Abstract
Inflammatory bowel diseases (IBD), including Crohn’s disease, ulcerative colitis and pouchitis, are chronic, relapsing intestinal inflammatory disorders mediated by dysregulated immune responses to resident microbiota. Current standard therapies that block immune activation with oral or biologic agent immunosuppression or surgical resection are generally effective, but each therapy induces a sustained remission in only a minority of patients. Furthermore, these approaches can have severe adverse events. Recent compelling evidence of a role of imbalanced of microbiota (dysbiosis) driving immune dysfunction and inflammation in IBD supports the therapeutic rationale for manipulating the dysbiotic microbiota. Traditional approaches using currently available antibiotics, probiotics, prebiotics, and synbiotics have not produced optimal results, but promising outcomes with fecal microbiota transplant (FMT) provide a proof of principle for targeting the resident microbiota. Rationally designed oral biotherapeutic products (LBP) composed of mixtures of protective commensal bacterial strains demonstrate impressive preclinical results. Resident microbial-based and microbial-targeted therapies are currently being studied with increasing intensity for IBD primary therapy with favorable early results. This review presents current evidence and therapeutic mechanisms of microbiota modulation, emphasizing clinical studies, and outlines prospects for future IBD treatment using new approaches, such as LBPs, bacteriophages, bacterial function-editing substrates and engineered bacteria. We believe that the optimal clinical use of microbial manipulation may be as adjuvants to immunosuppressive for accelerated and improved induction of deep remission and as potential safer solo approaches to sustained remission using personalized regimens based on an individual patient’s microbial profile.
Keywords: pouchitis, fecal microbiota transplantation, probiotics, prebiotics, synbiotics, diet, live biotherapeutic products, dysbiosis, microbiota
Introduction
Inflammatory bowel diseases (IBD), including Crohn's disease (CD), ulcerative colitis (UC) and pouchitis, are chronic intestinal inflammatory disorders characterized by dysregulated immune responses to enteric resident microbiota in genetically susceptible hosts [1-3]. Based on the requirement of microbiota colonization to develop colitis in germ-free (GF) susceptible rodents [4-7], gut microbiota play a crucial role in the pathogenesis of IBD [1,3,8]. Microbiota include potentially pathogenic microbes driving inflammation (pathobionts), as well as, potentially beneficial microbes inducing protective immune responses (commensals) [1,9,10]. However, most IBD patients exhibit unbalanced gut microbiota profiles (dysbiosis), with expanded potentially pathogenic Proteobacteria (especially Enterobacteriaceae that include E. coli and Klebsiella), Fusobacteria, Ruminococcus gnavus and Candida tropicalis [11] and reduced potentially protective Firmicutes (especially Faecalibacterium prausnitzii, Ruminococci and Clostridium clusters IV and XIVa) [12,13] (Table 1). The immunologic consequences of dysbiosis and its causal role in experimental colitis provide a strong rationale for therapeutically modifying the enteric microbiota in patients with IBD [1,3,8,14]. Current primary therapies in IBD, such as corticosteroids, methotrexate, 5-aminosalicylic acid (5-ASA), JAK inhibitors, anti-tumor necrosis factor (TNF)-α, anti-interleukin (IL)-12p40 antibody, and anti-integrin antibodies and surgical resection etc, mostly target effector immune responses [15-17]. These therapies can induce remission in many IBD patients, but can have severe adverse events with impaired quality of life (QOL). Microbiota-based therapies, including fecal microbiota transplant (FMT), probiotics and prebiotics, are suggested to be safe and can potentially correct the dysbiosis driving the dysregulated immune response [1,3,18]. Recent success of FMT in recurrent or refractory Clostridium difficile (rCDI) [19] achieved a major breakthrough in microbial-based therapy, which is being studied with increasing intensity as IBD primary therapy with favorable reported results [20]. In response to this trend, the United States Food and Drug Administration (FDA) created a new category, live biotherapeutic products (LBPs), for “live organisms, such as bacteria, which are applicable to the prevention, treatment, or cure of a disease or condition of human beings” and issued a guidance for clinical trials [21]. This review provides an overview of current microbial-based and microbiota-targeted therapies (Tables 2-5) and prospects for future treatments in IBD (Table 6) (Figure 1).
Table 1.
Representative dysbiotic microbiota in IBD
Ulcerative colitis | Pouchitis | Crohn's disease |
---|---|---|
Bacterial diversity ↓ | Bacterial diversity ↓↑ | Bacterial diversity ↓ |
Proteobacteria ↑ | Proteobacteria | Proteobacteria ↑ |
Enterobacteriaceae | Enterobacteriaceae | Enterobacteriaceae ↑ |
E. coli ↑ | E. coli ↑ | Adherent-invasive E. coli ↑ |
K. pneumoniae ↑ | ||
Pasteurellaceae ↑ | ||
Haemophilus ↑ | ||
Neisseriaceae ↑ | ||
Fusobacteria ↑ | Fusobacteria | Fusobacteria ↑ |
F. varium ↑ | Fusobacterium ↑ | Fusobacteriaceae ↑ |
Bacteroidetes ↑↓ | Bacteroidetes | Bacteroidetes |
Bacteroides ↑ | Bacteroides ↑ | Bacteroiales ↓↑ |
B. vulgatus ↑ | ||
Firmicutes ↓ | Firmicutes | Firmicutes ↓ |
Clostridiales ↓ | Clostridiales ↓ | |
clusters IV, XIVa, XVIII ↓ | clusters IV, XIVa ↓ | |
F. prausnitzii ↓ | F. prausnitzii ↓ | |
E. rectale ↓ | E. rectale ↓ | E. rectale ↓ |
Ruminococcaceae | Ruminococcaceae | Ruminococcaceae ↓ |
R. gnavus ↑ | R. gnavus ↑ | R. gnavus ↑ |
clusters I, II, IX, XI ↑ | ||
C. perfringens ↑ | C. perfringens ↑ | |
Peptostreptococci ↑ | Veillonellaceae ↑ | |
Lachnospiraceae | Lachnospiraceae ↓ | |
Roseburia hominis ↓ | Erysipelotrichaceae ↓ | |
Bacilli ↑ | Bacilli | Bacilli ↑ |
E. faecalis ↑ | Lactobacilli ↓ | Lactobacillus ↓↑ |
Actinobacteria | Actinobacteria | |
Bifidobacteria ↓ | Bifidobacteriaceae ↓ | |
Viral diversity ↑ | Viral diversity ↑ | |
Caudoviales bacteriopahge ↑ | ||
Fungal diversity | Fungal diversity | |
Candida ↑ | Candida ↑ |
Table 2.
Antibiotics for IBD (Randomized trials)
Author, year | Disease activity |
Case /Control |
Base therapy | Agent (s) | Route | Therapy duration |
Outcomes, therapy vs control | PMID |
---|---|---|---|---|---|---|---|---|
Ulcerative colitis | ||||||||
Dickinson, 1985 | active | 18/15 | Steroid | Vancomycin | oral | 7 d | NS | 3910524 |
Chapman, 1986 | active | 19/20 | Steroid | Metronidazole | i.v. | 5 d | NS | 3536677 |
Burke, 1990 | active | 42/42 | Steroid | Tobramycin | oral | 7 d | Improve clinical and histological score, 74% vs 43% | 2104079 |
Mantzaris, 1994 | active | 19/20 | Steroid | Metronidazole + Tobramycin | i.v. | 10 d | NS | 8273796 |
Mantzaris, 1997 | active | 34/36 | Olsalazine, steroid (oral/enema) | Ciprofloxacin | oral | 14 d | NS | 9068468 |
Casellas, 1998 | active | 19/11 | Steroid | Amoxicillin | oral | 5 d | Improve luminal IL-8 and other inflammatory mediaters | 9552221 |
Turunen, 1998 | active | 38/45 | Steroid, 5-ASA, sulfa | Ciprofloxacin | oral | 6 m | Improve endoscopic and histologic scores at 3 mo, but not at 6, 12mo. | 9797360 |
Gionchetti, 1999 | active | 14/12 | Steroid | Rifaximin | oral | 10 d | Improve clinical, endoscopic scores, 64% vs 42% | 10389700 |
Mantzaris, 2001 | active | 29/26 | Steroid | Ciprofloxacin | i.v. | 10 d | NS | 11521989 |
Ohkusa, 2005 | active | 10/10 | Steroid, 5-ASA, probiotics | Amoxicillin + Tetracycline + Metronidazole | oral | 14 d | Improve clinical, endoscopic, and histological scores | 16334443 |
Ohkusa, 2010 | active | 105/105 | Steroid, 5-ASA, immunosuppressant, sulfa | Amoxicillin + Tetracycline + Metronidazole | oral | 14 d | Improve clinical, endoscopic, and histological scores | 20216533 |
Turner, 2018 | active | 16/12 | Steroid (i.v.) | Amoxicillin + Vancomycin + Metronidazole + Doxycyclin/Ciprofloxacin | 5 d | Improve clinical score | Abst (a) | |
Pouchitis | ||||||||
Madden, 1994 | active | 11/11 | Metronidazole | oral | 2 w | Improve stool frequency (73% vs 0%) | 8200250 | |
Shen, 2001 | active | 7/9 | Ciprofloxacin vs Metronidazole | oral | 2 w | Both improve clinical and endoscopic scores. Efficacy: cipro>metro | 11720319 | |
Isaacs, 2007 | active | 8/9 | 5-ASA, probiotics, NSAIDs | Rifaximin | oral | 4 w | Improve remission rate (25% vs 0%) | 17567869 |
Crohn’s disease | ||||||||
Blichfeldt, 1978 | active | 20/20 | Sulfa, steroid | Metronidazole | oral | 2 m | NS | 345410 |
Kelleher, 1982 | inactive | 10/10 | Clofazimine | oral | 6 m | Improve relapse rate (0% vs 30%) | Abst (b) | |
Ambrose, 1985 | active | 18/16/21/17 | Steroid, 5-ASA, AZA | Metronidazole vs Cotrimoxazole vs Combined vs placebo | oral | 4 w | At 2w, improve symptoms. At 4w, NS | 3882364 |
Dickinson, 1985 | active | 4/3 | Steroid | Vancomycin | oral | 7 d | NS | 3910524 |
Sutherland, 1991 | active | 33/30/36 | Metronidazole low dose vs high dose vs placebo | oral | 16 w | Improve clinical score Efficacy: high dose > low dose | 1916494 | |
Afdhal, 1991 | active | 25/24 | Steroid | Clofazimine | oral | 3 m | NS | 2007362 |
Afdhal, 1991 | inactive | 16/12 | Clofazimine | oral | 12 m | NS (reduce clinical score) | 2007362 | |
Prantera, 1994 | inactive | 19/17 | Steroid tapering | Clofazimine + Rifampin + Ethambutol + Dapsone | oral | 9 m | Improve relapse rate | 8147352 |
Graham, 1995 | active | 7/8 | Clarithromycin | oral | 3 m | Improve remission rate (71% vs 13%) | Abst (c) | |
Goodgame, 2001 | active | 9/9 | Ethambutol + Clarithromycin | oral | 3 m | NS | 11736715 | |
Arnold, 2002 | active | 25/12 | Ciprofloxacin | oral | 1 m | Improve clinical score | 11837933 | |
Steinhart, 2002 | active | 66/64 | Ciprofloxacin + Metronidazole | oral | 8 w | NS | 12105831 | |
West, 2004 | fistula | 11/13 | Anti-TNF-α | Ciprofloxacin | oral | 12 w | NS (improve fistula 73% vs 39%) | 15606395 |
Rutgeerts, 2005 | inactive | 38/40 | Steroid | Ornidazole | oral | 54 w | Improve recurrence rate (8% vs 38%) | 15825069 |
Prantera, 2006 | active | 25/27/27 | Immunosuppressant, 5-ASA | Rifaximin o.d. vs b.d vs placebo | oral | 12 w | NS (improve remission rate) Efficacy: o.d.<b.d. | 16611272 |
Selby, 2007 | active | 102/111 | Steroid | Clarithromycin + Rifabutin +Clofazimine | oral | 16 w | Improve remission rate (66% vs 50%) | 17570206 |
Leiper, 2008 | active | 12/10 | Steroid, 5-ASA, AZA | Clarithromycin | oral | 3 m | At 1 m, improve clinical scores At 3m, NS | 18315579 |
Thia, 2009 | fistula | 9/2/7 | Immunosuppressant, steroid | Ciprofloxacin vs Metronidazole vs placebo | oral | 10 w | NS (remission: 30% vs 0% vs 13%) | 18668682 |
Maeda, 2010 | fistula | 33/41 | Steroid, 5-ASA, immunosuppressant, antibiotics, anti-TNF-α | Metronidazole | oral | 4 w | Improve clinical score and perianal discharge and pain | 20632322 |
Prantera, 2012 | active | 104/98/99/101 | Steroid, 5-ASA, immunosuppressant, antibiotics, anti-TNF-α | Rifaximin-EIR low vs mid vs high dose vs placebo | oral | 12 w | Improve remission rate by mid-dose 800mg/d (62% vs 43% placebo) | 22155172 |
Herfarth, 2013 | inactive | 17/16 | Steroid, 5-ASA, immunosuppressant | Ciprofroxacin | oral | 6 m | NS | 23511031 |
Jigaranu, 2014 | active | 83/83 | 5-ASA, AZA, anti-TNF-α | Rifaximin | oral | 12 w | Improve remission rate (100% vs 84%) | 24969283 |
Levine, 2019 | active | 35/38 child | Azithromycin+Metronidazole vs Metronidazole | oral | 8 w | Improve remission rate (66% vs 39%) | 29420227 |
IBD: inflammatory bowel disease, PMID: PubMed identifier, NS: no statistically significant difference in disease activity, 5-ASA: 5-aminosalicylic acid, sulfa: salazosulfapyridine, AZA; azathioprine, NSAID: nonsteroidal antiinflammator drug, o.d.: once a day, b.d. twice a day, EIR: extended intestinal release. Abst (a): JCC 2018,12,S366, Abst (b): Gut 1982,23,A449, Abst (c): Gastroenterology 1995,108,A826
Table 5.
