent on Pectin’s Methyl-Ester Number and Distribution. Mol. Nutr. Food Res. 2021, 65, e2100222. [Google Scholar] [CrossRef] [PubMed]
  192. Morelli, D.; Ménard, S.; Colnaghi, M.I.; Balsari, A. Oral administration of anti-doxorubicin monoclonal antibody prevents chem-otherapy-induced gastrointestinal toxicity in mice. Cancer Res. 1996, 56, 2082–2085. [Google Scholar] [PubMed]
  193. Wang, H.; Brook, C.L.; Whittaker, A.L.; Lawrence, A.; Yazbeck, R.; Howarth, G.S. Effects of Streptococcus thermophilus TH-4 in a rat model of doxorubicin-induced mucositis. Scand. J. Gastroenterol. 2013, 48, 959–968. [Google Scholar] [CrossRef] [PubMed]
  194. Shen, R.L.; Pontoppidan, P.E.L.; Rathe, M.; Jiang, P.; Hansen, C.F.; Buddington, R.K.; Heegaard, P.M.H.; Müller, K.; Sangild, P.T. Milk diets influence doxorubicin-induced intestinal toxicity in piglets. Am. J. Physiol. Liver Physiol. 2016, 311, G324–G333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Rtibi, K.; Selmi, S.; Grami, D.; Amri, M.; Sebai, H.; Marzouki, L. Contribution of oxidative stress in acute intestinal mucositis induced by 5 fluorouracil (5-FU) and its pro-drug capecitabine in rats. Toxicol. Mech. Methods 2018, 28, 262–267. [Google Scholar] [CrossRef] [PubMed]
  196. Méndez Utz, V.E.; Pérez Visñuk, D.; Perdigón, G.; de Moreno de LeBlanc, A. Milk fermented by Lactobacillus casei CRL431 administered as an immune adjuvant in models of breast cancer and metastasis under chemotherapy. Appl. Microbiol. Bio-Technol. 2021, 105, 327–340. [Google Scholar] [CrossRef]
  197. Van Sebille, Y.Z.; Gibson, R.J.; Wardill, H.R.; Carney, T.J.; Bowen, J.M. Use of zebrafish to model chemotherapy and targeted therapy gastrointestinal toxicity. Exp. Biol. Med. 2019, 244, 1178–1185. [Google Scholar] [CrossRef]
  198. Tang, S.; Ma, X.; Lu, J.; Zhang, Y.; Liu, M.; Wang, X. Preclinical toxicology and toxicokinetic evaluation of ailanthone, a natural product against castration-resistant prostate cancer, in mice. Fitoterapia 2019, 136, 104161. [Google Scholar] [CrossRef]
  199. Castellino, S.; Elion, G.B.; Griffith, O.W.; Dewhirst, M.; Kurtzberg, J.; Cattley, R.C.; Scott, P.; Bigner, D.D.; Friedman, H.S. Development of a model of melphalan-induced gastrointestinal toxicity in mice. Cancer Chemother. Pharmacol. 1993, 31, 376–380. [Google Scholar] [CrossRef]
  200. EL Pontoppidan, P.; Shen, R.L.; Cilieborg, M.S.; Jiang, P.; Kissow, H.; Petersen, B.L.; Thymann, T.; Heilmann, C.; Müller, K.; Sangild, P.T. Bovine Colostrum Modulates Myeloablative Chemotherapy–Induced Gut Toxicity in Piglets. J. Nutr. 2015, 145, 1472–1480. [Google Scholar] [CrossRef] [Green Version]
  201. Zhang, P.; Liu, J.; Xiong, B.; Zhang, C.; Kang, B.; Gao, Y.; Li, Z.; Ge, W.; Cheng, S.; Hao, Y.; et al. Microbiota from alginate oligosaccharide-dosed mice successfully mitigated small intestinal mucositis. Microbiome 2020, 8, 112. [Google Scholar] [CrossRef] [PubMed]
  202. Zuo, T.; Li, X.; Chang, Y.; Duan, G.; Yu, L.; Zheng, R.; Xue, C.; Tang, Q. Dietary fucoidan of Acaudina molpadioides and its enzy-matically degraded fragments could prevent intestinal mucositis induced by chemotherapy in mice. Food Funct. 2015, 6, 415–422. [Google Scholar] [CrossRef]
