A Phase II Randomized Clinical Trial and Mechanistic Studies Using Improved Probiotics to Prevent Oral Mucositis Induced by Concurrent Radiotherapy and Chemotherapy in Nasopharyngeal Carcinoma

Chaofei Xia, Chunling Jiang, Wenyu Li, Jing Wei, Hu Hong, Jingao Li, Liu Feng, Hong Wei, Hongbo Xin, Tingtao Chen, Chaofei Xia, Chunling Jiang, Wenyu Li, Jing Wei, Hu Hong, Jingao Li, Liu Feng, Hong Wei, Hongbo Xin, Tingtao Chen

Abstract

Earlier evidence has proven that probiotic supplements can reduce concurrent chemoradiotherapy (CCRT)-induced oral mucositis (OM) in nasopharyngeal cancer (NPC). The incidence of severe OM (grade 3 or higher) was the primary endpoint in this study. We first enrolled 85 patients with locally advanced NPC who were undergoing CCRT. Of them, 77 patients were finally selected and randomized (1:1) to receive either a probiotic cocktail or placebo. To investigate the protective effects and the mechanism of probiotic cocktail treatment on OM induced by radiotherapy and chemotherapy, we randomly divided the rats into the control (C) group, the model (M) group, and the probiotic (P) group. After treatment, samples from the tongue, blood, and fecal and proximal colon tissues on various days (7th, 14th, and 21st days) were collected and tested for the inflammatory response, cell apoptosis, intestinal permeability, and intestinal microbial changes. We found that patients taking the probiotic cocktail showed significantly lower OM. The values of the incidence of 0, 1, 2, 3, and 4 grades of OM in the placebo group and in the probiotic cocktail group were reported to be 0, 14.7, 38.2, 32.4, and 14.7% and 13.9, 36.1, 25, 22.2, and 2.8%, respectively. Furthermore, patients in the probiotic cocktail group showed a decrease in the reduction rate of CD3+ T cells (75.5% vs. 81%, p < 0.01), CD4+ T cells (64.53% vs. 79.53%, p < 0.01), and CD8+ T cells (75.59 vs. 62.36%, p < 0.01) compared to the placebo group. In the rat model, the probiotic cocktail could ameliorate the severity of OM, decrease the inflammatory response, cause cell apoptosis and intestinal permeability, and restore the structure of gut microbiota to normalcy. In conclusion, the modified probiotic cocktail significantly reduces the severity of OM by enhancing the immune response of patients with NPC and modifying the structure of gut microbiota. Clinical Trial Registration: The Clinical Trial Registration should be the NCT03112837.

Keywords: intestinal microbiota; nasopharyngeal cancer; oral mucositis; probiotics; radiotherapy and chemotherapy.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Xia, Jiang, Li, Wei, Hong, Li, Feng, Wei, Xin and Chen.

Figures

Figure 1
Figure 1
Selection of probiotic strains. (A) The non-metric multidimensional scaling (NMDS) analysis of healthy people (HP) from employees of the Jiangxi Cancer Hospital, tumor patients (TP) from the Jiangxi Cancer Hospital, and non-cancer people (NT) (healthy residents at the cancer-free village). (B) The Venn diagram of the intestinal microbiota among HP, TP, and NT. (C) The relative abundance of the bacteria among groups HP, TP, and NT. (D) The relative abundance of bacteria among the HP, TP, and NT groups with quantitative real-time PCR (qRT-PCR). The data are presented as means ± SD, where ***p < 0.001 and **** p < 0.0001.
Figure 2
Figure 2
Patients included in the study.
Figure 3
Figure 3
The combination of probiotics reduced oral mucositis (OM) by improving the immunity of patients with nasopharyngeal cancer (NPC). (A) Mucosal index. (B) Reduction rate of CD3+T cells. (C) Reduction rate of CD4+T cells. (D) Reduction rate of CD8+T cells. (E) Reduction rate of lymphocyte. (F) Reduction rate of hemoglobin. (G) Weight loss. The data are presented as means ± SD, where **p < 0.01.