FMT for IBD (RCTs and case serieses)
Author, year | Design | Disease activity |
Case /Ctrl |
FMT route and others | Outcomes, thrapy vs control | PMID |
---|---|---|---|---|---|---|
Ulcerative colitis | ||||||
Borody, 2003 | Cases | active | 6/0 | Enema, pre-antibiotics | Clinical response: 100% at 4 m | 12811208 |
Kunde, 2013 | Cases | active | 10/0 child | Enema | Clinical response: 78% at 1 w Clinical remission: 33% at 1 w | 23542823 |
Moayyedi, 2015 | RCT | active | 36/34 | Enema, multiple | Improve remission rate (24% vs 5%) at 7 w | 25857665 |
Rossen, 2015 | RCT | active | 23/25 | Nasoduodenal, multiple | NS (41% vs 25%, in per-protocol population) at 12 w | 25836986 |
Damman, 2015 | Cases | active | 7/0 | Colonoscopy | Clinical remission: 14% from 1 m until 3 m | 26288277 |
Suskind, 2015 | Cases | active | 4/0 child | Nasogastric | NS | 25647155 |
Wei, 2015 | Cases | active | 11/0 | Colonoscopy or nasojejunal | Improve Mayo score and IBDQ score at 4 w | 26146498 |
Wei, 2016 | Cases | active | 10/10 | FMT vs FMT +pectin (FMTP) | Mayo scores were significantly lower in the FMTP group than in the FMT at 4 w and 12 w | 27809778 |
Vermeire, 2016 | Cases | active | 8/0 | Colonoscopy or nasojejunal | Endoscopic remission: 25% at 8 w | 26519463 |
Goyal, 2016 | Cases | active | 7/0 | Colonoscopy or nasojejunal | Clinical response: 16% at 180 d | Abst (a) |
Paramsothy, 2017 | RCT | active | 42/43 | Single colonoscopy and multiple enema, pooled FMT | Improve remission rate (27% vs 8%) at 8 w | 28214091 |
Jacob, 2017 | Cases | active | 20/0 | Colonoscopy | Clinical response: 35% Clinical remission: 15% Mucosal healing 10% at 4 w | 28445246 |
Uygun, 2017 | Cases | active | 30/0 | Colonoscopy | Clinical response: 70% Clinical&Endoscopic remission: 43% at 12 w | 28422836 |
Ishikawa, 2017 | Cases | active | 17/19 | Colonoscopy, pre-AFM antibiotics + FMT vs AFM alone | Pre-AFM contributed to Bacteroidetes recovery associated with UC activity at 4 w | 27893543 |
Nishida, 2017 | Cases | active | 41/0 | Colonoscopy | Clinical response: 27% at 8 w | 27730312 |
Goyal, 2018 | Cases | active | 14/0 child | Single upper and lower endoscopy | Clinical response: 50% Clinical remission: 0% at 6 m | 29361092 |
Costello, 2019 | RCT | active | 38/35 | Enema and colonoscopy, anaerobically prepared pooled FMT | Improve remission rate (32% vs 9%) at w8 42% of responder keep remission at 12 m | 30644982 |
Pouchitis | ||||||
Landy, 2015 | Cases | active | 8/0 | Nasogastric | Clinical response: 25% Clinical remission: 0% | 26264409 |
El-Nachef, 2016 | Cases | active | 7/0 | Pouchoscopy | Improve symptoms (71%) | Abst (b) |
Stallmach, 2016 | Cases | active | 5/0 | multiple | Clinical response: 100% Clinical remission: 4/5 | 27018122 |
Herfarth, 2019 | RCT -> open | active | 6/0 | Single colonoscopy and daily oral | Clinical remission: 17% at 2 w | 31172007 |
Selvig, 2019 | Cases | active | 19/0 | Single colonoscopy | NS on clinical activity Improve bowel movement frequency and abd pain, microbial diversity at 4 w | 31302808 |
Crohn’s disease | ||||||
Suskind, 2015 | Cases | active | 9/0 child | Nasogastric | Remission: 78% at 2 w | 25647155 |
Cui, 2015 | Cases | active | child 30/0 | Gastroscope+mid-gut tube | Response (87%), remission (77%) | 25168749 |
Wei, 2015 | Cases | active | 3/0 | Conoloscopy or nasogastric | NS (clinical activity) Improve IBDQ score at 4 w | 26146498 |
Vanghn, 2016 | Cases | active | 19/0 | Colonoscopy | Clinical response (58%) Increase in colonic regulatory T cells at 12 w | 27542133 |
Vermeire, 2016 | Cases | active | 6/0 | Colonoscopy or nasojejunal | NS | 26519463 |
Goyal, 2016 | Cases | active | 4/0 child | Nasojejunal and colonoscopy | Response: 75% at 180 d | Abst (a) |
He, 2017 | Cases | active | 25/0 | Colonoscopy + tube | Response: 68%, Remission: 52% reduce inflammatory mass | 28684845 |
Goyal, 2018 | Cases | active | 7/0 child | Single upper and lower endoscopy | Response: 71%, remission: 29% at 6m | 29361092 |
Li, 2019 | Cases | active | 165/0 | Mid-gut/nasal-jejunal transendoscopic enteral tubing | Analysis of timing for sencond FMT Second<4 m is better | 30357440 |
IBD: inflammatory bowel disease, FMT, fecal microbiota transplantation, RCT: randomized controlled trial, PMID: PubMed identifier, NS: no statistically significant difference in disease activity, IBDQ: inflammatory bowel disease questionnaire, AFM: amoxicillin, fosfomycin, metronidazole. Abst (a): JPGN 2016, 63, S212, Abst (b): Gastroenterology 2016, 150, S544.
Table 6.
Ongoing microbial-based and microbial-targeted clinical trials in IBD (Randomized trials)
NCT number | Country | Start year |
Agent (s) | Target disease (s) |
Others | |
---|---|---|---|---|---|---|
Antibiotics | ||||||
NCT00061282 | US | 2002 | Clotrimazole | Pouchitis | ||
NCT00603616 | US | 2008 | Rifaximin | CD (active) | Induction of remission | |
NCT01951326 | US | 2013 | Anti-Mycobacterium | CD (active) | Induction of remission | |
NCT02033408 | Canada, Italy, Finland, Israel, Poland, Spain | 2014 | Antibiotics and FMT | UC, CD | ||
NCT01783106 | UK | 2014 | Ciprofloxacin, Doxycycline, Hydroxychloroquine, Budesonide | CD | ||
NCT02620007 | France | 2015 | Ciprofloxacin Rifaximin | CD (active) | Induction of remission AIEC targeted | |
NCT03537157 | Italy | 2017 | Rifaximin | CD (post-ope) | ||
NCT03476317 NCT02765256 |
US | 2018 | Vancomycin, Neomycin, Ciprofloxacin Polyethylene Glycol, Fluconazole | CD (refractory) | ||
NCT03221166 | Italy | 2018 | Thalidomide, Infliximab | CD (new onset) | ||
NCT04082559 | Israel | 2019 | Personalized antibiotics (Ciprofoxacin, Doxycycline) + diet (SCD, MSD) | Pouchitis, CD | ||
NCT03794765 | India | 2019 | Adjuvant ceftriaxone, metronidazole | UC (active) | ||
NCT03986996 | Israel | 2019 | Amoxicillin+Tetracycline+Metronidazole vs Amoxicillin+Tetracycline | UC | Induction of remission | |
LBP (probiotics) | ||||||
NCT03266484 | US | 2017 | Mixture (8 different bacterial strains) | CD, UC (inactive) | ||
NCT03415711 | Italy | 2017 | VSL#3 | UC (active) | Induction of clinical and endoscopic remission | |
NCT04102852 | Italy | 2019 | Lactobacillus rhamnosus GG | UC (active) | Induction of clinical remission | |
Prebiotics | ||||||
NCT02825914 | Denmark | 2016 | Casein glycomacropepptide | UC | ||
NCT03500653 | Israel | 2018 | Curcumin | IBD | ||
NCT03653481 | US | 2018 | Oligofructoseenriched Inulin | IBD | ||
NCT03847467 | US | 2019 | Fucosyllactose | CD, UC (child, adult) | Patients receiving stable maintenance anti-TNF therapy | |
NCT03998488 | US | 2019 | FMT + Psyllium | UC (active) | ||
NCT02277223 | Israel | 2019 | Curcumin | UC (pediatric) | ||
NCT02683759 | India | 2019 | Curcumin | UC | ||
Diet | ||||||
NCT04046913 | UK | 2013 | Low additive diet | CD | ||
NCT02472457 | US | 2015 | Crohn’s Disease Exclusion Diet | CD | ||
NCT02796339 | Greece | 2016 | Mastiha (Pistacia lentiscus) | IBD | ||
NCT02734589 | France, Israel, Italy | 2017 | Novel diet for the donor + FMT | UC | ||
NCT03000101 | Italy | 2017 | Pomegranate | CD, UC | Protocol in PMID 31171016 | |
NCT03012542 | US | 2017 | carbohydrates- or fiber- controled diet | CD | ||
NCT03053713 | Canada | 2017 | MSD | UC | ||
NCT03058679 | US | 2017 | MSD, SCD | CD | ||
NCT03301311 | US | 2018 | SCD, modified SCD | CD, UC (inactive) | ||
NCT02843100 | Canada, Ireland, Israel, Spain | 2018 | Modified Exclusive Enteral Nutrition, Crohn’s Disease Exclusion Diet, Partial Enteral Nutrition, Standard Exclusive Enteral Nutrition | CD | ||
NCT04143633 | Mexico | 2018 | Low FOMAP diet | UC | ||
NCT02201758 | Canada | 2018 | Flaxseed lignanenriched complex | UC | ||
NCT04147585 | US | 2019 | Intermittent reduced calorie diet | CD | ||
NCT03615690 | US | 2019 | Fasting mimicking diet | UC | ||
NCT04082559 | Israel | 2019 | Personalized antibiotics (Ciprofoxacin, Doxycycline) + diet (SCD, MSD) | Pouchitis | ||
NCT03850600 | US | 2019 | Diet intervention (no details) | CD (pregnancy) | ||
NCT04014517 | Italy | 2019 | Nestle IMPACT | CD | Prevention of reccurence after surgery | |
NCT03980405 | Israel | 2019 | Ulcerative collitis diet | UC | Adjuvant to 5-ASA | |
NCT04018040 | Australia | 2019 | Lacto-ovo vegetarian diet | UC | ||
Synbiotics | ||||||
NCT02865707 | Canada | 2016 | Synergy-1 | UC | Prevention of relapse | |
FMT | ||||||
NCT01790061 | China | 2012 | Standardized FMT vs traditional FMT | UC | ||
NCT02636517 | US | 2015 | FMT | CD, UC | ||
NCT02390726 | US | 2015 | FMT | UC (active) | ||
NCT03561532 | Finland | 2016 | FMT | UC | ||
NCT02291523 | US | 2016 | FMT | UC (pediatric) | ||
NCT02606032 | Canada | 2016 | 4 arms of pre-antibiotics + FMT | UC (active) | ||
NCT03078803 | Canada | 2017 | FMT via colonoscope or oral FMT | CD | ||
NCT03110289 | Belgium | 2017 | FMT superdonor vs FMT autologous | UC | Superdonor is based on abundance of taxa of the investigators’ interest | |
NCT03273465 | Israel | 2017 | FMT | UC (active) | ||
NCT03104036 | Czechia | 2017 | FMT vs 5-ASA enema | UC (active) | ||
NCT03006809 | US | 2017 | +/−Pre-Ax + low/high FMT | UC (active) | ||
NCT03378167 | Canada | 2018 | FMT oral | CD (pediatric) | ||
NCT03582969 | Israel | 2018 | FMT | UC (pediatric) | ||
NCT03378921 | Finland | 2018 | FMT | UC, Pouchitis | ||
NCT03716388 | India | 2018 | FMT vs FMT+5-ASA vs 5-ASA | UC (active) | ||
NCT04100291 | Denmark | 2019 | FMT | UC, Pouchitis | ||
NCT03843385 | Germany | 2019 | FMT-filtrated vs FMT vs Plaebo | UC | ||
NCT03829475 | US | 2019 | FMT | +/− Bezlotoxumab | IBD | |
NCT03998488 | US | 2019 | FMT+Psyllium | UC (active) | ||
NCT03483246 | France | 2019 | FMT | UC | ||
NCT03747718 | US | 2019 | FMT single vs maintainance | CD | ||
NCT04034758 | China | 2019 | Heterologous FMT | UC (active, adult, older adult) | ||
NCT03804931 | China | 2019 | 5-ASA/steroid +/− FMT | UC | ||
NCT03948919 | US | 2019 | Low sulfur FMT | UC (active) | ||
NCT03917095 | China | 2019 | Colonic transendoscopic enteral tubing | UC (active) | ||
Bacteriophage | ||||||
