Fecal Microbiota Transplantation for Ulcerative Colitis: The Optimum Timing and Gut Microbiota as Predictors for Long-Term Clinical Outcomes

Qianqian Li, Xiao Ding, Kangjian Liu, Cicilia Marcella, Xiaolin Liu, Ting Zhang, Yafei Liu, Pan Li, Liyuan Xiang, Bota Cui, Jun Wang, Jianling Bai, Faming Zhang, Qianqian Li, Xiao Ding, Kangjian Liu, Cicilia Marcella, Xiaolin Liu, Ting Zhang, Yafei Liu, Pan Li, Liyuan Xiang, Bota Cui, Jun Wang, Jianling Bai, Faming Zhang

Abstract

Introduction: The previous researches aimed to evaluate the efficacy and safety of fecal microbiota transplantation (FMT) for ulcerative colitis (UC) in a short-term observation. The present study aimed to explore the optimum timing of FMT for maintaining the long-term clinical benefits and to target the gut microbiota that may help to predict the long-term success or failure of FMT in UC.

Methods: Two hundred two patients with UC were recruited from November 2012 to September 2018. The primary endpoint of this study was the maintaining time of the first and second courses of FMT. Relapse was defined as partial Mayo score ≥2 after achieving clinical remission and an increase of partial Mayo score ≥1 after achieving clinical response. The stool samples were analyzed by 16S rRNA gene sequencing.

Results: The median maintaining time of the efficacy was 120 days (IQR, 45-180) and 182.5 days (IQR, 105-311.25) from the first course and second course of FMT, respectively. No FMT-related serious adverse events were observed. The differences of the relative abundance in Eggerthella, Lactobacillus, and Ruminococcus between pre-FMT and 5 days post-FMT were remarkably correlated with the long-term clinical remission (P < 0.05).

Discussion: This study demonstrated that patients with UC should undergo the second course of FMT within 4 months after the first course of FMT for maintaining the long-term clinical benefits. The short-term alterations of microbiota after FMT may be conducive to predicting the long-term efficacy of FMT in UC (see Visual Abstract, Supplementary Digital Content, http://links.lww.com/CTG/A363).

Trial registration: ClinicalTrials.gov NCT01790061.

Conflict of interest statement

Guarantor of the article: Faming Zhang, MD, PhD.

Specific author contributions: Qianqian Li, MD and Xiao Ding, MD, contributed equally to this work. F.Z., Q.L., and X.D. designed the study and edited the manuscript. Q.L., X.D., K.L., T.Z., L.X., B.C., P.L., and F.Z. were responsible for the recruitment and treatment of the patients. Q.L. and X.D. participated in the data analysis and wrote the manuscript. C.M. wrote and revised the manuscript. X.L., Y.L., and J.W. devoted to the visualization. J.B. provided scientific statistic analysis methods. All authors read and approved the final manuscript.

Funding support: This study was funded by the publicly donated Intestine Initiative Foundation; Primary Research & Development Plan of Jiangsu Province (BE2018751), Jiangsu Provincial Medical Innovation Team (Zhang F); National Natural Science Foundation of China (81873548, 81670495, 81600417); and the National Clinical Research Center for Digestive Diseases, Xi'an, China (2015BAI13B07).

Potential competing interests: None to report.

Ethics approval: This study was reviewed and approved by the Second Affiliated Hospital of Nanjing Medical University Institutional Review Board. All eligible subjects provided written informed consents before participation in this study.

Availability of data and materials: All data generated or analysed during this study are included in this published article (and its supplementary information files).

Trial registration: ClinicalTrials.gov NCT01790061.