Figure 4
Figure 4
Effects of the combination of probiotics on the composition of bacterial communities in patients with NPC. (A) The relative abundance of bacteria among HP from the employees of the Jiangxi Cancer Hospital, before the treatment of radiotherapy plus chemotherapy plus a placebo (BRCP), after treatment with radiotherapy plus chemotherapy plus a placebo (ARCP), before the treatment of radiotherapy plus chemotherapy plus the probiotic combination (BRCPM), and after treatment with radiotherapy plus chemotherapy plus the probiotic combination (ARCPM). (B) The Venn diagram of the intestinal microbiota among HP, BRCP, ARCP, BRCPM, and ARCPM groups. (C) The NMDS analysis of groups HP, BRCP, ARCP, BRCPM, and ARCPM.
Figure 5
Figure 5
The probiotic cocktail relieved tongue tissue inflammatory response in rats with OM caused by radiotherapy and chemotherapy. (A) Mucosal index. (B) The expression of IL-6 in the mRNA level. (C) The expression level of TNF-α in the mRNA level. (D) The expression of IL-1β in the mRNA level. (E) H&E staining of the tongue tissue among the C, M, and T groups on the 7th, 14th, and 21st days. The data are presented as means ± SD, where *p < 0.05, **p < 0.01, and ##p < 0.01.
Figure 6
Figure 6
The combination of probiotics ameliorated the upregulation of TLR4/NF-κB, tongue tissue apoptosis and improved the expression of intestinal tight junction (TJ) in rats with OM caused by radiotherapy and chemotherapy. (A) Protein expression level of TLR4, P-NF-κB, and NF-κB. (B) Protein expression level of apoptosis-associated factors, Bcl-2 and Bax. (C) The expression level of intestinal TJ proteins, ZO-1 and claudin-1. The data are presented as means ± SD, where *p < 0.05, **p < 0.01, #p < 0.05, and ##P < 0.01.
Figure 7
Figure 7
Effects of probiotic cocktail on the composition of bacterial communities in rats with OM caused by radiotherapy and chemotherapy. (A) Shannon index of intestinal bacterial communities among C, M, and T groups at days 7, 14, and 21. (B) Simpson index of intestinal bacterial communities among C, M, and T groups on the 7th, 14th, and 21st days. (C) Principle coordinate analysis (PCoA) of intestinal bacterial communities among C, M, and T groups on the 7th, 14th, and 21st days. (D) Relative abundance of Bacteroidetes of intestinal bacterial communities at the phylum level among C, M, and T groups on the 7th, 14th, and 21st days. (E) Relative abundance of Firmicutes of intestinal bacterial communities at the phylum level among C, M, and T groups on the 7th, 14th, and 21st days. (F) Relative abundance of Bacteroidetes of intestinal bacterial communities at the genus level among C, M, and T groups on the 7th, 14th, and 21st days. (G) Relative abundance of Lachnospiraceae of intestinal bacterial communities at the genus level among C, M, and T groups on the 7th, 14th, and 21st days. (H) Relative abundance of Ruminococcus of intestinal bacterial communities at the genus level among the C, M, and T groups on the 7th, 14th, and 21st days.