NCT03808103 | US | 2019 | EcoActive | CD |
Information from ClinicalTrials.gov., IBD: inflammatory bowel disease, CD: Crohn’s disease, UC: ulcerative colitis, FMT: fecal microbiota transplant, 5-ASA: 5-aminosalicylic acid, PMID: PubMed identifier, SCD: specific carbohydrate diet, MSD: mediterranean style diet, FOMAP:low fermentable oligosaccharide, disaccharide, monosaccharide, and polyol , AIEC: adherent-invasive Escherichia coli
Figure 1.
Graphic overview. The concept of manipulating microbiota to correct dysbiosis is a relatively new approach to treating inflammatory bowel disease (IBD). This review updates the status of current microbial-based and microbial-targeted therapies and prospects for future treatments in IBD. Tx: therapy, 5-ASA: 5-aminosalicylic acid, MTX: methotrexate, JAK: Janus kinase, IL: interleukin, TNF: tumor necrosis factor, SCFA: short chain fatty acid, PXR: pregnane X receptor, PPAR: peroxisome proliferator activated receptor, Treg: regulatory T cell, GOS: galacto-oligosaccharide, FOS: fructo-oligosaccharide, GBF: germinated barley foodstuff, OI: oligofructose-enriched inulin, BGS: bifidogenic growth stimulator, FODMAP: fermentable oligosaccharide, disaccharide, monosaccharide and polyol, FMT: fecal microbiota transplant, LBP: live biotherapeutic product, PolyP: polyphosphate, KFXL: Kangfuxin liquid, path: pathogenic, AIEC: adherent-invasive Escherichia coli, Images of antibiotics and prebiotics are adopted from KEGG. Image of prebiotic diet is adopted from Monash University (https://www.monashfodmap.com/blog/a-low-fodmap-mediterranean-style-diet/). Red: aggressive microbial species and cells, blue: protective microbial species and cells.
Microbiota in IBD: The rationale for therapeutic microbial manipulation
In general, ‘microbiota (or microbes)’ includes bacteria, fungi and viruses (mostly bacteriophages) while ‘microbiome’ refers to microbiota and their genes and metabolites [1,22,23]. The huge number of microbial cells in the distal intestine (1014 bacteria/g), species (approximately equal to human cells), genes (outnumber human genes by 100-fold), bacteriophages (outnumber bacteria by 10-fold) and their weight (1−2 kg) [13,22,24,25], are considered a ‘superorganism’ and ‘forgotten organ’ [26,27]. The colonic lumen contains the densest bacteria concentration in the human body (1011–1014 bacteria/g), followed by oral (108/g), ileum (107−108/g), jejunum (104/g), duodenum (103/g) and stomach (101/g) [22,23,28]. An individual’s enteric bacterial composition varies greatly and each individual harbors 100−150 diverse intestinal species [22,29]. This diversity allows humans to obtain a variety of benefits, such as digesting various foods (especially fiber), producing vitamins and other protective metabolites, activating homeostatic gut and systemic immune responses and preventing colonization by exogenous pathogens [1]. However, the diversity of bacteria in IBD patients is significantly decreased [12,13,30,31], whereas fungi and bacteriophages are expanded [11,31-33]. Furthermore, the composition and function of enteric microbiota in IBD patients is frequently disrupted, characterized by expanded potentially pathogenic microbes and reduced protective microbes producing short chain fatty acids (SCFAs) [12,13,34-39]. This microbial imbalance, termed as ‘dysbiosis’, was first noted in the intestine of IBD patients [12,13,30,34-38], but recently oral dysbiosis is also reported [40-42], the latter indicating that dysbiosis can be independent of local inflammatory processes. Although more careful assessment is needed in various patient subsets using modern detection techniques, consistent changes occur in CD and UC (Table 1). The link between this dysbiosis and gut inflammation is supported by many experimental studies. CD-associated adherent-invasive Escherichia coli (AIEC) invade epithelial cells and replicate within macrophages and can cause chronic experimental colitis [43,44]. Another Enterobacteriaceae, Klebsiella pneumoniae isolated from a CD patient, induces experimental colitis with high Th1 response compared to other control strains and species [42]. Fusobacterium varium strains from UC patients invade epithelial cells compared to strains from healthy controls and induce experimental colitis [45]. Alternatively, certain Clostridium species and F. prausnitzii are putative anti-inflammatory microbes. Clostridia are dominant intestinal microbes, accounting for over 60% of mucosa-associated bacteria [46]. A subset of resident Clostridium species produce SCFAs and can induce colonic regulatory T cells (Tregs) or IL-10-producing B cells and macrophages to protect against experimental colitis [47-50] with reduction in the abundance of Enterobacteriaceae [50]. F. prausnitzii another major SCFA producer, induces IL-10 production by human and murine dendritic cells [51]. Indeed, IBD-derived fecal bacteria stool did not induce colonic Treg in GF mice [9]. Interestingly, most expanded bacteria in IBD are aerotolerant species (aerobes or facultative anaerobes), such as E. coli, F. varium, Haemophilus, Enterococcus faecalis and Neisseriaceae. In contrast, the majority of reduced bacteria are obligate anaerobes, such as Clostridium clusters IV, XIVa, XVIII and F. prausnitzii. This trend gives rise to the ‘oxygen hypothesis’ wherein disruption in anaerobiosis indicates to a role for oxygen in intestinal dysbiosis [52]. Recent studies support this hypothesis by showing that Clostridium strains inhibit dysbiotic Enterobacteriaceae expansion by reducing luminal oxygen via activation of epithelial PPAR-γ [53,54]. Notably, decreased PPAR-γ gene expression is associated with IBD pathogenesis [55]. Dysbiosis of fungi and bacteriophages in IBD were also noted recently [31-33] with interactions between C. tropicalis, E. coli and Serratia marcescens [11]. Further investigations may determine the significance of elevated anti-fungus antibody in many CD patients [56]. A causal association between dysbiosis and IBD is further supported by results from recent FMT trials, as a shift of the recipient’s dysbiotic microbiota towards the donor’s non-dysbiotic microbiota is associated with clinical response [57-60]. In addition to its causal role in driving inflammation, microbiota influence efficacy of certain immunomodulatory therapies, including anti-TNF-α [61,62], steroids [63] and PD-1-based treatments [64]. Understanding microbial dynamics is necessary for optimal current and future IBD therapies, particularly personalized management. Technologic developments and ongoing human microbiome projects have improved the culture of previously ‘unculturable’ human microbiota [65], access to more extensive multi-omics databases [66] and gene catalogues established by metagenomic sequencing [12,24].
Antibiotics
Antibiotics, antimicrobial substances active against bacteria, are widely used treat complications of IBD (bacteremia, abscess, opportunistic and surgical site infections) [1]. Antibiotics are also used as primary therapy for inducing or maintaining remission based on the hypothesis that certain bacteria cause IBD, the pathologic similarities between CD and Mycobacterium avium subspecies paratuberculosis infection and isolation of this organism in some CD patients [67]. IBD is considered to be caused by intricately intertwined gut microbiota, host genes, immune system and environmental factors rather than a specific infectious colitis [1,3]. However, as potential pathobionts are expanded in dysbiotic IBD intestines, targeted antibiotic therapy is a rational strategy. Unfortunately, most antibiotics decrease overall bacterial diversity and inhibit not only pathobionts but also beneficial bacteria, which can lead to overgrowth of pathogenic bacteria (C. difficile), fungi (candida) and bacteriophages [32]. Despite their inhibitory effects, some antibiotics increase protective bacteria [68-70] and modulate host immune functions [71]. This section updates clinical efficacy of antibiotics in IBD (Table 2) and their therapeutic mechanisms.
Ulcerative colitis
Two meta-analyses of antibiotic therapy for active UC demonstrated improved remission rates overall (64% vs 48% placebo) [72,73]. With a broad variety of different agents and protocols (vancomycin, metronidazole, tobramycin, ciprofloxacin, amoxicillin, ethambutol, tetracycline and rifamycin), it is difficult to choose optimal antibiotic agents. Of note, all randomized controlled trials (RCTs) using intravenous antibiotics failed to achieve therapeutic benefit over control treatment. In contrast, most oral antibiotics achieved clinical response except for 2 RCTs of ciprofloxacin. Two RCTs of a promising 2-week triple antibiotic primary therapy cocktail including oral Amoxicillin, Tetracycline and Metronidazole (ATM) showed significantly improved remission rates, clinical and endoscopic scores [74,75]. This regimen, designed based on susceptibility testing of F. varium [75], significantly reduced mucosal F. varium abundance in Japanese UC patients [76]. Further, a new RCT of the ATM triple cocktail versus AT cocktail, excluding metronidazole, has been initiated (ClinicalTrials.gov identifier: NCT03986996), given metronidazole’s potential negative effect on gut barrier function and poor patient acceptance. Only a few reports address the long-term outcomes of antibiotics: one trial reported that 7 days of oral tobramycin significantly improved remission rates at 1 week (74% vs 43% placebo) [77], but no statistical difference in relapse rates at 2-year follow-up (24% vs 12%) [78]. Another trial showed 6 month-oral ciprofloxacin improved endoscopic and histological appearances at early 3 month, but the benefit disappeared by 6 and 12 months [79]. In contrast, the ATM cocktail therapy demonstrated significantly higher remission rates and lower clinical and endoscopic scores at both intermediate (3–5 months) and long-term (12–14 months) follow-ups [74,75].