  203. Xiang, D.; Guo, Y.; Zhang, J.; Gao, J.; Lu, H.; Zhu, S.; Wu, M.; Yu, Y.; Han, W. Interleukin-1 receptor antagonist attenuates cyclo-phosphamide-induced mucositis in a murine model. Cancer Chemother. Pharmacol. 2011, 67, 1445–1453. [Google Scholar] [CrossRef] [PubMed]
  204. Zuo, T.; Cao, L.; Li, X.; Zhang, Q.; Xue, C.; Tang, Q. The Squid Ink Polysaccharides Protect Tight Junctions and Adherens Junctions from Chemotherapeutic Injury in the Small Intestinal Epithelium of Mice. Nutr. Cancer 2015, 67, 364–371. [Google Scholar] [CrossRef] [PubMed]
  205. Liu, T.; Wu, Y.; Wang, L.; Pang, X.; Zhao, L.; Yuan, H.; Zhang, C. A More Robust Gut Microbiota in Calorie-Restricted Mice Is Associated with Attenuated Intestinal Injury Caused by the Chemotherapy Drug Cyclophosphamide. mBio 2019, 10, e02903-18. [Google Scholar] [CrossRef] [Green Version]
  206. Moriya, T.; Fukatsu, K.; Noguchi, M.; Okamoto, K.; Murakoshi, S.; Saitoh, D.; Miyazaki, M.; Hase, K.; Yamamoto, J. Intravenous ad-ministration of high-dose Paclitaxel reduces gut-associated lymphoid tissue cell number and respiratory immunoglobulin A concentrations in mice. Surg. Infect. 2014, 15, 50–57. [Google Scholar] [CrossRef]
  207. Ramos, M.G.; Bambirra, E.A.; Cara, D.C.; Vieira, E.C.; Alvarez-Leite, J.I. Oral administration of short-chain fatty acids reduces the intestinal mucositis caused by treatment with Ara-C in mice fed commercial or elemental diets. Nutr. Cancer 1997, 28, 212–217. [Google Scholar] [CrossRef]
  208. Ypsilantis, P.; Tentes, I.; Assimakopoulos, S.F.; Kortsaris, A.; Scopa, C.D.; Simopoulos, C. Mesna ameliorates intestinal mucosa damage after ifosfamide administration in the rabbit at a dose-Related manner. J. Surg. Res. 2004, 121, 84–91. [Google Scholar] [CrossRef]
  209. Sasu, A.; Herman, H.; Mariasiu, T.; Rosu, M.; Balta, C.; Anghel, N.; Miutescu, E.; Cotoraci, C.; Hermenean, A. Protective effects of silymarin on epirubicin-induced mucosal barrier injury of the gastrointestinal tract. Drug Chem. Toxicol. 2015, 38, 442–451. [Google Scholar] [CrossRef]
  210. Cao, Y.-N.; Wang, Y.; Zhang, L.; Hou, Y.; Shan, J.; Li, M.; Chen, C.; Zhou, Y.; Shan, E.; Wang, J. Protective effect of endoplasmic reticulum stress inhibition on 5-fluorouracil-induced oral mucositis. Eur. J. Pharmacol. 2022, 919, 174810. [Google Scholar] [CrossRef]
  211. Sottili, M.; Mangoni, M.; Gerini, C.; Salvatore, G.; Castiglione, F.; Desideri, I.; Bonomo, P.; Meattini, I.; Greto, D.; Loi, M.; et al. Peroxisome proliferator activated receptor-gamma stimulation for prevention of 5-fluorouracil-induced oral mucositis in mice. Head Neck 2018, 40, 577–583. [Google Scholar] [CrossRef] [PubMed]
  212. Liu, Y.; Qi, X.; Wang, Y.; Li, M.; Yuan, Q.; Zhao, Z. Inflammation-targeted cannabidiol-loaded nanomicelles for enhanced oral mucositis treatment. Drug Deliv. 2022, 29, 1272–1281. [Google Scholar] [CrossRef] [PubMed]
  213. Takeuchi, I.; Kamiki, Y.; Makino, K. Tacids reduces the intestinal muherapeutic efficacy of rebamipide-loaded PLGA nanoparticles coated with chitosan in a mouse model for oral mucositis induced by cancer chemotherapy. Colloids Surf. B Biointerfaces 2018, 167, 468–473. [Google Scholar] [CrossRef] [PubMed]
