Rifaximin is associated with modest, transient decreases in multiple taxa in the gut microbiota of patients with diarrhoea-predominant irritable bowel syndrome

Anthony A Fodor, Mark Pimentel, William D Chey, Anthony Lembo, Pamela L Golden, Robert J Israel, Ian M Carroll, Anthony A Fodor, Mark Pimentel, William D Chey, Anthony Lembo, Pamela L Golden, Robert J Israel, Ian M Carroll

Abstract

Rifaximin, a non-systemic antibiotic, is efficacious for the treatment of diarrhoea-predominant irritable bowel syndrome (IBS-D). Given the emerging association between the gut microbiota and IBS, this study examined potential effects of rifaximin on the gastrointestinal microbial community in patients with IBS-D. TARGET 3 was a randomised, double-blind, placebo-controlled, phase 3 study. Patients with IBS-D initially received open-label rifaximin 550 mg 3 times daily (TID) for 2 weeks. Patients who responded to the initial treatment and then relapsed were randomised to receive 2 repeat courses of rifaximin 550 mg TID or placebo for 2 weeks, with each course separated by 10 weeks. Stool samples were collected at the beginning and end of open-label treatment, at the beginning and end of the first double-blind treatment, and at the end of the study. As a secondary analysis to the TARGET 3 trial, the composition and diversity of the gut microbiota were assessed, from a random subset of patients, using variable 4 hypervariable region 16S ribosomal RNA gene sequencing. Samples from 103 patients were included. After open-label rifaximin treatment for 2 weeks, 7 taxa (e.g. Peptostreptococcaceae, Verrucomicrobiaceae, Enterobacteriaceae) had significantly lower relative abundance at a 10% false discovery rate threshold. The effects of rifaximin were generally short-term, as there was little evidence of significantly different changes in taxa relative abundance at the end of the study (up to 46 weeks) versus baseline. The results suggest that rifaximin has a modest, largely transient effect across a broad range of stool microbes. Future research may determine whether the taxa affected by rifaximin are causally linked to IBS-D. ClinicalTrials.gov identifier number: NCT01543178.

Keywords: Antibiotic therapy; colonic microflora; diarrhoea; gastrointestinal immune response; irritable bowel syndrome.

Figures

Figure 1.
Figure 1.
Study design. Adapted with permission from Lembo, et al. EOS, end of study; TID: 3 times daily, V: clinic visit
Figure 2.
Figure 2.
Multidimensional scaling (MDS) ordination at the family level for all samples colored by visit (left panel) and batch (right panel). Batch 1 and batch 2 show substantial overlap, as do batch 3 and batch 4. Tests from 9999 ADONIS permutations yielded a significant difference (p = 0.016) for V3 vs V4 and the effect of patient (p

Figure 3.

Rifaximin induces a small, transient…

Figure 3.

Rifaximin induces a small, transient reduction in the relative abundance of multiple taxa.…

Figure 3.
Rifaximin induces a small, transient reduction in the relative abundance of multiple taxa. (A) For 74 non‒rare taxa observed in ≥10% of all samples, the mean relative abundance of each taxon is shown in V3 (n = 101) and V4 (n = 102) samples. (B) For the same taxa, the mean relative abundance of taxa is shown in V3 and V11. The symbols in red indicate taxa that were significantly different in paired Wilcoxon test comparisons of log-transformed data (Supplementary Table S3). The black line is the identity line (V3 = V4). V: clinic visit

Figure 4.

The number of taxa observed…

Figure 4.

The number of taxa observed as different from V3 to V4 (A) and…

Figure 4.
The number of taxa observed as different from V3 to V4 (A) and to V11 (B) using smaller sample sizes. At each sample size, the number of patients indicated on the x-axis was subsampled 50 times (without replacement). The mean and SD of the number of taxa that were detected at a 10% FDR by the paired t-test are shown for taxa with relative abundances that were depressed (black) or enhanced (red) in association with rifaximin treatment. FDR: false discovery rate; SD: standard deviation; V: clinic visit

Figure 5.

Comparison between the first (V3–V4)…

Figure 5.