Pouchitis
All three RCT (including metronidazole, rifaximin and ciprofloxacin) showed therapeutic benefit, matching widespread clinical use. Three meta-analyses including RCTs and cohort studies support the favorable results [80-82]. A meta-analysis concludes that antibiotics and biologics (anti-TNF-α) are more beneficial for chronic refractory pouchitis than are corticosteroids, bismuth, elemental diet and tacrolimus [81]. Furthermore, ciprofloxacin is suggested to be more effective than metronidazole [82]. Ciprofloxacin reduced Clostridium perfringens and E. coli and did not affect abundance of anaerobic bacteria, while metronidazole reduced C. perfringens, but not E. coli, and reduced anaerobic bacteria in a cohort study [83], indicating that ciprofloxacin is more active against pathogenic species and less harmful to beneficial species. Major concerns with sustained or intermittent use of antibiotics include antibiotic resistance and side effects, such as tendon rupture with ciprofloxacin and peripheral neuropathy with metronidazole. Regarding antibiotic-dependency observed in many pouchitis cases, a recent clinical trial proposed a unique hypothesis that antibiotics enrich antibiotic-resistance in non-pathogenic species, which might prevent colonization with pathogenic species as long as antibiotics are used [84]. Based on this, a new RCT is currently underway that alternates antibiotics short-term with dietary interventions to support growth of beneficial species to avoid progression to antibiotic-dependent disease (NCT04082559).
Crohn’s disease
A meta-analysis of studies designed to maintain remission after surgical resections demonstrates significant benefit of antibiotics alone and as adjuvants to immunomodulators (azathioprine or 6-mercaptopurine) or anti-TNF-α therapies [85]. However, three meta-analyses suggest that the benefit of antibiotics is weak for overall treatment of CD [80,86,87]. However, anti-Mycobacterium agents (especially rifamycin-containing regimens), demonstrate some benefit for inducing remission [73,88] but do not induce a sustained remission to support clearance of a pathogen [89,90]. Long-term responses have been studied in a few reports, indicating that the protective effects of antibiotics wane over time; Aberra et al. suggested efficacy over 60 days [90,91]. Multiple additional RCTs of rifamycin for active CD (NCT02240108, NCT00603616, NCT02240121, NCT02620007) (Table 6) and prevention of postoperative CD recurrence (NCT03185624, NCT03185611) are ongoing or recently completed (NCT01951326). One of these trials specifically targets CD-associated E. coli-colonized patients (NCT02620007). Other agents, including ciprofloxacin and metronidazole alone or in combination, also show remission induction [72,73]. Given the fungal dysbiosis in CD, an ongoing RCT investigates whether addition antifungal fluconazole to an antibiotic cocktail can improve remission rates is underway (NCT02765256). For specific conditions such as anal or internal fistula, ciprofloxacin and metronidazole reduce drainage [73], improve symptoms and improve fistula closure rates [92-94]. For abscesses, the first choice is surgical treatment, but frequently emergency surgery can be avoided by antibiotics and percutaneous drainage [95]. Although meta-analyses of antibiotics in IBD are quite positive, clinical use of these agents are largely restricted to patients with active pouchitis and septic complications of CD. This disparity is in part due to publication bias that favors publication of positive results.
Therapeutic mechanisms
Several mechanisms mediate therapeutic actions of antibiotics. (1) Inhibiting pathobionts. Each antibiotic has a unique spectrum against bacteria and most antibiotics inhibit pathogenic species and decrease overall bacterial diversity. Long-term metronidazole eliminates Bacteroides, with bacterial concentrations correlated with disease activity [96]. Ciprofloxacin is effective against enteric pathogens and most Gram-negative Enterobacteriaceae. Although rifamycin does to alter the overall microbiota composition in IBD patients [71], it reduces bacterial attachment [71]. (2) Increasing beneficial bacteria. Despite many antibiotics reducing beneficial species, such as F. prausnitzii [84], some antibiotics can increase protective species. For example, rifamycin increases Lactobacillus [70], Bifidobacterium and F. prausnitzii [69]. (3) Modifying bacterial metabolites. Shifts in microbiota composition alter microbial metabolites, with increased SCFAs and other beneficial products [12,69] that correlate with clinical response in IBD patients [69,97]. (4) Immunomodulatory effect. Rifamycins, ciprofloxacin, metronidazole and macrolides have mucosal immunomodulatory effects [71,98-100]. Specifically, rifaximin is a gut-specific agonist of the human pregnane X receptor (PXR) that helps maintain mucosal homeostasis [71,99].
Clinical concerns
Safety:
In clinical trials IBD patients exhibited no increased risk of severe adverse events with antibiotics compared to placebo [86], but safety issues must be considered. Anti-Mycobacterium therapy has more frequent adverse events, such as rashes and skin pigmentation, but not increased withdrawal rate [88]. Long-term use of metronidazole can cause peripheral neuropathy [80,101].
Risk of resistance:
Probably due to higher antibiotic exposure, the prevalence rates of methicillin-resistant Staphylococcus aureus, vancomycin-resistant Enterococcus, and extended-spectrum beta-lactamases (ESBL)-producing E. coli are significantly higher among IBD patients [102].
Risk of CDI:
Antibiotics increase CDI infection in IBD patients [103] through decreased lactate-producing bacteria numbers and increased succinic acid [104] although others reported rare CDI in CD patients [105]. Antibiotic-resistant probiotics may prevent CDI after antibiotic therapy [106].
Effective protocols:
While oral antibiotics appear to be effective as adjunctive therapy for IBD flares based on their direct effects on luminal bacteria and mucosal immune function, the benefits of intravenous antibiotics is not proven [107]. For fulminant colitis, such as toxic megacolon at risk for severe bacteremia, especially when receiving corticosteroids, intravenous broad-spectrum antibiotics appear to be reasonable [108]. The most effective therapy duration remains unclear, but Ledder provides recommendations [109]. The short-term benefit (induction of remission) of antibiotics is promising, while the long-term benefits (maintenance) appear low with increased toxicity or antibiotic resistant bacteria [78,90,91]. Sequential maintenance approaches, such as protective nutrients, probiotics or FMT, need to be considered after induction of remission with antibiotics.
Risk of dysbiosis:
Compelling epidemiologic evidence indicate that multiple early childhood exposures to antibiotics carry higher risk of developing CD [1]. It is unclear whether this risk is due to antibiotics themselves, an infection that required antibiotic use, or early IBD symptoms.
Conclusions
Despite the many different antibiotics, protocols and endpoint assessments in clinical trials with publication bias, oral antibiotics provide a promising primary or adjuvant therapy for inducing remission of IBD. Specific pathobiont-targeted strategy have recently emerged as an area of interest (F. varium, AIEC, C. perfringens), supporting future personalized antibiotic use. For active UC, the oral ATM cocktail is promising. For active pouchitis, ciprofloxacin > metronidazole are effective. For active CD, rifamycins are promising. Given the negative potential effects of long-term use such as host toxicity and antibiotic-resistance, short-term use followed by alternative maintenance therapies, such as probiotics, prebiotics, diet, standard immunotherapies, etc. should be considered.
Standard probiotics and LBPs using resident protective microbiota
Probiotics are living microorganisms such as bacteria or yeast with beneficial health effects [110], which included LBP [21]. Since Metchnikoff first published the concept of probiotics (Yogurt containing Lactobacillus bulgaricus) in 1907, many probiotic strains have been studied in clinical trials including IBD [108,110] (Table 3). Probiotic strains used in IBD trials have mostly belonged to two genera, Bifidobacterium and Lactobacillus, and isolated from limited sources (Yogurt, milk etc.) [108,110]. Recently, a variety of LBP candidates (Clostridium, Firmicutes spores, Bacteroides, Roseburia) isolated from healthy human microbiota have been investigated [1,3].
Table 3.