  214. Chang, C.-T.; Hsiang, C.-Y.; Ho, T.-Y.; Wu, C.-Z.; Hong, H.-H.; Huang, Y.-F. Comprehensive Assessment of Host Responses to 5-Fluorouracil-Induced Oral Mucositis through Transcriptomic Analysis. PLoS ONE 2015, 10, e0135102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Sun, H.; Zhou, Y.; Ma, R.; Zhang, J.; Shan, J.; Chen, Y.; Li, X.; Shan, E. Metformin protects 5-Fu-induced chemotherapy oral mucositis by reducing endoplasmic reticulum stress in mice. Eur. J. Pharm. Sci. 2022, 173, 106182. [Google Scholar] [CrossRef] [PubMed]
  216. Gupta, N.; Quah, S.; Yeo, J.; Ferreira, J.; Tan, K.; Hong, C. Role of oral flora in chemotherapy-induced oral mucositis In Vivo. Arch. Oral Biol. 2019, 101, 51–56. [Google Scholar] [CrossRef]
  217. Cuba, L.D.F.; Salum, F.G.; Guimarães, F.S.; Cherubini, K.; Borghetti, R.L.; de Figueiredo, M.A.Z. Cannabidiol on 5-FU-induced oral mucositis in mice. Oral Dis. 2020, 26, 1483–1493. [Google Scholar] [CrossRef]
  218. Gupta, N.; Ferreira, J.; Hong, C.H.L.; Tan, K.S. Lactobacillus reuteri DSM 17938 and ATCC PTA 5289 ameliorates chemothera-py-induced oral mucositis. Sci. Rep. 2020, 10, 16189. [Google Scholar] [CrossRef]
  219. Katagiri, H.; Fukui, K.; Nakamura, K.; Tanaka, A. Systemic hematogenous dissemination of mouse oral candidiasis is induced by oral mucositis. Odontology 2018, 106, 389–397. [Google Scholar] [CrossRef] [Green Version]
  220. Shimamura, Y.; Takeuchi, I.; Terada, H.; Makino, K. Therapeutic Effect of GGsTop, Selective Gamma-glutamyl Transpeptidase Inhibitor, on a Mouse Model of 5-Fluorouracil-induced Oral Mucositis. Anticancer Res. 2019, 39, 201–206. [Google Scholar] [CrossRef]
  221. Tancharoen, S.; Shakya, P.; Narkpinit, S.; Dararat, P.; Kikuchi, K. Anthocyanins Extracted from Oryza sativa L. Prevent Fluorouracil-Induced Nuclear Factor-κB Activation in Oral Mucositis: In Vitro and In Vivo Studies. Int. J. Mol. Sci. 2018, 19, 2981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Thieme, S.; Ribeiro, J.T.; dos Santos, B.G.; Zieger, R.D.A.; Severo, M.L.B.; Martins, M.A.T.; Matté, C.; Martins, M.D. Comparison of photobiomodulation using either an intraoral or an extraoral laser on oral mucositis induced by chemotherapy in rats. Support. Care Cancer 2020, 28, 867–876. [Google Scholar] [CrossRef] [PubMed]
  223. Lima, G.D.M.G.; Severo, M.C.; Santana-Melo, G.D.F.; Carvalho, M.A.; Vilela-Goulart, M.D.G.; Salgado, M.A.C.; Gomes, M.F. Amniotic membrane as a biological dressing for 5-fluoruracil-induced oral mucositis in rats. Int. J. Oral. Maxillofac. Surg. 2015, 44, 845–851. [Google Scholar] [CrossRef] [PubMed]
  224. Miyano, K.; Eto, M.; Hitomi, S.; Matsumoto, T.; Hasegawa, S.; Hirano, A.; Nagabuchi, K.; Asai, N.; Uzu, M.; Nonaka, M.; et al. The Japanese herbal medicine Hangeshashinto enhances oral keratinocyte migration to facilitate healing of chemotherapy-induced oral ulcerative mucositis. Sci. Rep. 2020, 10, 625. [Google Scholar] [CrossRef] [Green Version]