Comparison between the first (V3–V4) and second (V6–V7) treatment of uncorrected p values…

Figure 5.
Comparison between the first (V3–V4) and second (V6–V7) treatment of uncorrected p values for non‒rare taxa for the null hypothesis that patients responded to rifaximin (A) or placebo (B) as evaluated by the paired t-test. The y-axis shows the p value for the null hypothesis that treatment had no effect for each taxa for the V6–V7 time point (second treatment) where patients were treated at this time point with either rifaximin (A) or placebo (B). The x-axis shows the results of evaluation of the null hypothesis for the V3 and V4 time points (first treatment), where all patients were treated with rifaximin. To distinguish the direction of response of each treatment, if the mean after treatment was higher than before treatment, the log p value was multiplied by –1. Only patients for whom the researchers had samples for all 4 time points were included in this analysis (33 patients treated with rifaximin for V6–V7; 30 patients in the placebo group for V6–V7) V: clinic visit
Figure 3.
Figure 3.
Rifaximin induces a small, transient reduction in the relative abundance of multiple taxa. (A) For 74 non‒rare taxa observed in ≥10% of all samples, the mean relative abundance of each taxon is shown in V3 (n = 101) and V4 (n = 102) samples. (B) For the same taxa, the mean relative abundance of taxa is shown in V3 and V11. The symbols in red indicate taxa that were significantly different in paired Wilcoxon test comparisons of log-transformed data (Supplementary Table S3). The black line is the identity line (V3 = V4). V: clinic visit
Figure 4.
Figure 4.
The number of taxa observed as different from V3 to V4 (A) and to V11 (B) using smaller sample sizes. At each sample size, the number of patients indicated on the x-axis was subsampled 50 times (without replacement). The mean and SD of the number of taxa that were detected at a 10% FDR by the paired t-test are shown for taxa with relative abundances that were depressed (black) or enhanced (red) in association with rifaximin treatment. FDR: false discovery rate; SD: standard deviation; V: clinic visit
Figure 5.
Figure 5.
Comparison between the first (V3–V4) and second (V6–V7) treatment of uncorrected p values for non‒rare taxa for the null hypothesis that patients responded to rifaximin (A) or placebo (B) as evaluated by the paired t-test. The y-axis shows the p value for the null hypothesis that treatment had no effect for each taxa for the V6–V7 time point (second treatment) where patients were treated at this time point with either rifaximin (A) or placebo (B). The x-axis shows the results of evaluation of the null hypothesis for the V3 and V4 time points (first treatment), where all patients were treated with rifaximin. To distinguish the direction of response of each treatment, if the mean after treatment was higher than before treatment, the log p value was multiplied by –1. Only patients for whom the researchers had samples for all 4 time points were included in this analysis (33 patients treated with rifaximin for V6–V7; 30 patients in the placebo group for V6–V7) V: clinic visit