Probiotics for IBD (Randomized trials)
Author, year | Disease activity |
Case /Control |
Base Therapy | Agent (s) | Route | Therapy duration |
Outcomes, therapy vs control | PMID |
---|---|---|---|---|---|---|---|---|
Ulcerative colitis | ||||||||
Kruis, 1997 | remission | 50/53 | Sterpod, salicylates | Escherichia coli Nissle1917 vs 5-ASA | oral | 12 w | Equivalent relapse rate to 5-ASA | 9354192 |
Rembacken, 1999 | active | 57/59 | Steroid, pre-GEM | Escherichia coli Nissle1917 vs 5-ASA | oral | 3 m | Equivalent remission rate to 5-ASA | 10466665 |
Rembacken, 1999 | remission | 44/39 | Steroid | Escherichia coli Nissle1917 vs 5-ASA | oral | 12 m | Equivalent remission rate to 5-ASA | 10466665 |
Ishikawa, 2003 | remission | 11/10 | Steroid, sulfa | bifidobacteria-fermented milk | oral | 1 y | Improve relapse rate (27% vs 90%) | 12569115 |
Cui, 2004 | remission | 15/15 | BIFICO (Enterococci, Bifidobacteria, Lactobacilli) | oral | 8 w | Improve ralapse rate (20% vs 93%) and cytokine profile | 15133865 | |
Kruis, 2004 | remission | 162/165 | Escherichia coli Nissle1917 vs 5-ASA | oral | 12 m | Equivalent relapse rate to 5-ASA (36% vs 34%) | 15479682 | |
Tursi, 2004 | active | 30/30/30 | 5-ASA | Low balsalazide + VSL#3 vs med balsalazide vs mesalazine | oral | 8 w | Improve remission rate (80% vs 77% vs 53%) | 15507864 |
Kato, 2004 | active | 10/10 | Sulfa | bifidobacteria-fermented milk | oral | 12 w | Improve clinical, endoscopi and histological scores | 15569116 |
Shanahan, 2006 | Lactobacillus salivarius UCC118 | NS | Abst (a) | |||||
Zocco, 2006 | remission | 65/60/62 | LGG vs 5-ASA vs conbination | oral | 12 m | Improve relapse-free time (ralapse rate is similar) | 16696804 | |
Miele, 2009 | active, newly diagnosed child | 14/15 | 5-ASA, steroid | VSL#3 | oral | 1 y | Improve remission rate (93% vs 36%), relapse rate (21% vs 73%) and endoscopic and histological scores | 19174792 |
Fujimori, 2009 | inactive, mild | 40/40/40 | 5-ASA, steroid | Bifidobacterium longum vs Psyllium vs synbiotics | oral | 4 w | NS (synbiotics significantly improve remission rate) | 19201576 |
Sood, 2009 | active | 77/70 | 5-ASA | VSL#3 | oral | 12 w | Improve remission rate (43% vs 16%) and clinical scores | 19631292 |
Tursi, 2010 | active | 65/66 | 5-ASA, MNZ, AZA | VSL#3 | oral | 8 w | Improve clinical score (63% vs 41%) Remission rate (48% vs 32%, P=0.069) | 20517305 |
Matthes, 2010 | active | 24/23/23/20 | Steroid | Escherichia coli Nissle1917 | rectal | 2w- | Dose-dependent benefit | 20398311 |
Ng, 2010 | active | 14/14 | 5-ASA, AZA | VSL#3 | oral | 8 w | Improve DC cytokine profiles (↑IL-10, ↓IL-12p40) Clinical response (71% vs 36%, P=0.06) | 20155842 |
Wildt, 2011 | remission | 20/12 | 5-ASA, salazopyrine | Probio-Tec (Lactobacillus acidophilus La-5, Bifidobacterium animalis subsp. lactis BB-12) | oral | 52 w | NS (relapse rate: 75% vs 92%) | 21453880 |
D’Inca, 2011 | active | 8/11/7 | 5-ASA | Oral Lactobacillus casei DG + oral 5-ASA vs rectal Lactobacillus casei DG + oral 5-ASA vs oral 5-ASA | oral vs rectal | 8 w | TLR4, IL1b,microbiota: only rectal is effective. rectally administered L. casei DG, it modified colonic microbiota by increasing Lactobacillus spp. and reducing Enterobacteriaceae. | 20737210 |
Oliva, 2012 | active, child | 16/15 | 5-ASA | Lactobacillus reuteri ATCC55730 | rectal | 8 w | Improve clinical and endoscopic Mayo scores, histological scores and cytokine profiles | 22150569 |
Groeger, 2013 | active | 13/9 | Bifidobacterium infantis 35624 | oral | 6-8 w | Reduced CRP, TNF-a, IL-6 | 23842110 | |
Petersen, 2014 | active | 25/25/25/25 | 5-ASA, AZA, 6-MP, steroid | +/− pre-cipro (1w) +/− E.coli Nissle1917 (7w) | oral | 7 w | NS | 24972748 |
Yoshimatsu, 2015 | remission | 9/9 | 5-ASA, salazopyrin | Bio-three (Streptococcus faecalis T-110, Clostridium butyricum TO-A, Bificobacterium mesentericus TO-A) | oral | 12 m | Relapse: 0% vs 17%(3m), 9% vs 26%(6m), 22% vs 35%(9m) Remission: 70% vs 57% (12m) P=0.248 | 26019464 |
Tamaki, 2016 | active | 28/28 | 5-ASA, AZA, steroid | Bificobacterium longum BB536 | oral | 8 w | Improve clinical and endoscopic scores | 26418574 |
Palumbo, 2016 | active | 30/30 | 5-ASA | Lactobacillus salivarius, Lactobacillus acidophilus, Bifidobacterium bifidus BGN4 | oral | 24 m | Improve clinical and endoscopic scores | 27623957 |
Matsuoka, 2018 | remission | 98/97 | 5-ASA | bifidobacteria-fermented milk | oral | 48 w | NS | 29450747 |
Bharat, 2018 | active | 11/15/15/17 | SER-287 (cocktail of Firmicutes spores) placebo/placebo daily vs vanco/SER287 daily vs placebo/SER287 weekly vs vanco/SER-287 weekly | oral | 8 w | Improve remission rate (placebo/placebo daily: 0%, vanco/SER287 daily: 40%, placebo/SER287 weekly: 13.3%, vanco/SER-287 weekly: 17.7%) | Abst (b) | |
Pouchitis | ||||||||
Gionchetti, 2000 | remission | 20/20 | VSL#3 | oral | 9 m | Improve relapse rate (15% vs 100%) | 10930365 | |
Gionchetti, 2003 | inactive | 20/20 | VSL#3 | oral | 12 m | Improve onset rate (10% vs 40%) and IBDQ | 12730861 | |
Kuisma, 2003 | active | 10/10 | Lactobacillus rhamnosus GG | oral | 3 m | NS | 12622759 | |
Mimura, 2004 | inactive | 20/16 | VSL#3 | oral | 12 m | Improve relapse rate (15% vs 94%) and IBDQ | 14684584 | |
Tomasz, 2014 | active | 22/21 | Lactobacillus acidophilus, Lactobacillus delbrueckii supsp. bulgaricus, Bificobacterium bifidus | oral | 9m | Improve clinical and endoscopic scores and calprotectin | 24579075 | |
Yasueda, 2016 | inactive | 9/8 | Clostridium butyricum MIYAIRI | oral | 24 m | Improve relapse rate (11% vs 50%) and CRP | 26510664 | |
Bengtsson, 2016 | poor pouch function | 16/16 | Lactobacillus plantarum 299 + Bifidobacterium infantis Cure21 | oral | 21 d | NS | 27150635 | |
Crohn’s disease | ||||||||
Plein, 1993 | inactive | 10/7 | Saccharomyces boulardii | oral | 10 w | Improve CDAI (<150: 90% vs 14%) | 8465554 | |
Malchow, 1997 | inactive | 16/12 | Steroid | Escherichia coli Nissle1917 | oral | 3 m | NS | 9451682 |
Malchow, 1997 | active, inactive | 12/11 | Steroid | Escherichia coli Nissle1917 | oral | 12 m | NS (relapse rate: 33% vs 64%) | 9451682 |
Guslandi, 2000 | remission | 16/16 | 5-ASA | Saccharomyces boulardii | oral | 6 m | Improve relapse rate (6% vs 38%) | 10961730 |
Campieri, 2000 | remission | 20/20 | pre-rifaximin + VSL#3 vs 5-ASA | oral | 12 m | Improve endoscopic score (80% vs 60%) | Abst (c) | |
Prantera, 2002 | remission | 23/22 | Lactobacillus casei subsp. rhamnosus | oral | 12 m | NS | 12171964 | |
Schultz, 2004 | active | 5/6 | Steroid, antibiotics | Lactobacillus GG | oral | 6 m | NS | 15113451 |
Bousvaros, 2005 | remission | 39/36 | 5-ASA, AZA, 6-MP, steroid | Lactobacillus GG | oral | 2 y | NS | 16116318 |
Marteau, 2006 | inactive | 48/50 | Steroid | Lactobacillus johnsonii LA1 | oral | 6 m | NS | 16377775 |
Van Gossum, 2007 | inactive | 34/36 | Lactobacillus johnsonii LA1 | oral | 3 m | NS | 17206696 | |
Garcia Vilela, 2008 | inactive | 12/13 | 5-ASA, AZA, steroid, thalidomide. metronidazole | Saccharomyces boulardii | oral | 3 m | Improve intestinal permeability | 18584523 |
Bourreille, 2013 | inactive | 59/66 | AZA, 6-MP, steroid, MTX, anti-TNF-α | Saccharomyces boulardii | oral | 1 y | NS | 23466709 |
Fedorak, 2015 | inactive after surgery | 59/60 | VSL#3 | oral | 1 y | Improve mucosal inflammatory cytokine levels Reccurence of lesions (10% vs 27%, P=0.09) | 25460016 |
IBD: inflammatory bowel disease, PMID: PubMed identifier, NS: no statistically significant difference in disease activity, 5-ASA: 5-aminosalicylic acid, sulfa: salazosulfapyridine, AZA; azathioprine, 6-MP: 6-Mercaptopurine, MTX: methotrexate, DC: dendritic cell, TLR: toll-like receptor, IBDQ: inflammatory bowel disease questionnaire, CDAI: Crohn’s disease activity index,VSL#3: Lactobacillus casei, L. plantarum, L. acidophilus, L. delbrueckii subsp. bulgaricus, Bifidobacterium longum, B. breve, B. infantis, Streptococcus salivarius subsp. thermophilus, bifidobacteria-fermented milk containing Bifidobacterium breve Yakult, Bifidobacterium bifidum Yakult, Lactobacillus acidophillus YIT0168, ↑:increase, ↓:decrease. Abst (a): Gastroenterology 2006, 130, A44, Abst (b): Gastroenterology 2018, 154, S85, Abst (c): Gastroenterology 2000, 118, A781.
Ulcerative colitis
A meta-analysis of 18 RCTs in UC patients, including pediatric, demonstrates therapeutic benefit over placebo [111]. Also another meta-analysis including Chinese-based RCTs supports the use of adjuvant probiotics with 5-ASA in active UC [112]. Multiple strains have been investigated with favorable results (Table 3). A systematic sub-analysis suggests Bifidobacterium-containing probiotics significantly benefit active UC [113]. Because different strains have different metabolomic and immunomodulatory activities and provide complementary help, a cocktail of different strains may be more efficient than a single strain. Indeed, VSL#3, a cocktail of 8 strains, Lactobacillus casei, L. plantarum, L. acidophilus, L. delbrueckii subspecies bulgaricus, Bifidobacterium longum, B. breve, B. infantis and Streptococcus salivarius subspecies thermophilus, improved remission and relapse rates [111,112]. Additional RCTs of VSL#3 (NCT03415711) and L. rhamnosus (NCT04102852) in active UC are underway. More recently, a SERES Therapeutics (Boston, MA) cocktail of purified Firmicutes spore (SER-287) from feces of healthy screened donors was tested in a Phase 1B RCT in active UC [114-116]. Treatment arms included 6 days of vancomycin pretreatment followed by 8 weeks of SER-287 either daily or weekly or placebo pretreatment followed by weekly SER-287 vs placebo/placebo. Vancomycin improve engraftment of microbes from SER-287 [114] and improved remission rates (placebo/placebo daily: 0%, vanco/SER287 daily: 40%, placebo/SER287 weekly: 13.3%, vanco/SER-287 weekly: 17.7%) and endoscopic scores [115]. SER-287-treated remitters exhibited widespread transcriptional shifts from baseline, with by decreased expression of inflammatory genes and increased expression of homeostatic mediators [116]. These promising results led to a Phase 2B, 3-arm RCT in active UC (NCT03759041). Based on improved engraftment of SER-287 by vancomycin pretreatment, two patient groups receive different doses of SER-287, both following short courses of oral vancomycin. In pediatric UC, 2 RCTs demonstrated that oral VSL#3 [117] or rectal L. reuteri ATCC55730 [118] significantly improved clinical and endoscopic scores. Long-term follow-up data are limited, but a 2-year-follow-up showed promising results [119].
Pouchitis
Meta-analyses indicate that probiotics significantly induce remission and prevent relapse in pouchitis [80,120,121]. A RCT of oral C. butyricum MIYAIRI showed improved relapse rates (11% vs 50%) [122]. Gionchetti and colleagues reported strikingly decreased relapses (15% vs 100% placebo) in recurrent pouchitis after 9 months of oral VSL#3 therapy [123], but these positive results were not replicated in the USA [124]. In naive ileal pouches within a year after surgery, VSL#3 prevented onset of pouchitis over placebo (10% vs 40%, P<0.05) [125]. Microbial analysis revealed that the probiotic enriched Lactobacilli and Bifidobacteria and increased bacteria diversity while reducing fungal diversity [126]. In contrast, probiotics containing Lactobacillus and Bifidobacterium did not improve pouch dysfunction nor pouchitis activity [127].
Crohn’s disease
In contrast to UC and pouchitis, several meta-analyses in CD suggest very weak or no benefit of standard probiotics [111,128] with benefits limited to maintaining remission after surgery [111,129,130]. However, there is a strong strain-specific effect [131]; VSL#3 improved endoscopic features [129,130] and decreased mucosal inflammatory cytokine levels [129]. E. coli Nissle1917 and other Lactobacillus strains tested in RCTs lacked benefit [111,128-130,132], but significantly induced of Treg numbers in peripheral blood [132]. Bifidobacterium strains, some of which are included in VSL#3, have not been tested alone, but the combination with prebiotics (synbiotics) significantly improved remission rates, clinical activity and histological scores in CD patients with active disease compared to placebo [133].