References

    1. Chua MLK, Wee JTS, Hui EP, Chan ATC. Nasopharyngeal carcinoma. Lancet. (2016) 387:1012–24. 10.1016/S0140-6736(15)00055-0
    1. Nishimura N, Nakano K, Ueda K, Kodaira M, Yamada S, Mishima Y, et al. . Prospective evaluation of incidence and severity of oral mucositis induced by conventional chemotherapy in solid tumors and malignant lymphomas. Support Care Cancer. (2012) 20:2053–9. 10.1007/s00520-011-1314-6
    1. Villa S, Sonis T. Mucositis: pathobiology and management. Curr Opin Oncol. (2015) 27:159–64. 10.1097/CCO.0000000000000180
    1. Daugelaite G, Uzkuraityte K, Jagelaviciene E, Filipauskas A. Prevention and treatment of chemotherapy and radiotherapy induced oral mucositis. Medicina. (2019) 55:25. 10.3390/medicina55020025
    1. Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature. (2016) 535:75–84. 10.1038/nature18848
    1. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. (2016) 16:341–52. 10.1038/nri.2016.42
    1. Cui M, Xiao H, Li Y, Zhou L, Zhao S, Luo D, et al. . Faecal microbiota transplantation protects against radiation-induced toxicity. EMBO Mol Med. (2017) 9:448–61. 10.15252/emmm.201606932
    1. Roy S, Trinchieri G. Microbiota: a key orchestrator of cancer therapy. Nat Rev Cancer. (2017) 17:271–85. 10.1038/nrc.2017.13
    1. Lakritz JR, Poutahidis T, Levkovich T, Varian BJ, Ibrahim YM, Chatzigiagkos A, et al. . Beneficial bacteria stimulate host immune cells to counteract dietary and genetic predisposition to mammary cancer in mice. Int J Cancer. (2014) 135:529–40. 10.1002/ijc.28702
    1. Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, et al. . Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. (2015) 350:1079–84. 10.1126/science.aad1329
    1. Daillere R, Vetizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, et al. . Enterococcus hirae and barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. (2016) 45:931–43. 10.1016/j.immuni.2016.09.009
    1. Sivan, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. . Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. (2015) 350:1084–9. 10.1126/science.aac4255
    1. Jiang C, Wang H, Xia C, Dong Q, Chen E, Qiu Y, et al. . A randomized, double-blind, placebo-controlled trial of probiotics to reduce the severity of oral mucositis induced by chemoradiotherapy for patients with nasopharyngeal carcinoma. Cancer. (2019) 125:1081–90. 10.1002/cncr.31907
    1. Gao W, Howden BP, Stinear TP. Evolution of virulence in Enterococcus faecium, a hospital-adapted opportunistic pathogen. Curr Opin Microbiol. (2018) 41:76–82. 10.1016/j.mib.2017.11.030
    1. Liu Y, Gibson GR, Walton GE. An in vitro approach to study effects of prebiotics and probiotics on the faecal microbiota and selected immune parameters relevant to the elderly. PLoS ONE. (2016) 11:e0162604. 10.1371/journal.pone.0162604
    1. Munoz-Quezada S, Chenoll E, Vieites JM, Genoves S, Maldonado J, Bermudez-Brito M, et al. . Isolation, identification and characterisation of three novel probiotic strains (Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035 and Lactobacillus rhamnosus CNCM I-4036) from the faeces of exclusively breast-fed infants. Br J Nutr. (2013) 109 (Suppl. 2):S51–62. 10.1017/S0007114512005211
    1. Jiang M, Deng K, Jiang C, Fu M, Guo C, Wang X, et al. . Evaluation of the antioxidative, antibacterial, and anti-inflammatory effects of the aloe fermentation supernatant containing Lactobacillus plantarum HM218749.1. Mediators Inflamm. (2016) 2016:2945650. 10.1155/2016/2945650
    1. Charteris WP, Kelly PM, Morelli L, Collins JK. Antibiotic susceptibility of potentially probiotic Lactobacillus species. J Food Prot. (1998) 61:1636–43. 10.4315/0362-028X-61.12.1636