References

    1. Lacy BE, Mearin F, Chang L, Chey WD, Lembo AJ, Simren M, Spiller R. Bowel disorders. Gastroenterology 2016; 150:1393–407.
    1. Chey WD, Kurlander J, Eswaran S. Irritable bowel syndrome: a clinical review. JAMA 2015; 313:949–58.
    1. Knight JR, Locke GR, Zinsmeister AR, Schleck CD, Talley NJ. Family history of mental illness or alcohol abuse and the irritable bowel syndrome. J Psychosom Res 2015; 78:237–41.
    1. Bashashati M, Rezaei N, Shafieyoun A, McKernan DP, Chang L, Ohman L, Quigley EM, Schmulson M, Sharkey KA, Simren M. Cytokine imbalance in irritable bowel syndrome: a systematic review and meta-analysis. Neurogastroenterol Motil 2014; 26:1036–48.
    1. Rana SV, Sharma S, Sinha SK, Parsad KK, Malik A, Singh K. Pro-inflammatory and anti-inflammatory cytokine response in diarrhoea-predominant irritable bowel syndrome patients. Trop Gastroenterol 2012; 33:251–56.
    1. Guilarte M, Santos J, de Torres I, Alonso C, Vicario M, Ramos L, Martinez C, Casellas F, Saperas E, Malagelada JR. Diarrhoea-predominant IBS patients show mast cell activation and hyperplasia in the jejunum. Gut 2007; 56:203–09.
    1. Vivinus-Nébot M, Frin-Mathy G, Bzioueche H, Dainese R, Bernard G, Anty R, Filippi J, Saint-Paul MC, Tulic MK, Verhasselt V, et al. . Functional bowel symptoms in quiescent inflammatory bowel diseases: role of epithelial barrier disruption and low-grade inflammation. Gut 2014; 63:744–52.
    1. Ahn JY, Lee KH, Choi CH, Kim JW, Lee HW, Kim JW, Kim MK, Kwon GY, Han S, Kim SE, et al. . Colonic mucosal immune activity in irritable bowel syndrome: comparison with healthy controls and patients with ulcerative colitis. Dig Dis Sci 2014; 59:1001–11.
    1. Holliday EG, Attia J, Hancock S, Koloski N, McEvoy M, Peel R, D'Amato M, Agreus L, Nyhlin H, Andreasson A, et al. . Genome-wide association study identifies two novel genomic regions in irritable bowel syndrome. Am J Gastroenterol 2014; 109:770–72.
    1. Wouters MM, Lambrechts D, Knapp M, Cleynen I, Whorwell P, Agreus L, Dlugosz A, Schmidt PT, Halfvarson J, Simren M, et al. . Genetic variants in CDC42 and NXPH1 as susceptibility factors for constipation and diarrhoea predominant irritable bowel syndrome. Gut 2014; 63:1103–11.
    1. Zhou Q, Zhang B, Verne GN. Intestinal membrane permeability and hypersensitivity in the irritable bowel syndrome. Pain 2009; 146:41–46.
    1. Shulman RJ, Jarrett ME, Cain KC, Broussard EK, Heitkemper MM. Associations among gut permeability, inflammatory markers, and symptoms in patients with irritable bowel syndrome. J Gastroenterol 2014; 49:1467–76.
    1. Zanini B, Ricci C, Bandera F, Caselani F, Magni A, Laronga AM, Lanzini A. Incidence of post-infectious irritable bowel syndrome and functional intestinal disorders following a water-borne viral gastroenteritis outbreak. Am J Gastroenterol 2012; 107:891–99.
    1. Nair P, Okhuysen PC, Jiang ZD, Carlin LG, Belkind-Gerson J, Flores J, Paredes M, DuPont HL. Persistent abdominal symptoms in US adults after short-term stay in Mexico. J Travel Med 2014; 21:153–58.
    1. Durbán A, Abellán JJ, Jimenéz-Hernández N, Salgado P, Ponce M, Ponce J, Garrigues V, Latorre A, Moya A. Structural alterations of faecal and mucosa-associated bacterial communities in irritable bowel syndrome. Environ Microbiol Rep 2012; 4:242–47.
    1. Durbán A, Abellán JJ, Jiménez-Hernández N, Artacho A, Garrigues V, Ortiz V, Ponce J, Latorre A, Moya A. Instability of the faecal microbiota in diarrhoea-predominant irritable bowel syndrome. FEMS Microbiol Ecol 2013; 86:581–89.
    1. Carroll IM, Ringel-Kulka T, Keku TO, Chang YH, Packey CD, Sartor RB, Ringel Y. Molecular analysis of the luminal- and mucosal-associated intestinal microbiota in diarrhea-predominant irritable bowel syndrome. Am J Physiol Gastrointest Liver Physiol 2011; 301:G799–G807.
    1. Codling C, O'Mahony L, Shanahan F, Quigley EMM, Marchesi JR. A molecular analysis of fecal and mucosal bacterial communities in irritable bowel syndrome. Dig Dis Sci 2010; 55:392–97.
    1. Hungin APS, Mulligan C, Pot B, Whorwell P, Agreus L, Fracasso P, Lionis C, Mendive J, Philippart de Foy JM, Rubin G, et al. . Systematic review: probiotics in the management of lower gastrointestinal symptoms in clinical practice – an evidence-based international guide. Aliment Pharmacol Ther 2013; 38:864–86.
    1. Frissora CL, Cash BD. Review article: the role of antibiotics vs. conventional pharmacotherapy in treating symptoms of irritable bowel syndrome. Aliment Pharmacol Ther 2007; 25:1271–81.
    1. Pimentel M, Chatterjee S, Chow EJ, Park S, Kong Y. Neomycin improves constipation-predominant irritable bowel syndrome in a fashion that is dependent on the presence of methane gas: subanalysis of a double-blind randomized controlled study. Dig Dis Sci 2006; 51:1297–301.
    1. Jiang ZD, DuPont HL. Rifaximin: in vitro and in vivo antibacterial activity–a review. Chemotherapy 2005; 51:67–72.