Therapeutic mechanisms
Most orally administered current probiotics pass through, although E. coli Nissle1917 [134] can colonize, the intestine and perform several documented protective functions [135,136]. Subsets of resident microbiota have extensive evidence of preventing and treating experimental colitis mediated by well-documented mechanisms [1,48,50,137]. Several comprehensive reviews more extensively document protective effects of probiotics and commensal bacteria [135,136]. The protective mechanisms of traditional probiotics and LBPs include the bacteria themselves (DNA, cytoplasmic and cell wall contents) and their metabolites, such as organic acids, SCFAs, lactic acid that stimulate homeostatic immune and mucosal protection [1,138]. (1) Inhibiting pathobionts. Certain protective bacteria inhibit resident potentially pathogenic microbiota, such as Enterobacteriaceae [50,139], Fusobacterium [50] and Bacteroideceae [140]. Many pathobionts adhere intestinal epithelial cells to induce inflammation, i.e. AIEC and F. varium [45,141,142]. Probiotic E. coli Nissle1917 and L. johnsonii La1 can compete for ecologic niches, epithelial binding and nutrients with pathobionts and inhibit their adhesion and proliferation [143]. Furthermore, decreased luminal pH by organic acids (SCFAs etc.) produced by probiotics and protective resident bacteria [144], anti-bacterial peptides (bacteriocins) [145] and bile-acids modulated by probiotics [146] can inhibit pathobionts. In addition to these bacterial cross-talk, probiotics and resident bacterial species can indirectly (via host cells) affect pathobionts: mucosal PPAR-γ signaling activated by probiotic Clostridium and VSL#3 reduce luminal oxygen and inhibit aerobic Enterobacteriaceae [53,147]; E.coli Nissle1917 induces defensin production by epithelial cells through flagellin-toll like receptor binding (TLR) [148]. (2) Increasing beneficial resident bacteria. Probiotics and LBTs can increase growth of other resident beneficial bacterial species and improve the intestinal eco-system [50,123,149]; increase Lactobacilli [126,139], Bifidobacteria [123,126,149], S. thermophilus [123] and bacterial diversity [126], while reducing fungal diversity [126]. (3) Improving mucosal barrier function. Bifidobacterium strains strengthen epithelial barrier function in UC [150]. SCFAs provide the main energy source of colonic epithelial cells, improve mucosal barrier function and activate colonic Tregs [151,152]. However, in clinical studies, some probiotics work without elevating SCFA [153]. Other protective bacterial metabolites include indoles that bind aryl hydrolase receptors, PXR and sphingolipids [135]. TLR and NOD2-recognition pathways mediate some bacterial protective functions [154]. (4) Mucosal and systemic immunomodulation. Probiotics and resident bacteria can induce anti-inflammatory cytokines (IL-10, TGF-β etc.) and mucosal and systemic regulatory cells (Treg, IgA+ and regulatory B cells) and attenuate inflammatory cytokines (IFN-γ, IL-12p40, TNF-α etc.) [47,48,118,132,154-156].
Clinical concerns
Safety:
Probiotics are well-tolerated with low rates of adverse effects [157,158] although rare cases of sepsis, endocarditis and liver abscess with use of Lactobacillus and fungemia by S. boulardii have occurred, primarily in hospitalized and severely ill or immunocompromised patients with intravenous catheters [158].
Optimal protocols:
Resident microbiota compete with exogenous microbes. Therefore, antibiotic pretreatment seems reasonable to improve engraftment of exogenous probiotics and LBTs. SERES’s RCT demonstrates that pretreatment with vancomycin enhances engraftment of Firmicutes spores [114,115]. Some papers suggest that beneficial effects require 106–108 probiotics/g stool [159]. Given that only 20% of probiotic cells survive [160,161] and stool weight is 1kg, 5×108–1011 probiotic cells administration may be required. However, lower doses may be sufficient for resident LBT strains that proliferate in the intestine. The rectal route (enema) can be considered for pouchitis and pouchitis. D’Inca et al. compared oral versus enema with L. casei GG in active UC and showed a significant advantage of the rectal route for reducing mucosal inflammatory cytokines and modifying the microbiota (Lactobacillus increased, Enterobacteriaceae decreased) [139]. Matthes et al. showed that E. coli Nissle1917 enemas are effective in a dose-dependent manner in active UC patients [162].
Conclusions
Some standard probiotics benefit UC and pouchitis activities. In contrast, the benefits of probiotics for CD seem to be strain specific and limited in maintaining remission after surgery for CD patients. However, VSL#3 containing Bifidobacterium, Lactobacillus and S. salivarius, have more beneficial effects. These positive reported results are subject to publication bias. Although clinical trials of LBTs are just beginning, providing protective resident microbiota to reverse dysbiosis and restore homeostatic microbial community structure and function is an attractive approach that will be actively investigated in the near future.
Prebiotics
Prebiotics are food ingredients that are selectively fermented by host microbes to confer a health benefit. Examples include dietary fiber and oligosaccharides naturally contained in fruits, vegetables and grains [163]. IBD patients are traditionally advised to lower their fiber intake and sometimes fast during flares to reduce mechanical stimulation of the damaged mucosa [164]. However, many studies of various dietary fiber and oligosaccharides suggest favorable results as an emerging treatment approaches (Table 4). Their ability to increase potentially beneficial bacteria and beneficial metabolic effects (SCFAs etc.) have been verified in humans and murine models [163,165].
Table 4.
Prebiotics and synbiotics for IBD (Randomized trials)
Author, year | Disease activity |
Case /Control |
Base Therapy | Agent (s) | Therapy duration |
Outcomes, therapy vs control | PMID |
---|---|---|---|---|---|---|---|
Prebiotics | |||||||
Ulcerative colitis | |||||||
Hallert, 1991 | inactive | 16/13 | Psyllium | 4 m | Improve symptoms | 1654592 | |
Ejderhamn, 1992 | inactive, child | 10/10 | Sulfa | Wheat bran vs psyllium | 6 m | Reduce total bile acid and modified composition | 1360699 |
Fernandez-Banares, 1999 | inactive | 35/37/30 | Psyllium vs 5-ASA vs combined | 12 m | Equivalent relapse rate (40%, 35%, 30%) Increase butyrate | 10022641 | |
Hallert, 2003 | inactive | 22/10 | Wheat bran | 3 m | Increase butyrate | 12769445 | |
Hanai, 2004 | inactive | 22/37 | GBF | 12 m | Improve remission period and tapering steroid | 15067363 | |
Hafer, 2007 | active | 7/7 | 5-ASA, steroid, immunosuppress ant or antibiotics | Lactulose | 4 m | NS on clinical, endoscopic scores Improve QOL | 17784949 |
Casellas, 2007 | active | 10/9 | 5-ASA | Oligofructose-enriched inulin | 2 w | Improve fecal calprotectin, symptoms | 17439507 |
Fujimori, 2009 | inactive, mild | 40/40/40 | 5-ASA, steroid | Psyllium vs Bifidobacterium longum vs synbiotics | 4 w | NS on clinical, endoscopic scores Improve QOL | 19201576 |
Faghfoori, 2011 | inactive | 21/20 | Standard drugs | GBF | 2 m | Improve inflammatory cytokines | 21367884 |
Faghfoori, 2014 | inactive | 23/23 | Standard drugs | GBF | 2 m | Improve CRP, symptoms | 25097845 |
James, 2015 | inactive | 7/7 | 5-ASA, steroid, thiopurine | Inulin-type fructans etc. | 17 d | Tend to normalise gut transit, but does not increase the proportion of carbohydrate fermented, nor increase short-chain fatty acids | 25037189 |
Pouchitis | |||||||
Alles, 1997 | inactive | Crossover 15 | Steroid | Fructo-oligosaccharide (FOS) or resistant starch | 7 d | Resistant starch increases butyrate FOS reduced isobutyrate and isovalerate excretion | 9356550 |
Meijer, 2000 | active | Crossover 20 | Inulin | 3 w | NS (increase butyrate, not significant, correlated with disease activity | 11052521 | |
Welters, 2002 | active | Crossover 20 | Inulin | 3 w | Improve endoscopic and histological scores | 12004211 | |
Crohn’s disease | |||||||
Heaton, 1979 (retro) | active | 22/32 | Steroid, AZA, or sulfa | Fiber-rich, unrefined-carbohydrate diet | 4.3 y | Improve hospital admission (fewer and shoter: 111 d vs 533 d), surgical rate (5% vs 16%) | 519185 |
Jones, 1985 | inactive | 10/10 | Fiber-rich, unrefined-carbohydrate diet | 6 m | Improve relapse rate (0% vs 70%) and ESR | 2862371 | |
Levenstein, 1985 | inactive, active | 28/30 | Normal diet vs fiber-restricted diet | 29 m | Fiber-restriction does not improve outcome (symptoms, need for surgery or hospitalization) | 2996991 | |
Hafer, 2007 | active | 8/9 | 5-ASA, steroid, immunosuppress ant or antibiotics | Lacturose | 4 m | NS | 17784949 |
Benjamin, 2011 | active | 54/49 | Fructo-oligosaccharide | 4 w | NS (clinical response: 22% vs 39%) Reduce IL-6+ DC, increase IL-10+ DC | 21262918 | |
Brotherton, 2011 | inactive | 4/3 | Wheat bran | 4 w | Improve QOL | 24871666 | |
Joossens, 2012 | inactive, active | 34/33 | Oligofructose-enriched inulin | 4 w | Improve disease activity | 21749983 | |
De Preter, 2013 | inactive, active | 25/20 | Oligofructose-enriched inulin | 4 w | Improve clinical diseaese activity Increase acetaldehyde and butyrate | 23303175 | |
Synbiotics (Probiotics + Prebiotics) | |||||||
Ulcerative colitis | |||||||
Furrie, 2005 | active | 8/8 | Bifidobacterium longum + fructo-oligosaccharide/inulin | 4 w | Improve endoscopic score (P=0.06) and b-defencin (P<0.05), TNF-α (P=0.018), IL-1 (P=0.023) | 15647189 | |
Fujimori, 2009 | inactive, mild | 40/40/40 | 5-ASA, steroid | Psyllium/Bifidobacterium longum /synbiotics | 4 w | Improve QOL and CRP | 19201576 |
Ishikawa, 2011 | inactive, active | 21/20 | Bifidobacterium breve Yakult + galacto-oligosaccharide | 2 w | Improve endoscopic score | 21525768 | |
Crohn’s disease | |||||||
Chermesh, 2007 | inactive | 20/10 | Pediacoccus pentoseceus, Lactobacillus raffinolactis, L. paracasei subsp. paracasei 19, L. plantarum 2362 and beta-glucans, inulin, pectin, resistant starch | 24 m | NS | 17211699 | |
Steed, 2010 | active | 13/11 | Bifidobacterium longum + fructo-oligosaccharide/inulin | 6 m | Improve remission rate (62% vs 45%), clinial and histological scores and inflammatory cytokines | 20735782 |
IBD: inflammatory bowel disease, PMID: PubMed identifier, NS: no statistically significant difference in disease activity, AZA; azathioprine, sulfa: salazosulfapyridine, 5-ASA: 5-aminosalicylic acid, DC: dendritic cell, QOL: quality of life, ESR: erythrocyte sedimentation rate, GBF: germinated barley foodstuff, CRP: C-reactive protein, CDAI: Crohn’s disease activity index.
Ulcerative colitis
Many studies focus on QOL, symptoms and bacterial metabolites in UC treated with various prebiotics. Psyllium, germinated barley foodstuff (GBF), lactulose and oligofructose-enriched inulin significantly improve QOL and symptoms in UC patients [166-169]. Intake of psyllium and wheat bran significantly increased fecal butyrate [170,171]. A large RCT with psyllium demonstrated equivalent effectiveness to 5-ASA to maintain remission in UC [171] and a crossover trial in active UC is underway (NCT03998488). GBF contains low-lignified hemicellulose that are efficiently fermented by colonic microbiota [161,165]. GBF reduced CRP [172] and improved clinical and endoscopic scores in active UC in an uncontrolled long-term study [173]. Supplemental oligofructose-enriched inulin with 5-ASA significantly reduced fecal calprotectin (4-fold change) at day 7 compared with 5-ASA alone [168]; a RCT is underway (NCT03653481). A RCT of Synergy1, composed of equal proportions of fructo-oligosaccharide and inulin in active UC has been completed without published results (NCT02093767) and an additional RCT in inactive UC is currently recruiting (NCT02865707). Curcumin, the biologically active component of turmeric with anti-inflammatory and antioxidant effects, can support the growth of protective bacteria [174], so is a promising prebiotic. A large RCT in UC improved remission rates with clinical and endoscopic scores compared to controls [175]. Additional RCTs of curcumin have recently been registered in pediatric (NCT02277223) and adult (NCT02683759) UC patients. Fucosyllactose modifies microbiota [176]; 2 RCTs are underway (NCT03847467, NCT03847467). Glycomacropeptide can modify microbiota and metabolites, reducing Proteobacteria and increasing SCFAs [177]; the first RCT is recruiting UC patients (NCT02825914).