  225. Tanideh, N.; Davarmanesh, M.; Andisheh-Tadbir, A.; Ranjbar, Z.; Mehriar, P.; Koohi-Hosseinabadi, O. Healing acceleration of oral mucositis induced by 5-fluorouracil with Pistacia atlantica (bene) essential oil in hamsters. J. Oral Pathol. Med. 2017, 46, 725–730. [Google Scholar] [CrossRef]
  226. Leitão, R.F.C.; Ribeiro, R.A.; Lira, A.M.S.; Silva, L.R.; Bellaguarda, E.A.L.; Macedo, F.D.B.; Sousa, R.B.; Brito, G.A.C. Glutamine and ala-nyl-glutamine accelerate the recovery from 5-fluorouracil-induced experimental oral mucositis in hamster. Cancer Chemother. Pharmacol. 2008, 61, 215–222. [Google Scholar] [CrossRef]
  227. Fonseca, K.M.; RodriguesCosta, D.M.; da Silva, V.F.; de Carvalho, J.L.; Oliveira, A.P.; Sousa, F.B.D.M.; Lopes, A.L.F.; Martins, C.D.S.; Chaves, L.D.S.; Nicolau, L.A.D.; et al. Anti-inflammatory effect of l-cysteine (a semi-essential amino acid) on 5-FU-induced oral mucositis in hamsters. Amino Acids 2021, 53, 1415–1430. [Google Scholar] [CrossRef]
  228. Lima, V.; Brito, G.; Cunha, F.D.Q.; Rebouças, C.; Falcão, B.; Augusto, R.; Souza, M.; Leitão, B.; Ribeiro, R. Effects of the tumour necrosis factor-α inhibitors pentoxifylline and thalidomide in short-term experimental oral mucositis in hamsters. Eur. J. Oral Sci. 2005, 113, 210–217. [Google Scholar] [CrossRef]
  229. Medeiros, C.A.C.X.; Leitão, R.F.C.; Macedo, R.N.; Barboza, D.R.M.M.; Gomes, A.S.; Nogueira, N.A.P.; Alencar, N.M.N.; Ribeiro, R.A.; Brito, G.A.C. Effect of atorvastatin on 5-fluorouracil-induced experimental oral mucositis. Cancer Chemother. Pharmacol. 2011, 67, 1085–1100. [Google Scholar] [CrossRef]
  230. Clarke, J.; Butler, R.; Howarth, G.; Read, L.; Regester, G. Exposure of oral mucosa to bioactive milk factors reduces severity of chemotherapy-induced mucositis in the hamster. Oral Oncol. 2002, 38, 478–485. [Google Scholar] [CrossRef]
  231. Yoshino, F.; Yoshida, A.; Nakajima, A.; Wada-Takahashi, S.; Takahashi, S.-S.; Lee, M.C.-I. Alteration of the Redox State with Reactive Oxygen Species for 5-Fluorouracil-Induced Oral Mucositis in Hamsters. PLoS ONE 2013, 8, e82834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Sonis, S.; Muska, A.; O’Brien, J.; VanVugt, A.; Langer-Safer, P.; Keith, J. Alteration in the frequency, severity and duration of chemotherapy-induced mucositis in hamsters by interleukin-11. Oral Oncol. 1995, 31, 261–266. [Google Scholar] [CrossRef] [PubMed]
  233. Freitas, A.P.F.; Bitencourt, F.S.; Brito, G.A.C.; de Alencar, N.M.N.; Ribeiro, R.A.; Lima-Júnior, R.C.P.; Ramos, M.V.; Vale, M.L. Protein fraction of Calotropis procera latex protects against 5-fluorouracil-induced oral mucositis associated with downregulation of pivotal pro-inflammatory mediators. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2012, 385, 981–990. [Google Scholar] [CrossRef] [PubMed]
  234. Horii, K.; Kanayama, T.; Miyamoto, H.; Kohgo, T.; Tsuchimochi, T.; Shigetomi, T.; Yokoi, M. Platelet-rich fibrin has a healing effect on chemotherapy-induced mucositis in hamsters. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2014, 117, 445–453. [Google Scholar] [CrossRef]