Figures

Figure 1.
Figure 1.
Flowchart of the study. FMT, fecal microbiota transplantation; UC, ulcerative colitis.
Figure 2.
Figure 2.
Maintaining time of the efficacy from FMT. (a) The maintaining time of the first course of FMT in 122 patients with UC. Compared with the maintaining time of the NCT group (n = 99), the CT group (n = 23) achieved longer maintaining time significantly (P < 0.001). The median maintaining time of the first course of FMT in the nonconsolidation group was 120 days (IQR, 45–180). (b) The maintaining time of the second course of FMT in 24 patients with UC. The median maintaining time of the second course of FMT was 182.5 days (IQR, 105–311.25). Kaplan–Meier survival curves were used to describe the time for relapse. P value less than 0.05 was considered statistically significant. CT, consolidation treatment; FMT, fecal microbiota transplantation; NCT, nonconsolidation treatment.
Figure 3.
Figure 3.
The partial Mayo scores at various visits. (a) The partial Mayo scores of 99 patients before and after the first course of FMT. (b) The partial Mayo scores of 24 patients before and after the second course of FMT. More than 2 groups were analyzed by the ANOVA test. P value less than 0.05 was considered statistically significant. Significance levels: *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. ANOVA, analysis of variance; FMT, fecal microbiota transplantation.
Figure 4.
Figure 4.
The diversity of microbiota in donors and patients with UC before and after FMT. (a and b) Alpha diversity was calculated by Shannon evenness and Chao 1 index. The donors' and post-FMT patients' samples were compared with pre-FMT patients' samples. (c and d) Beta diversity was calculated by MDS and CAP coordinates. Each dot stands for 1 sample. P value less than 0.05 was considered statistically significant. Significance levels: *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. The number of samples: donors (n = 19), patients with UC pre-FMT (n = 22), patients with UC post-FMT (n = 20). CAP, canonical analysis of principal; FMT, fecal microbiota transplantation; MDS, multidimensional scaling; UC, ulcerative colitis.
Figure 5.
Figure 5.
Nineteen significant differential genera between patients with UC and donors. Differences were analyzed by the Kruskal-Wallis test. P value less than 0.05 was considered statistically significant. Significance levels: *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. (a) Pre-FMT vs donor, (b) Post-FMT vs donor, and (c) Pre-FMT vs post-FMT. The number of samples: donors (n = 19), patients with UC pre-FMT (n = 22), patients with UC post-FMT (n = 20). FMT, fecal microbiota transplantation; UC, ulcerative colitis.
Figure 6.
Figure 6.
Microbiota related to the long-term efficacy of FMT in patients with UC. (a) The partial Mayo score of the 22 patients at various visits (pre-FMT, 1 week post-FMT and 1 month post-FMT). The 7 patients (boxed) achieved clinical remission and maintaining time was at least 4 months. (b and c) Beta diversity was calculated by MDS and CAP coordinates in the remission (R) and nonremission (NR) groups. Each dot stands for 1 sample. (df) The relative abundance differences of genera Eggerthella (r = 0.542, P = 0.014), Lactobacillus (r = 0.471, P = 0.036), and Ruminococcus (r = 0.523, P = 0.018) between pre-FMT and 5 days post-FMT were positively correlated to the long-term efficacy (4 months after FMT) significantly. P value less than 0.05 was considered statistically significant. The number of samples: Rdonor (n = 7), NRdonor (n = 12), Rpre (n = 7), NRpre (n = 15), Rpost (n = 5), and NRpost (n = 15). CAP, canonical analysis of principal; FMT, fecal microbiota transplantation; NRpre, genera in the nonremission group before FMT; NRpost, genera in the nonremission group after FMT; Rdonor, genera in the remission group of donor; Rpre, relative abundance of genera in the remission group before FMT; Rpost, genera in the remission group after FMT; MDS, multidimensional scaling; NRdonor, genera in the nonremission group of donor; UC, ulcerative colitis.
Figure 7.
Figure 7.
Comparisons of the 19 genera relative abundance between UC patients with sustained remission and nonremission from FMT. NRpre, genera in the nonremission group before FMT; NRpost, genera in the nonremission group after FMT; Rpost, genera in the remission group after FMT; Rpre, relative abundance of genera in the remission group before FMT. P value less than 0.05 was considered statistically significant. Significance levels: *P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001. The number of samples: donor (n = 19), Rpre (n = 7), NRpre (n = 15), Rpost (n = 5), and NRpost (n = 15).