    1. Gillis JC, Brogden RN. Rifaximin. A review of its antibacterial activity, pharmacokinetic properties and therapeutic potential in conditions mediated by gastrointestinal bacteria. Drugs 1995; 49:467–84.
    1. Marchese A, Salerno A, Pesce A, Debbia EA, Schito GC. In vitro activity of rifaximin, metronidazole and vancomycin against Clostridium difficile and the rate of selection of spontaneously resistant mutants against representative anaerobic and aerobic bacteria, including ammonia-producing species. Chemotherapy 2000; 46:253–66.
    1. Xifaxan(rifaximin) tablets, for oral use [package insert]. Bridgewater, NJ: Salix Pharmaceuticals; 2015.
    1. Xu D, Gao J, Gillilland M, Wu X, Song I, Kao JY, Owyang C. Rifaximin alters intestinal bacteria and prevents stress-induced gut inflammation and visceral hyperalgesia in rats. Gastroenterology 2014; 146:484–96.
    1. Pimentel M, Lembo A, Chey WD, Zakko S, Ringel Y, Yu J, Mareya SM, Shaw AL, Bortey E, Forbes WP. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med 2011; 364:22–32.
    1. Pimentel M, Park S, Mirocha J, Kane SV, Kong Y. The effect of a nonabsorbed oral antibiotic (rifaximin) on the symptoms of the irritable bowel syndrome: a randomized trial. Ann Intern Med 2006; 145:557–63.
    1. Meyrat P, Safroneeva E, Schoepfer AM. Rifaximin treatment for the irritable bowel syndrome with a positive lactulose hydrogen breath test improves symptoms for at least 3 months. Aliment Pharmacol Ther 2012; 36:1084–93.
    1. Pimentel M, Morales W, Chua K, Barlow G, Weitsman S, Kim G, Amichai MM, Pokkunuri V, Rook E, Mathur R, et al. . Effects of rifaximin treatment and retreatment in nonconstipated IBS subjects. Dig Dis Sci 2011; 56:2067–72.
    1. Schoenfeld P, Pimentel M, Chang L, Lembo A, Chey WD, Yu J, Paterson C, Bortey E, Forbes WP. Safety and tolerability of rifaximin for the treatment of irritable bowel syndrome without constipation: a pooled analysis of randomised, double-blind, placebo-controlled trials. Aliment Pharmacol Ther 2014; 39:1161–68.
    1. Lembo A, Pimentel M, Rao SS, Schoenfeld P, Cash B, Weinstock LB, Paterson C, Bortey E, Forbes WP. Repeat treatment with rifaximin is safe and effective in patients with diarrhea-predominant irritable bowel syndrome. Gastroenterology 2016; 151:1113–21.
    1. Panda S, El khader I, Casellas F, Lopez VJ, Garcia CM, Santiago A, Cuenca S, Guarner F, Manichanh C. Short-term effect of antibiotics on human gut microbiota. PLoS One 2014; 9:e95476.
    1. Perez-Cobas AE, Artacho A, Knecht H, Ferrus ML, Friedrichs A, Ott SJ, Moya A, Latorre A, Gosalbes MJ. Differential effects of antibiotic therapy on the structure and function of human gut microbiota. PLoS One 2013; 8:e80201.
    1. Acosta A, Camilleri M, Shin A, Linker Nord S, O'Neill J, Gray AV, Lueke AJ, Donato LJ, Burton DD, Szarka LA, et al. . Effects of rifaximin on transit, permeability, fecal microbiome, and organic acid excretion in irritable bowel syndrome. Clin Transl Gastroenterol 2016; 7:e173.
    1. Zeber-Lubecka N, Kulecka M, Ambrozkiewicz F, Paziewska A, Goryca K, Karczmarski J, Rubel T, Wojtowicz W, Mlynarz P, Marczak L, et al. . Limited prolonged effects of rifaximin treatment on irritable bowel syndrome-related differences in the fecal microbiome and metabolome. Gut Microbes 2016; 7:397–413.
    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; 73:5261–67.
    1. Tourlousse DM, Yoshiike S, Ohashi A, Matsukura S, Noda N, Sekiguchi Y. Synthetic spike-in standards for high-throughput 16S rRNA gene amplicon sequencing. Nucleic Acids Res 2017; 45:e23.
    1. Sanapareddy N, Legge RM, Jovov B, McCoy A, Burcal L, Araujo-Perez F, Randall TA, Galanko J, Benson A, Sandler RS, et al. . Increased rectal microbial richness is associated with the presence of colorectal adenomas in humans. ISME J 2012; 6:1858–68.
    1. McMurdie PJ, Holmes S. Waste not, want not: why rarefying microbiome data is inadmissible. PLoS Comput Biol 2014; 10:e1003531.
    1. Maccaferri S, Vitali B, Klinder A, Kolida S, Ndagijimana M, Laghi L, Calanni F, Brigidi P, Gibson GR, Costabile A. Rifaximin modulates the colonic microbiota of patients with Crohn's disease: an in vitro approach using a continuous culture colonic model system. J Antimicrob Chemother 2010; 65:2556–65.
    1. Swidsinski A, Loening-Baucke V, Verstraelen H, Osowska S, Doerffel Y. Biostructure of fecal microbiota in healthy subjects and patients with chronic idiopathic diarrhea. Gastroenterology 2008; 135:568–79.
    1. Ringel Y, Maharshak N, Ringel-Kulka T, Wolber EA, Sartor RB, Carroll IM. High throughput sequencing reveals distinct microbial populations within the mucosal and luminal niches in healthy individuals. Gut Microbes 2015; 6:173–81.
    1. Fouhy F, Ross RP, Fitzgerald GF, Stanton C, Cotter PD. Composition of the early intestinal microbiota: knowledge, knowledge gaps and the use of high-throughput sequencing to address these gaps. Gut Microbes 2012; 3:203–20.
    1. Tu Q, He Z, Zhou J. Strain/species identification in metagenomes using genome-specific markers. Nucleic Acids Res 2014; 42:e67.

Source: PubMed

3
Abonnere