Pouchitis
Two cross-over studies focused on disease activity. Three weeks of inulin supplementation significantly improved clinical and histological scores from baselines associated with increased butyrate levels and reduced pH, B. fragilis and increased secondary bile acids levels [178]. However, the same treatment protocol failed to show significant benefit although a slightly increased butyrate level correlated with reduced disease activity [179]. One possible explanation is variable microbiota in individuals, since efficacy of prebiotics can depend on abundance of resident Bifidobacteria [180].
Crohn’s disease
Restricted dietary fiber did not improve symptoms need for surgery or hospitalization in CD patients [181]. On the contrary, fiber-rich diets significantly reduced surgery in active CD [182] and prevented relapse during remission [183]. Two recent RCTs using oligofructose-enriched inulin inhibited disease activity of active CD associated with increased SCFAs [184] and B. longum and reduced R. gnavus, a potential pathogen in CD [185]. An additional RCT is underway (NCT03653481). 2 RCTs are underway (NCT03847467, NCT03847467) testing fucosyllactose a prebiotic that modifies microbiota [176].
Therapeutic mechanisms
Prebiotics are substrates fermented by resident microbiota to organic acids (SCFAs), CO2, H2 and methane gas [163,165]. (1) Increasing beneficial bacteria. Prebiotics enriched Bifidobacteria [185,186], lactobacilli [186], F. prausnitzii [187] and Clostridium clusters IV and XIVa [188]. (2) Inhibiting pathobionts. Prebiotics can decrease Proteobacteria [177], Bacteroides [178,186], R. gnavus [185] and Candida [186]. (3) Improving mucosal barrier. SCFAs improve mucosal barrier function by providing a key nutrient for colonic epithelial cells [47,48,135], while inulin prevents mucus defects [189]. (4) Mucosal and systemic immunomodulation. Some prebiotics induce Tregs [47,48,135], likely through SCFA production, and intestinal IgA [190]. (5) Absorption of toxic substances. Dietary fiber can adsorb toxic substances, cholesterol, bile acids, and provide bacterial scaffolds to benefit inflammation [191].
Clinical concerns
Safety:
Because prebiotics derive from natural foods, prebiotics are considered to be safe [166,167,169,192]. There were no severe adverse events reported in RCT, although a few food-allergy events occurred [192,193]. Of note, psyllium may cause gastrointestinal obstruction, especially at stenotic sites [192,193] and has not used in CD trials.
Tolerability:
Clinical use is limited by high participant dropout due to bloating and discomfort among IBD patients [184].
Conclusions
Improved beneficial bacteria community structure and metabolism by prebiotics are documented in human volunteers and IBD patients, but relatively few clinical RCTs have been conducted. Although more high quality and disease activity-focused clinical studies are needed, prebiotic therapy is a promising safe and physiologic treatment and maintenance approach to IBD, perhaps in combination with LBPs.
Prebiotic diets
Diet greatly affect microbiota composition and metabolism and IBD dysbiosis is associated with diet [194-196]. Many rigorously designed RCTs have been newly registered. Exclusive enteral nutrition (EEN) is used as first-line therapy for inducing remission in CD with mucosal healing and histological improvement [194,197]. This approach is most widely used in pediatric patients. Responses may be partially attributed to EEN-mediated microbial changes, despite decreased diversity [32]. The Mediterranean style diet (MSD), Asian and semi-vegetarian diets increase beneficial bacteria [198], potentially reduce pathobionts [199] and may benefit IBD patients [200], leading to a RCT investigating the effectiveness of MSD in UC (NCT03053713). The specific carbohydrate diet (SCD), consisting of mostly meat, fruits, vegetables, nuts, oils, and honey with the elimination of grains, has shown efficacy in a retrospective IBD study [201] leading to multiple RCTs, investigating its effects microbial profile and clinical outcome (NCT02858557) (NCT02412553) and efficacy in pediatric (NCT02610101, NCT03301311) and adult (NCT03058679, NCT02412553, NCT02858557) IBD and comparison vs MSD (NCT04082559, NCT03058679). Other promising diets are under investigation, such as the fasting-mimicking diet in UC (NCT03165690), Mashiha in IBD (NCT02796339); the low fermentable oligosaccharide, disaccharide, monosaccharide, and polyol (FODMAP) diet in UC (NCT02469220). Several of these diets, including SCD and low FODMAP, are low in fiber and prebiotics, so they may affect symptoms more than disease efficacy. A better understanding of prebiotics may provide improved advice for patients’ food choices.
Synbiotics
Synbiotics are mixtures of probiotics and prebiotics that beneficially affect the host by improving the survival and implantation of live microbial dietary supplements in the gastrointestinal tract by selectively stimulating the growth and/or by activating the metabolism of one or a limited number of health-promoting bacteria, thus improving host welfare [163]. Given that IBD patients harbor less beneficial intestinal bacteria (Table 1), administration of synbiotics may improve treatment with probiotics or LBPs. Indeed, some papers demonstrated that the benefit of prebiotics depends on baseline abundance of resident protective species [180]. However, clinical studies of synbiotics are limited (Table 4). Ishikawa et al. demonstrated that Bifidobacterial strains plus galacto-oligosaccharide synbiotics improved endoscopic scores and decreased inflammatory markers in treated UC patients [202]. Furrie et al. detected higher numbers of total Bifidobacteria on the mucosal surface in active UC patients fed a synbiotic containing B. longum and Synergy1 than in those taking placebo [203]. In CD, B. longum plus Synergy1 was effective [133]. Overall, prebiotic therapy appears safe and promising, but RCTs are needed to assess the efficacy of dietary/prebiotic interventions. The concept of combined therapies is supported by observations that partial EN plus an exclusion diet high in fiber and fresh fruits and vegetables was better tolerated and induced a more sustained remission in pediatric CD patients compared with standard EEN therapy [197].
FMT
After the breakthrough success of FMT therapy in rCDI in 2013 [19], several accessible fecal banks have been established (OpenBiome etc.) and multiple clinical studies have been performed in IBD patients. This section updates FMT clinical trials in IBD (Table 5), which have been extensively reviewed [57,204,205].
Ulcerative colitis
After initial success of FMT for induction of remission in UC in 1989 [206], four RCTs and several case series have provided promising results. Three out of four RCTs demonstrated a significantly improved clinical, endoscopic and histological scores [58,59,207] although clinical response rates (24–32%) are not as dramatic as in FMT for rCDI (93%). In the unsuccessful RCT [60], the FMT group showed a higher clinical remission rate over controls (41% vs 25%) without statistical significance, likely due to limited subject numbers. However, clinical efficacy was strikingly different with different donors [58]. Indeed, pooled analyses show effectiveness of FMT for active UC [57,204,205]. Bacterial taxa analyses revealed that FMT significantly improved bacterial diversity, which correlates with clinical responses [59,204]. Interestingly, several bacteria taxa associated with remission after FMT, such as Clostridium clusters IV and XVIII, while the presence of Proteobacteria (Sutterella spp) and Fusobacterium species was associated with lack of remission [59]. Ishikawa et al. modified the Fusobacterium-targeted antibiotic ATM cocktail (tetracycline replaced by fosfomycin, AFM cocktail) and showed that the pre-AFM+FMT combination improved outcomes [208]. Moreover, they showed that the reduced abundance of Bacteroidetes by AFM antibiotics pretreatment was clearly restored in FMT responders, but not non-responders. Bacteroidetes is one of the symbiotic taxa [209], that can inhibit C. perfringens [145] and induce Treg [47,48,137]. Two RCTs investigating antibiotics prior to FMT are currently underway (NCT02606032, NCT02033408). Based on limited long-term follow-up reports, the effects of FMT seem to gradually decrease over 3 months [210,211]. However, some responders exhibit long-term remission (>1–2 years) [207,212]. Multiple RCTs are underway in several countries.
Pouchitis
Herfarth et al. demonstrated the difficulty of engraftment of FMT: one out of six patients showed successful engraftment and remission [213]. This could be due to several factors, including donor selection, the dose, frequency and route of administration of FMT, and the pouch microenvironment. Pouches are constructed from the small intestine where potentially beneficial Firmicutes bacteria such as Clostridia are rarely detectable in normal conditions. Of note, Stallmach et al. showed impressive clinical benefits and engraftment by multiple FMT in antibiotic-refractory pouchitis patients [214]: all 5 patients who received FMT achieved clinical response (4/5 remission) and 3/5 patients maintained remission with sequential FMTs. A RCT is currently recruiting (NCT02049502).
Crohn’s disease
Although some case series showed less benefit of FMT in CD patients compared with UC [212,215], many promising case reports and series describe induction of CD remission [204]. Meta-analysis of 6 prospective and uncontrolled trials [204] shows 52% clinical remission rate with publication bias. For adult CD, 58–87% clinical responses were reported [216,217]. Responders showed greater improvement in microbial diversity with a significant shift in fecal microbial composition towards their donor’s profile than non-responders and increased lamina propria Tregs following FMT [216]. FMT via nasogastric tube induced remission in 77.8% of pediatric CD patients 2 weeks after FMT with evidence of engraftment [218]. As seen in UC, responders of FMT in CD showed rapidly improved symptoms and clinical activity several weeks after FMT, but this effect diminished over several months after FMT [216-219] with return to bacterial composition patterns close to pre-FMT levels [219]. To maintain the clinical benefits from FMT, Li et al. suggested performing the next course of FMT less than 4 months after the previous FMT, based on the large scale clinical trial [220]. Multiple RCTs are currently underway.
Therapeutic mechanisms
The therapeutically relevant components of FMT remain elusive [221]. Increased bacterial diversity is clearly associated with successful response of FMT in IBD [59,204,216]. Further, the recipient microbiota after successful FMT resemble donor microbiota, likely due to implantation of donor bacteria and/or donor feces promoting growth of resident bacteria that resemble the donor’s species [221]. Although several potentially relevant species are reported [59], more data are required to support the protective species. FMT is a complex material containing bacteriophages, fungi and metabolites as well as bacteria [59]. Given the therapeutic benefits of filtrated-FMT in rCDI studies [222], cell-free components (bacteriophages and metabolites) in FMT need to be included as research targets. A RCT of filtrated-FMT in UC has been registered (NCT03843385).
Clinical concerns
Safety:
FMT is safe and well tolerated in IBD clinical trials [57,204,223-225]. Importantly, two bacteremias (one death) by ESBL E. coli were reported in immunocompromised patients who received donor stool harboring these strains [226]. Exclusion criteria now include ESBL-producing species. Fecal banks are one source for donor stool.
Effective donor:
FMT success depends on microbial diversity and composition of the donor’s stool, leading to the proposed existence of FMT ‘super donors’ [58,212,227]. The optimal microbial characteristics of donor feces have not defined in IBD. A family member is often chosen as a donor. Because siblings and relatives share similar gut microbiota because of similar lifestyles, diets and genetics [228,229], they may not be optimal donors if the goal is to modulate the recipient’s microbial composition. Switching donors rescued non-responders in a rCDI trial [19]. Whether donors can be optimized by various methods is under investigation.