    1. do Carmo MS, Noronha FM, Arruda MO, Costa EP, Bomfim MR, Monteiro AS, et al. . Lactobacillus fermentum ATCC 23271 displays in vitro inhibitory activities against Candida spp. Front Microbiol. (2016) 7:1722. 10.3389/fmicb.2016.01722
    1. Nishino M, Jagannathan JP, Ramaiya NH, Van den Abbeele AD. Revised RECIST guideline version 1.1: what oncologists want to know and what radiologists need to know. AJR Am J Roentgenol. (2010) 195:281–9. 10.2214/AJR.09.4110
    1. Velasquez-Mejia EP, de la Cuesta-Zuluaga J, Escobar JS. Impact of DNA extraction, sample dilution, and reagent contamination on 16S rRNA gene sequencing of human feces. Appl Microbiol Biotechnol. (2018) 102:403–11. 10.1007/s00253-017-8583-z
    1. Patel A, Biswas S, Shoja MH, Ramalingayya GV, Nandakumar K. Protective effects of aqueous extract of Solanum nigrum Linn. leaves in rat models of oral mucositis. ScientificWorldJournal. (2014) 2014:345939. 10.1155/2014/345939
    1. Lee SW, Jung KI, Kim YW, Jung HD, Kim HS, Hong JP. Effect of epidermal growth factor against radiotherapy-induced oral mucositis in rats. Int J Radiat Oncol Biol Phys. (2007) 67:1172–8. 10.1016/j.ijrobp.2006.10.038
    1. Zheng B, Zhu X, Liu M, Yang Z, Yang L, Lang J, et al. . Randomized, double-blind, placebo-controlled trial of shuanghua baihe tablets to prevent oral mucositis in patients with nasopharyngeal cancer undergoing chemoradiation therapy. Int J Radiat Oncol Biol Phys. (2018) 100:418–26. 10.1016/j.ijrobp.2017.10.013
    1. Magoc T, Salzberg SL. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics. (2011) 27:2957–63. 10.1093/bioinformatics/btr507
    1. Edgar RC. UPARSE: highly accurate OTU sequences from microbial amplicon reads. Nat Methods. (2013) 10:996–8. 10.1038/nmeth.2604
    1. Wang Q, Garrity GM, Tiedje JM, Cole JR. Naive bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl Environ Microbiol. (2007) 73:5261–7. 10.1128/AEM.00062-07
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. . QIIME allows analysis of high-throughput community sequencing data. Nat Methods. (2010) 7:335–6. 10.1038/nmeth.f.303
    1. Lozupone C, Lladser ME, Knights D, Stombaugh J, Knight R. UniFrac: an effective distance metric for microbial community comparison. Isme J. (2011) 5:169–72. 10.1038/ismej.2010.133
    1. Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, et al. . Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. (2013) 342:967–70. 10.1126/science.1240527
    1. Couzin-Frankel J. Breakthrough of the year 2013. Cancer immunotherapy. Science. (2013) 342:1432–3. 10.1126/science.342.6165.1432
    1. Hu J, Wang C, Ye L, Yang W, Huang H, Meng F, et al. . Anti-tumour immune effect of oral administration of Lactobacillus plantarum to CT26 tumour-bearing mice. J Biosci. (2015) 40:269–279. 10.1007/s12038-015-9518-4
    1. Perez-Chanona E, Trinchieri G. The role of microbiota in cancer therapy. Curr Opin Immunol. (2016) 39:75–81. 10.1016/j.coi.2016.01.003
    1. Mi H, Dong Y, Zhang B, Wang H, Peter CCK, Gao P, et al. . Bifidobacterium infantis ameliorates chemotherapy-induced intestinal mucositis via regulating T cell immunity in colorectal cancer rats. Cell Physiol Biochem. (2017) 42:2330–41. 10.1159/000480005
    1. Thomas S, Izard J, Walsh E, Batich K, Chongsathidkiet P, Clarke G, et al. . The host microbiome regulates and maintains human health: a primer and perspective for non-microbiologists. Cancer Res. (2017) 77:1783–812. 10.1158/0008-5472.CAN-16-2929
    1. Zitvogel L, Ayyoub M, Routy B, Kroemer G. Microbiome and anticancer immunosurveillance. Cell. (2016) 165:276–87. 10.1016/j.cell.2016.03.001