References

    1. Ungaro R, Mehandru S, Allen PB, et al. Ulcerative colitis. Lancet 2017;389(10080):1756–70.
    1. Ordas I, Eckmann L, Talamini M, et al. Ulcerative colitis. Lancet 2012;380(9853):1606–19.
    1. Caruso R, Lo BC, Nunez G. Host-microbiota interactions in inflammatory bowel disease. Nat Rev Immunol 2020;20(7):411–426.
    1. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature 2007;448(7152):427–34.
    1. Garrett WS, Lord GM, Punit S, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell 2007;131(1):33–45.
    1. Dianda L, Hanby AM, Wright NA, et al. T cell receptor-alpha beta-deficient mice fail to develop colitis in the absence of a microbial environment. Am J Pathol 1997;150(1):91–7.
    1. Dalal SR, Chang EB. The microbial basis of inflammatory bowel diseases. J Clin Invest 2014;124(10):4190–6.
    1. Mortha A, Chudnovskiy A, Hashimoto D, et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 2014;343(6178):1249288.
    1. Cui B, Li P, Xu L, et al. Step-up fecal microbiota transplantation strategy: A pilot study for steroid-dependent ulcerative colitis. J Transl Med 2015;13:298.
    1. Timmer A, Patton PH, Chande N, et al. Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev 2016;5:CD000478.
    1. Sandborn WJ, van Assche G, Reinisch W, et al. Adalimumab induces and maintains clinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology 2012;142(2):257–65 e251-253.
    1. Feagan BG, Rutgeerts P, Sands BE, et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2013;369(8):699–710.
    1. Ding X, Li Q, Li P, et al. Long-term safety and efficacy of fecal microbiota transplant in active ulcerative colitis. Drug Saf 2019;42(7):869–80.
    1. Costello SP, Hughes PA, Waters O, et al. Effect of fecal microbiota transplantation on 8-week remission in patients with ulcerative colitis: A randomized clinical trial. JAMA 2019;321(2):156–64.
    1. Moayyedi P, Surette MG, Kim PT, et al. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology 2015;149(1):102–9.e106.
    1. Rossen NG, Fuentes S, van der Spek MJ, et al. Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis. Gastroenterology 2015;149(1):110–8.e114.
    1. Nishida A, Imaeda H, Ohno M, et al. Efficacy and safety of single fecal microbiota transplantation for Japanese patients with mild to moderately active ulcerative colitis. J Gastroenterol 2017;52(4):476–82.
    1. Paramsothy S, Nielsen S, Kamm MA, et al. Specific bacteria and metabolites associated with response to fecal microbiota transplantation in patients with ulcerative colitis. Gastroenterology 2019;156(5):1440–54.e1442.
    1. Sood A, Mahajan R, Juyal G, et al. Efficacy of fecal microbiota therapy in steroid dependent ulcerative colitis: A real world intention-to-treat analysis. Intest Res 2019;17(1):78–86.
    1. Li P, Zhang T, Xiao Y, et al. Timing for the second fecal microbiota transplantation to maintain the long-term benefit from the first treatment for Crohn's disease. Appl Microbiol Biotechnol 2019;103(1):349–60.
    1. Schirmer M, Denson L, Vlamakis H, et al. Compositional and temporal changes in the gut microbiome of pediatric ulcerative colitis patients are linked to disease course. Cell Host Microbe 2018;24(4):600–10.e604.
    1. Maukonen J, Kolho KL, Paasela M, et al. Altered fecal microbiota in paediatric inflammatory bowel disease. J Crohns Colitis 2015;9(12):1088–95.
    1. Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012;491(7422):119–24.
    1. He Z, Li P, Zhu J, et al. Multiple fresh fecal microbiota transplants induces and maintains clinical remission in Crohn's disease complicated with inflammatory mass. Sci Rep 2017;7(1):4753.
    1. Zhang T, Lu G, Zhao Z, et al. Washed microbiota transplantation vs. manual fecal microbiota transplantation: Clinical findings, animal studies and in vitro screening. Protein Cell 2020;11(4):251–266.
    1. Fecal Microbiota Transplantation-standardization Study Group. Nanjing consensus on methodology of washed microbiota transplantation. Chin Med J 2020. [Epub ahead of print July 21, 2020.]. doi:10.1097/CM9.0000000000000954
    1. Peng Z, Xiang J, He Z, et al. Colonic transendoscopic enteral tubing: A novel way of transplanting fecal microbiota. Endosc Int Open 2016;4(6):E610–613.
    1. Long C, Yu Y, Cui B, et al. A novel quick transendoscopic enteral tubing in mid-gut: Technique and training with video. BMC Gastroenterol 2018;18(1):37.
    1. Baumgart DC, Bokemeyer B, Drabik A, et al. Vedolizumab induction therapy for inflammatory bowel disease in clinical practice—A nationwide consecutive German cohort study. Aliment Pharmacol Ther 2016;43(10):1090–102.
    1. Aden K, Rehman A, Waschina S, et al. Metabolic functions of gut microbes associate with efficacy of tumor necrosis factor antagonists in patients with inflammatory bowel diseases. Gastroenterology 2019;157(5):1279–1292.
    1. Sood A, Mahajan R, Singh A, et al. Role of fecal microbiota transplantation for maintenance of remission in patients with ulcerative colitis: A pilot study. J Crohns Colitis 2019;13(10):1311–1317.
    1. Golinska E, Tomusiak A, Gosiewski T, et al. Virulence factors of Enterococcus strains isolated from patients with inflammatory bowel disease. World J Gastroenterol 2013;19(23):3562–72.
    1. Balish E, Warner T. Enterococcus faecalis induces inflammatory bowel disease in interleukin-10 knockout mice. Am J Pathol 2002;160(6):2253–7.
    1. Seishima J, Iida N, Kitamura K, et al. Gut-derived Enterococcus faecium from ulcerative colitis patients promotes colitis in a genetically susceptible mouse host. Genome Biol 2019;20(1):252.
    1. Kellingray L, Gall GL, Defernez M, et al. Microbial taxonomic and metabolic alterations during faecal microbiota transplantation to treat Clostridium difficile infection. J Infect 2018;77(2):107–18.
    1. Hyams JS, Davis Thomas S, Gotman N, et al. Clinical and biological predictors of response to standardised paediatric colitis therapy (PROTECT): A multicentre inception cohort study. Lancet 2019;393(10182):1708–20.
    1. Cerqueira FM, Photenhauer AL, Pollet RM, et al. Starch digestion by gut bacteria: Crowdsourcing for carbs. Trends Microbiol 2020;28(2):95–108.
    1. Wopereis H, Sim K, Shaw A, et al. Intestinal microbiota in infants at high risk for allergy: Effects of prebiotics and role in eczema development. J Allergy Clin Immunol 2018;141(4):1334–42.e1335.
    1. Goh YJ, Barrangou R. Harnessing CRISPR-Cas systems for precision engineering of designer probiotic lactobacilli. Curr Opin Biotechnol 2019;56:163–71.
    1. Yilmaz B, Juillerat P, Øyås O, et al. Microbial network disturbances in relapsing refractory Crohn's disease. Nat Med 2019;25(2):323–336.

Source: PubMed

3
Předplatit