  235. Park, J.W.; Oh, J.; Ko, S.J.; Chang, M.S.; Kim, J. Effects of Onchung-eum, an Herbal Prescription, on 5-Fluorouracil–Induced Oral Mucositis. Integr. Cancer Ther. 2018, 17, 1285–1296. [Google Scholar] [CrossRef] [Green Version]
  236. Wilder-Smith, P.; Hammer-Wilson, M.J.; Zhang, J.; Wang, Q.; Osann, K.; Chen, Z.; Wigdor, H.; Schwartz, J.; Epstein, J. In Vivo Imaging of Oral Mucositis in an Animal Model Using Optical Coherence Tomography and Optical Doppler Tomography. Clin. Cancer Res. 2007, 13, 2449–2454. [Google Scholar] [CrossRef] [Green Version]
  237. Schmidt, T.R.; Curra, M.; Wagner, V.P.; Martins, M.A.T.; de Oliveira, A.C.; Batista, A.C.; Valadares, M.C.; Marreto, R.N.; Martins, M.D. Mucoadhesive formulation containing Curcuma longa L. reduces oral mucositis induced by 5-fluorouracil in hamsters. Phytother. Res. PTR 2019, 33, 881–890. [Google Scholar] [CrossRef] [Green Version]
  238. Da Cruz, É.D.P.; Campos, L.; da Silva Pereira, F.; Magliano, G.C.; Benites, B.M.; Arana-Chavez, V.E.; Ballester, R.Y.; Simões, A. Clinical, biochemical and histological study of the effect of antimicrobial photodynamic therapy on oral mucositis induced by 5-fluorouracil in hamsters. Photodiagn. Photodyn. Ther. 2015, 12, 298–309. [Google Scholar] [CrossRef]
  239. Koohi-Hosseinabadi, O.; Ranjbar, Z.; Sepehrimanesh, M.; AndisheTadbir, A.; Poorbaghi, S.L.; Bahranifard, H.; Tanideh, N.; Koohi-Hosseinabadi, M.; Iraji, A. Biochemical, hematological, and pathological related healing effects of Elaeagnus angustifolia hydroalcoholic extract in 5-fluorouracil-induced oral mucositis in male golden hamster. Environ. Sci. Pollut. Res. 2017, 24, 24447–24453. [Google Scholar] [CrossRef] [PubMed]
  240. Morvan, F.O.; Baroukh, B.; Ledoux, D.; Caruelle, J.-P.; Barritault, D.; Godeau, G.; Saffar, J.-L. An Engineered Biopolymer Prevents Mucositis Induced by 5-Fluorouracil in Hamsters. Am. J. Pathol. 2004, 164, 739–746. [Google Scholar] [CrossRef] [Green Version]
  241. Davarmanesh, M.; Miri, R.; Haghnegahdar, S.; Tadbir, A.A.; Tanideh, N.; Saghiri, M.A.; Garcia-Godoy, F.; Asatourian, A. Protective effect of bilberry extract as a pretreatment on induced oral mucositis in hamsters. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2013, 116, 702–708. [Google Scholar] [CrossRef] [PubMed]
  242. Sacono, N.T.; Costa, C.A.; Bagnato, V.S.; Abreu-E-Lima, F.C. Light-emitting diode therapy in chemotherapy-induced mucositis. Lasers Surg. Med. 2008, 40, 625–633. [Google Scholar] [CrossRef] [PubMed]
  243. Clarke, J.; Edwards, B.; Srpek, L.; Regester, G. Evaluation of bovine lactoferrin as a topical therapy for chemotherapy-induced mucositis in the golden Syrian hamster. Oral Oncol. 1999, 35, 197–202. [Google Scholar] [CrossRef] [PubMed]
  244. Sonis, S.T.; Costa, J.W.; Evitts, S.M.; Lindquist, L.E.; Nicolson, M. Effect of epidermal growth factor on ulcerative mucositis in hamsters that receive cancer chemotherapy. Oral Surg. Oral Med. Oral Pathol. 1992, 74, 749–755. [Google Scholar] [CrossRef] [PubMed]
  245. Cotomacio, C.C.; Calarga, C.C.; Yshikawa, B.K.; Arana-Chavez, V.E.; Simões, A. Wound healing process with different photobio-modulation therapy protocols to treat 5-FU-induced oral mucositis in hamsters. Arch. Oral Biol. 2021, 131, 105250. [Google Scholar] [CrossRef] [PubMed]