    1. Clemente M, Rizzetto L, Castronovo G, Perissi E, Tanturli M, Cozzolino F, et al. . Effects of near-infrared laser radiation on the survival and inflammatory potential of Candida spp. involved in the pathogenesis of chemotherapy-induced oral mucositis. Eur J Clin Microbiol Infect Dis. (2015) 34:1999–2007. 10.1007/s10096-015-2443-5
    1. Tancharoen S, Shakya P, Narkpinit S, Dararat P, Kikuchi K. Anthocyanins Extracted from Oryza sativa L. Prevent fluorouracil-induced nuclear factor-kappab activation in oral mucositis: in vitro and in vivo studies. Int J Mol Sci. (2018) 19:2981. 10.3390/ijms19102981
    1. Luo J, Bian L, Blevins MA, Wang D, Liang C, Du D, et al. . Smad7 promotes healing of radiotherapy-induced oral mucositis without compromising oral cancer therapy in a xenograft mouse model. Clin Cancer Res. (2019) 25:808–18. 10.1158/1078-0432.CCR-18-1081
    1. Viet CT, Corby PM, Akinwande A, Schmidt BL. Review of preclinical studies on treatment of mucositis and associated pain. J Dent Res. (2014) 93:868–75. 10.1177/0022034514540174
    1. Meirovitz A, Kuten M, Billan S, Abdah-Bortnyak R, Sharon A, Peretz T, et al. . Cytokines levels, severity of acute mucositis and the need of PEG tube installation during chemo-radiation for head and neck cancer–a prospective pilot study. Radiat Oncol. (2010) 5:16. 10.1186/1748-717X-5-16
    1. Mangoni M, Sottili M, Gerini C, Desideri I, Bastida C, Pallotta S, et al. . A PPAR gamma agonist protects against oral mucositis induced by irradiation in a murine model. Oral Oncol. (2017) 64:52–8. 10.1016/j.oraloncology.2016.11.018
    1. Mu Q, Kirby J, Reilly CM, Luo XM. Leaky gut as a danger signal for autoimmune diseases. Front Immunol. (2017) 8:598. 10.3389/fimmu.2017.00598
    1. Suzuki T. Regulation of intestinal epithelial permeability by tight junctions. Cell Mol Life Sci. (2013) 70:631–59. 10.1007/s00018-012-1070-x
    1. van Vliet MJ, Harmsen HJ, de Bont ES, Tissing WJ. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog. (2010) 6:e1000879. 10.1371/journal.ppat.1000879
    1. Touchefeu Y, Montassier E, Nieman K, Gastinne T, Potel G, Bruley des Varannes S, et al. . Systematic review: the role of the gut microbiota in chemotherapy- or radiation-induced gastrointestinal mucositis - current evidence and potential clinical applications. Aliment Pharmacol Ther. (2014) 40:409–21. 10.1111/apt.12878
    1. Li Y, Dong J, Xiao H, Zhang S, Wang B, Cui M, et al. . Gut commensal derived-valeric acid protects against radiation injuries. Gut Microbes. (2020) 11:789–806. 10.1080/19490976.2019.1709387
    1. Laval L, Martin R, Natividad JN, Chain F, Miquel S, Desclee de Maredsous C, et al. . Lactobacillus rhamnosus CNCM I-3690 and the commensal bacterium Faecalibacterium prausnitzii A2-165 exhibit similar protective effects to induced barrier hyper-permeability in mice. Gut Microbes. (2015) 6:1–9. 10.4161/19490976.2014.990784
    1. Celiberto LS, Pinto RA, Rossi EA, Vallance BA, Cavallini DCU. Isolation and characterization of potentially probiotic bacterial strains from mice: proof of concept for personalized probiotics. Nutrients. (2018) 10:1684. 10.3390/nu10111684
    1. Hong BY, Sobue T, Choquette L, Dupuy AK, Thompson A, Burleson JA, et al. . Chemotherapy-induced oral mucositis is associated with detrimental bacterial dysbiosis. Microbiome. (2019) 7:66. 10.1186/s40168-019-0679-5
    1. Al-Qadami G, Van Sebille Y, Le H, Bowen J. Gut microbiota: implications for radiotherapy response and radiotherapy-induced mucositis. Expert Rev Gastroenterol Hepatol. (2019) 13:485–96. 10.1080/17474124.2019.1595586
    1. Okubo R, Kinoshita T, Katsumata N, Uezono Y, Xiao J, Matsuoka YJ. Impact of chemotherapy on the association between fear of cancer recurrence and the gut microbiota in breast cancer survivors. Brain Behav Immun. (2019) 85:186–91. 10.1016/j.bbi.2019.02.025
    1. Montassier E, Gastinne T, Vangay P, Al-Ghalith GA, Bruley des Varannes S, Massart S, et al. . Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment Pharmacol Ther. (2015) 42:515–28. 10.1111/apt.13302
    1. Simon-Soro A, Tomas I, Cabrera-Rubio R, Catalan MD, Nyvad B, Mira A. Microbial geography of the oral cavity. J Dent Res. (2013) 92:616–21. 10.1177/0022034513488119
    1. Zhu XX, Yang XJ, Chao YL, Zheng HM, Sheng HF, Liu HY, et al. . The potential effect of oral microbiota in the prediction of mucositis during radiotherapy for nasopharyngeal carcinoma. EBioMedicine. (2017) 18:23–31. 10.1016/j.ebiom.2017.02.002
    1. Swidsinski A, Weber J, Loening-Baucke V, Hale LP, Lochs H. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J Clin Microbiol. (2005) 43:3380–9. 10.1128/JCM.43.7.3380-3389.2005
    1. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. . Richness of human gut microbiome correlates with metabolic markers. Nature. (2013) 500:541–6. 10.1038/nature12506
    1. Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermudez-Humaran LG, Gratadoux JJ, et al. . Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci USA. (2008) 105:16731–6. 10.1073/pnas.0804812105
    1. Chakraborti CK. New-found link between microbiota and obesity. World J Gastrointest Pathophysiol. (2015) 6:110–9. 10.4291/wjgp.v6.i4.110
    1. Canani RB, Costanzo MD, Leone L, Pedata M, Meli R, Calignano A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J Gastroenterol. (2011) 17:1519–28. 10.3748/wjg.v17.i12.1519
    1. Uchiyama K, Naito Y, Takagi T. Intestinal microbiome as a novel therapeutic target for local and systemic inflammation. Pharmacol Ther. (2019) 199:164–72. 10.1016/j.pharmthera.2019.03.006
    1. Joossens M, De Preter V, Ballet V, Verbeke K, Rutgeerts P, Vermeire S. Effect of oligofructose-enriched inulin (OF-IN) on bacterial composition and disease activity of patients with Crohn's disease: results from a double-blinded randomised controlled trial. Gut. (2012) 61:958. 10.1136/gutjnl-2011-300413
    1. Braat H, Rottiers P, Hommes DW, Huyghebaert N, Remaut E, Remon JP, et al. . A phase I trial with transgenic bacteria expressing interleukin-10 in Crohn's disease. Clin Gastroenterol Hepatol. (2006) 4:754–9. 10.1016/j.cgh.2006.03.028
    1. Gerhard D, Sousa F, Andraus RAC, Pardo PE, Nai GA, Neto HB, et al. . Probiotic therapy reduces inflammation and improves intestinal morphology in rats with induced oral mucositis. Braz Oral Res. (2017) 31:e71. 10.1590/1807-3107bor-2017.vol31.0071
    1. Prisciandaro LD, Geier MS, Butler RN, Cummins AG, Howarth GS. Evidence supporting the use of probiotics for the prevention and treatment of chemotherapy-induced intestinal mucositis. Crit Rev Food Sci Nutr. (2011) 51:239–47. 10.1080/10408390903551747
    1. Pico-Monllor JA, Mingot-Ascencao JM. Search and selection of probiotics that improve mucositis symptoms in oncologic patients. A systematic review. Nutrients. (2019) 11:2322. 10.3390/nu11102322
    1. Kato S, Hamouda N, Kano Y, Oikawa Y, Tanaka Y, Matsumoto K, et al. . Probiotic Bifidobacterium bifidum G9-1 attenuates 5-fluorouracil-induced intestinal mucositis in mice via suppression of dysbiosis-related secondary inflammatory responses. Clin Exp Pharmacol Physiol. (2017) 44:1017–25. 10.1111/1440-1681.12792
    1. Chang CW, Liu CY, Lee HC, Huang YH, Li LH, Chiau JC, 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. 10.3389/fmicb.2018.00983
    1. Cereda E, Caraccia M, Caccialanza R. Probiotics and mucositis. Curr Opin Clin Nutr Metab Care. (2018) 21:399–404. 10.1097/MCO.0000000000000487

Source: PubMed

3
Se inscrever