  246. Mitsuhashi, H.; Suemaru, K.; Li, B.; Cui, R.; Araki, H. Evaluation of topical external medicine for 5-fluorouracil-induced oral mucositis in hamsters. Eur. J. Pharmacol. 2006, 551, 152–155. [Google Scholar] [CrossRef]
  247. Sonis, S.T.; Lindquist, L.; Van Vugt, A.; A Stewart, A.; Stam, K.; Qu, G.Y.; Iwata, K.K.; Haley, J.D. Prevention of chemotherapy-induced ulcerative mucositis by transforming growth factor beta 3. Cancer Res. 1994, 54, 1135–1138. [Google Scholar]
  248. Sonis, S.T.; Van Vugt, A.G.; Brien, J.P.; Muska, A.D.; Bruskin, A.M.; Rose, A.; Haley, J.D. Transforming growth factor-beta 3 mediated modulation of cell cycling and attenuation of 5-fluorouracil induced oral mucositis. Oral Oncol. 1997, 33, 47–54. [Google Scholar] [CrossRef]
  249. Cotomacio, C.C.; Campos, L.; De Souza, D.N.; Arana-Chavez, V.E.; Simões, A. Dosimetric study of photobiomodulation therapy in 5-FU-induced oral mucositis in hamsters. J. Biomed. Opt. 2017, 22, 18003. [Google Scholar] [CrossRef]
  250. Sonis, S.T.; Tracey, C.; Shklar, G.; Jenson, J.; Florine, D. An animal model for mucositis induced by cancer chemotherapy. Oral Surg. Oral Med. Oral Pathol. 1990, 69, 437–443. [Google Scholar] [CrossRef]
  251. Skeff, M.A.; Brito, G.A.C.; de Oliveira, M.G.; Braga, C.M.; Cavalcante, M.M.; Baldim, V.; Holanda-Afonso, R.C.; Silva-Boghossian, C.M.; Colombo, A.P.; Ribeiro, R.A.; et al. S-Nitrosoglutathione Accelerates Recovery from 5-Fluorouracil-Induced Oral Mucositis. PLoS ONE 2014, 9, e113378. [Google Scholar] [CrossRef] [Green Version]
  252. Freire, M.D.R.S.; Freitas, R.; Colombo, F.; Valença, A.; Marques, A.M.C.; Sarmento, V.A. LED and laser photobiomodulation in the prevention and treatment of oral mucositis: Experimental study in hamsters. Clin. Oral. Investig. 2014, 18, 1005–1013. [Google Scholar] [CrossRef]
  253. Leitão, R.F.C.; Ribeiro, R.A.; Bellaguarda, E.A.L.; Macedo, F.D.B.; Silva, L.R.; Oriá, R.; Vale, M.; Cunha, F.Q.; Brito, G.A.C. Role of nitric oxide on pathogenesis of 5-fluorouracil induced experimental oral mucositis in hamster. Cancer Chemother. Pharmacol. 2007, 59, 603–612. [Google Scholar] [CrossRef]
  254. Al-Azri, A.R.; Gibson, R.J.; Bowen, J.M.; Stringer, A.M.; Keefe, D.M.; Logan, R.M. Involvement of matrix metalloproteinases (MMP-3 and MMP-9) in the pathogenesis of irinotecan-induced oral mucositis. J. Oral Pathol. Med. 2015, 44, 459–467. [Google Scholar] [CrossRef]
  255. Bayar Muluk, N.; Kaymaz, F.F.; Cakar, A.N. Effects of topotecan treatment on nasal, buccal, and lingual mucosa in the rabbit: Light and transmission electron microscopic evaluation. Eur. Arch. Oto-Rhino-Laryngol. 2007, 264, 197–203. [Google Scholar] [CrossRef]
  256. A LaFerriere, C.; Pang, D.S. Review of Intraperitoneal Injection of Sodium Pentobarbital as a Method of Euthanasia in Laboratory Rodents. J. Am. Assoc. Lab. Anim. Sci. 2020, 59, 254–263. [Google Scholar] [CrossRef]
  257. Bertolini, M.; Sobue, T.; Thompson, A.; Dongari-Bagtzoglou, A. Chemotherapy Induces Oral Mucositis in Mice Without Additional Noxious Stimuli. Transl. Oncol. 2017, 10, 612–620. [Google Scholar] [CrossRef]
  258. Bowen, J.M.; Gibson, R.J.; Keefe, D.M. Animal Models of Mucositis: Implications for Therapy. J. Support. Oncol. 2011, 9, 161–168. [Google Scholar] [CrossRef]
  259. Sonis, S.T.; Elting, L.S.; Keefe, D.; Peterson, D.E.; Schubert, M.; Hauer-Jensen, M.; Bekele, B.N.; Raber-Durlacher, J.; Donnelly, J.P.; Ru-benstein, E.B. Perspectives on cancer therapy-induced mucosal injury: Pathogenesis, measurement, epidemiology, and con-sequences for patients. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2004, 100, 1995–2025. [Google Scholar]
  260. Lee, C.S.; Ryan, E.J.; Doherty, G.A. Gastro-intestinal toxicity of chemotherapeutics in colorectal cancer: The role of inflammation. World J. Gastroenterol. 2014, 20, 3751. [Google Scholar] [CrossRef]
  261. Nguyen, H.; Sangha, S.; Pan, M.; Shin, D.H.; Park, H.; Mohammed, A.I.; Cirillo, N. Oxidative Stress and Chemoradiation-Induced Oral Mucositis: A Scoping Review of In Vitro, In Vivo and Clinical Studies. Int. J. Mol. Sci. 2022, 23, 4863. [Google Scholar] [CrossRef]
Figure 1. Flowchart of the data collection and selection process in accordance with PRISMA-ScR guidelines.
Figure 1. Flowchart of the data collection and selection process in accordance with PRISMA-ScR guidelines.
Ijms 23 15434 g001
Table 1. Summary of data collected from in vivo animal studies involving 5-fluorouracil-induced intestinal mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models.
Table 1. Summary of data collected from in vivo animal studies involving 5-fluorouracil-induced intestinal mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models.
Animal ModelsNumber of ArticlesDosage of DrugReferences
Mice72100 ng/kg i.p. a; 25–450 mg/kg i.p.; 50–200 mg/kg i.v. injection/infusion; 50 mg/kg orally.[7,8,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80]
Rats3040–400 mg/kg i.p.; 20–50 mg/kg orally;[81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109]
Pigs112 mg/kg orally[110]
Total103
i.p.: intraperitoneal injection as a route of chemotherapy administration, i.v.: intravenous injection. a A dose of 100 ng/kg was found in one paper reporting a combination of 5-FU with doxorubicin.
Table 2. Summary of data collected from in vivo animal studies involving irinotecan (CPT-11)-induced intestinal mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models.
Table 2. Summary of data collected from in vivo animal studies involving irinotecan (CPT-11)-induced intestinal mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models.
Animal ModelsNumber of ArticlesDosage of DrugReferences
Mice2210–270 mg/kg i.p.[46,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131]
Rats1220–200 mg/kg i.p.[108,132,133,134,135,136,137,138,139,140,141,142]
Total34
i.p.: intraperitoneal injection as a route of chemotherapy administration, i.v.: intravenous injection.
Table 3. Summary of data collected from in vivo animal studies involving platinum-based chemotherapy drugs-induced intestinal mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models.
Table 3. Summary of data collected from in vivo animal studies involving platinum-based chemotherapy drugs-induced intestinal mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models.
Animal ModelsNumber of ArticlesDosage of DrugReferences
CarboplatinMice1100 mg/kg i.p.[143]
CisplatinMice52–11 mg/kg i.p. [75,144,145,146,147]
Rats65–7 mg/kg i.p.[148,149,150,151,152,153]
OxaliplatinMice41–5 mg/kg i.p.[7,8,154,155]
Total16
i.p.: intraperitoneal injection as a route of chemotherapy administration, i.v.: intravenous injection.
Table 4. Summary of data collected from in vivo animal studies involving methotrexate-induced intestinal mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models.
Table 4. Summary of data collected from in vivo animal studies involving methotrexate-induced intestinal mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models.
Animal ModelsNumber of ArticlesDosage of DrugReferences
Mice620–500 mg/kg i.p.; 12.5 mg/kg s.c. [156,157,158,159,160,161]
Rats272.5–90 mg/kg i.p.; 1.5–3.5 mg/kg s.c.; 1.5 mg/kg i.m.; 20–150 mg/kg i.v.; 5 mg/kg orally[6,86,88,108,157,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183]
Total33
i.p.: intraperitoneal injection as a route of administration, i.v.: intravenous injection, s.c.: subcutaneous injection, i.m.: intramuscular injection.
Table 5. Summary of data collected from in vivo animal studies involving 5-fluorouracil-induced oral mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models, including the route of administration.
Table 5. Summary of data collected from in vivo animal studies involving 5-fluorouracil-induced oral mucositis. Drug concentrations obtained from studies are listed in a range of minimum to maximum dosage administered in animal models, including the route of administration.
Animal ModelsNumber of ArticlesDosage of DrugReferences
Mice1310–100 mg/kg i.p.; 50 mg/kg intravenously (i.v.)[44,70,210,211,212,213,214,215,216,217,218,219,220]
Rats840–150 mg/kg i.p.[88,99,108,109,221,222,223,224]
Hamsters2940–100 mg/kg i.p.[225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253]
Total50
i.p.: intraperitoneal injection as a route of chemotherapy administration.
Table 6. Additional stimuli used in oral mucositis animal models other than the chemotherapeutic agents.
Table 6. Additional stimuli used in oral mucositis animal models other than the chemotherapeutic agents.
Additional StimuliNumber of ArticlesReferences
Mechanical irritation33[70,109,217,221,222,225,226,227,228,229,230,232,233,234,235,236,237,238,239,240,241,242,243,244,245,246,247,248,249,250,251,252,253]
Chemical injury8[99,212,213,219,220,223,224,231]
No additional stimuli12[44,88,108,200,210,211,214,215,216,218,254,255]
Total53
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Huang, J.; Hwang, A.Y.M.; Jia, Y.; Kim, B.; Iskandar, M.; Mohammed, A.I.; Cirillo, N. Experimental Chemotherapy-Induced Mucositis: A Scoping Review Guiding the Design of Suitable Preclinical Models. Int. J. Mol. Sci. 2022, 23, 15434. https://doi.org/10.3390/ijms232315434

AMA Style

Huang J, Hwang AYM, Jia Y, Kim B, Iskandar M, Mohammed AI, Cirillo N. Experimental Chemotherapy-Induced Mucositis: A Scoping Review Guiding the Design of Suitable Preclinical Models. International Journal of Molecular Sciences. 2022; 23(23):15434. https://doi.org/10.3390/ijms232315434

Chicago/Turabian Style

Huang, Junhua, Alan Yaw Min Hwang, Yuting Jia, Brian Kim, Melania Iskandar, Ali Ibrahim Mohammed, and Nicola Cirillo. 2022. "Experimental Chemotherapy-Induced Mucositis: A Scoping Review Guiding the Design of Suitable Preclinical Models" International Journal of Molecular Sciences 23, no. 23: 15434. https://doi.org/10.3390/ijms232315434

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

Article Metrics

Back to TopTop