Optimal protocol:
Engraftment is important factor of efficacy in FMT [230]. A 2018 meta-analysis of FMT protocols indicated that fresh or frozen donor stools, delivery route, and antibiotic pretreatment have no impact on FMT efficacy in IBD [205]. Multiple administrations appear more effective rather than single FMT [205]. Costello et al. demonstrated marked benefit of anaerobically prepared FMT [207], while Cui et al. established a laboratory preparation of fecal materials [217]. Vermeire et al. demonstrated that increased CRP levels at week 2 were an early marker of failure [212], which could allow early rescue therapy in those IBD patients that will not benefit from FMT or guide repeat FMT with a different donor. Because mucosal inflammation reduces microbial diversity and increase pathobionts [231], pre-treatment with immunosuppression to reduce local inflammation and antibiotics to eliminate competing native microbiota may improve engraftment of beneficial species.
Conclusions
Successful FMT have been reported primarily in UC patients. A few positive results exist for CD and pouchitis from case reports and open-label studies. Ongoing multiple RCTs and efforts to optimize protocols, engraftment, donor and recipient selection and matching the optimal donor with individual recipients based on microbial sequencing could improve FMT as a primary therapy. However, we continuously need to consider possible transmission of ‘undefined’ infectious agents in human stools, in contrast to the safety of defined therapeutic LBP cocktails.
Emerging options (bugs as drugs)
This section discusses recent and ongoing pre-clinical studies, technologies and emerging therapeutic concepts [1-3].
Rationally-defined human-derived bacterial consortia - LBPs
A potentially better, more consistent therapeutic approach uses well-characterized, rationally defined and orally-delivered LBPs from resident bacterial species from the intestine of healthy subjects. The most advanced LBP for IBD investigation is a Clostridium cocktail. Atarashi et al. isolated 17 Clostridium strains from healthy human stool screened for induction of FOXP3+ murine CD4+ Tregs [48]. These strains protected several experimental colitis models with high production of SCFAs and induction of colonic IL-10-producing Tregs [48]. All 17 strains belong to Clostridium clusters IV, XIVa or XVIII, which are reduced in IBD patients [13,30]. Administering these strains is designed to restore a normal ecology in IBD patients [232]. Based on these results [40] and mechanistic preclinical studies that identified additional mechanisms beyond the Treg and SCFA pathways, such as correcting dysbiosis and altering non-SCFAs metabolites [50], Janssen Research & Development and Vedanta initiated a Phase 1 clinical study in healthy volunteers. Many other LBTs based on different in vitro and in vivo screening methods are in development and should reach clinical trials soon.
Screening LBPs
Choosing protective resident bacterial strains has been performed in vivo by reductionist [48] and combinatorial [233] approaches in gnotobiotic mice by screening for Treg activation and in vitro with cell lines and human blood lymphocytes [234]. However, results of in vitro studies do not always predict in vivo effects [235]. Peran et al. showed that a specific strain of Lactobacillus salivarius prevented colitis in a TNBS rat model [236]. This strain was selected from 30 laboratory strains for eliciting the highest IL-10/IL-12 and IL-10/TNFα ratios in macrophages. Unfortunately, no strains exhibiting a moderate or low IL-10/IL-12 profile were included in the in vivo study. Similarly strains ranked on their induction of in vitro IL-10/IL-12 cytokine induction closely matches their in vivo attenuation of experimental colitis [234,237]. Our group established a novel in vivo/in vitro combined method using gnotobiotic IL-10-reporter mice to measure individual resident bacterial strain induction of IL-10/IFNγ ratios, with high ratio strains preventing and reversing experimental colitis induced by low IL-10/IFNγ-inducing strains [10].
Substrates from microbiota
Although SCFAs are an important anti-inflammatory substrate in microbe-microbe and microbe-host interactions [144], many other candidate bacterial metabolites affect microbiota and host responses to attenuate mucosal inflammation. Because microbial-based therapies have strong strain- and donor- specific effects [131], purified substrates from defined microbes may provide more consistent results. Examples of established protective bioactive substances produced by probiotic and resident bacteria include p75/p40 from L. rhamnosus that acts through an EGFR-dependent mechanism [238], polyphosphate from lactobacilli [239], lactocepin from VSL#3 [240], polysaccharide-A from B. fragilis [137], an anti-inflammatory protein from F. prausnitzii [241] and kangfuxin liquid extracted from Periplaneta americana dried worms [242]. Most investigators focus on ‘beneficial’ strains to discover a new therapeutic microbial-based tool. In contrast, a unique product from a ‘pathogenic’ E. coli strain, QBECO, is used to immunize hosts (Qu Biologics). Interestingly, purified major macromolecules of an inactivated pathogenic strain of E. coli isolated from a patient with an E. coli infection restored the immune system’s ability to respond productively to invading bacteria in the gastrointestinal tract and rebuild normal barrier function. QBECO treatment improved endoscopic and histological scores in active UC [243] and CD patients cohort [244]. A RCT in CD patients is ongoing [245].
Editing microbiota
Improved understanding of microbiology and metabolic functions suggests ways to modify or block bacterial functions (enzymes and surface molecules) that provide virulence traits. In mice, tungstate treatment, which inhibits molybdenum-cofactor-dependent microbial respiratory pathways, inhibit Enterobacteriaceae expansion and experimental inflammation [246]. Of note, this effect on microbiota was observed only during inflammation. Additional approaches to selectively inhibit pathobiont numbers and functions in IBD include blocking AIEC epithelial attachment through FimH (NCT03709628) and pathobiont-specific bacteriophages (NCT03808103).
Bacteriophages, yeasts and engineered bacteria
Virus (mostly bacteriophages targeting specific bacteria) improves intestinal homeostasis and protects against intestinal injury and pathogen infection [247]. This is potentially clinically relevant, since specific bacteria-targeted bacteriophages may act without affecting beneficial resident bacteria. A RCT examining therapeutic effects of a bacteriophage against AIEC is recruiting CD patients (NCT03808103). Interestingly, bacteriophage DNA can induce colitis and activate IFN-γ responses [248], so clinical toxicity must be examined. Probiotic yeast, including Candida glabrata, produce chitin that reduces bacteria/fungus overgrowth and attenuate DSS colitis with activation of PPAR-γ and induction of IL-10 [249,250]. Although genetically engineered organisms must be carefully handled, several unique bacterial strain express anti-inflammatory substrates such as IL-10, IL-35, trefoil factors, elafin [251-254].
Screening patients: personalized treatment
Based on the heterogeneity of individual IBD patient’s microbiota and patient therapeutic responses, a pilot RCT investigating effects of personalized microbiota-based therapy (antibiotics and prebiotics) is underway in pouchitis (NCT04082559). As described above, efficacy of probiotics, prebiotics and FMT (and anti-TNFα therapy) depends on a patient’s microbiota [255]. Therefore, the best strategy for personalized management of IBD is to identify intestinal microbial profiles prior to beginning therapy or in non-responders [2] to guide optimal microbiome-based therapies.
Conclusions
Microbial-based and microbial-targeted therapies for IBD are emerging with favorable results. The rational for correcting the established dysbiosis in CD, UC and pouchitis patients is well established. Certain antibiotics are promising short-term primary therapies with relatively safety. However, the risk of resistant bacteria and CDI and their uncertain long-term benefit/ toxicity profiles limit maintenance use of antibiotics. FMT is also a promising primary therapy with well-designed RCTs underway. However, the risk of transmission of ‘unknown’ pathogens and long-term benefits remain unclear. A major limitation is variable responses from different donors. In contrast, LBPs, prebiotics and diet are well-defined, safe for long-term use and could be designed for personalized use based on the microbial community structure of individual recipients. Hopefully, these new generation microbial-related therapies will be validated by high quality preclinical and clinical trials. A major discussion point is the best clinical applications for microbial therapy in IBD. Current studies concentrate on single agents inducing remission of active UC. However, we believe that preventing relapse after achieving clinical remission with corticosteroids or biologic therapies in UC or CD patients or with antibiotics in chronic relapsing or antibiotic-resistant pouchitis might be more important areas to investigate. Other clinical needs possibly fulfilled by microbial-based therapies are to use these agents as adjuncts to standard biologic or immunologic therapies to hasten or increase the frequency of deep remission or to maintain quiescent disease after removing the more toxic immune-suppressing agent. Long-term use of this physiologic approach to restore microbial homeostatic function would, in theory, be less toxic and more acceptable to patients (and physicians) who are concerned about risk of infection and neoplasia with sustained immunosuppression. We advocate use of concomitant companion diagnostic tests to profile an individual’s microbiota to guide optimal personalized microbial therapies, determine best timing of intervention and ultimately prevent disease onset in high risk individuals.
Acknowledgement of grant support:
This research supported by National Institute of Health grants, P01DK094779, P30DK34987, P40OD010995 to RBS and by the Crohn’s & Colitis Foundation, 407007 to AO.
Footnotes
Disclosure of financial interest: Dr. Oka declares no competing financial interests. Dr. Sartor has grant support from Janssen, Gusto Global, SERES, BiomX and Vedanta and serves on advisory boards for BiomX, Second Genome, Qu Biologics and Biomica.
References
- 1.Sartor RB; Wu GD Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology 2017, 152, 327–339.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mishima Y; Sartor RB Manipulating resident microbiota to enhance regulatory immune function to treat inflammatory bowel diseases. J. Gastroenterol 2019, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Cohen LJ; Cho JH; Gevers D; Chu H Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Diseases. Gastroenterology 2019, 156, 2174–2189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Sellon RK; Tonkonogy S; Schultz M; Dieleman LA; Grenther W; Balish E; Rennick DM; Sartor RB Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect. Immun 1998, 66, 5224–5231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Stepankova R; Powrie F; Kofronova O; Kozakova H; Hudcovic T; Hrncir T; Uhlig H; Read S; Rehakova Z; Benada O; et al. Segmented filamentous bacteria in a defined bacterial cocktail induce intestinal inflammation in SCID mice reconstituted with CD45RBhigh CD4+ T cells. Inflamm. Bowel Dis 2007, 13, 1202–11. [DOI] [PubMed] [Google Scholar]
- 6.Matsumoto S; Okabe Y; Setoyama H; Takayama K; Ohtsuka J; Funahashi H; Imaoka A; Okada Y; Umesaki Y Inflammatory bowel disease-like enteritis and caecitis in a senescence accelerated mouse P1/Yit strain. Gut 1998, 43, 71–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Rath HC; Herfarth HH; Ikeda JS; Grenther WB; Hamm TE; Balish E; Taurog JD; Hammer RE; Wilson KH; Sartor RB Normal luminal bacteria, especially Bacteroides species, mediate chronic colitis, gastritis, and arthritis in HLA-B27/human beta2 microglobulin transgenic rats. J. Clin. Invest 1996, 98, 945–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Sartor RB Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology 2004, 126, 1620–33. [DOI] [PubMed] [Google Scholar]
- 9.Britton GJ; Contijoch EJ; Mogno I; Vennaro OH; Llewellyn SR; Ng R; Li Z; Mortha A; Merad M; Das A; et al. Microbiotas from Humans with Inflammatory Bowel Disease Alter the Balance of Gut Th17 and RORγt+ Regulatory T Cells and Exacerbate Colitis in Mice. Immunity 2019, 50, 212–224.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Liu B; Oka A; Jang J; Holt L; Britton GJ; Faith J; Sartor RB Protective and Potentially Aggressive Bacterial Species Occur Simultaneously in Healthy Hosts [Abstract]. Gastroenterology 2019, 156, S-78. [Google Scholar]
- 11.Hoarau G; Mukherjee PK; Gower-Rousseau C; Hager C; Chandra J; Retuerto MA; Neut C; Vermeire S; Clemente J; Colombel JF; et al. Bacteriome and mycobiome interactions underscore microbial dysbiosis in familial Crohn’s disease. MBio 2016, 7, 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lloyd-Price J; Arze C; Ananthakrishnan AN; Schirmer M; Avila-Pacheco J; Poon TW; Andrews E; Ajami NJ; Bonham KS; Brislawn CJ; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Frank DN; St Amand AL; Feldman RA; Boedeker EC; Harpaz N; Pace NR Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. U. S. A 2007, 104, 13780–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kim SC; Tonkonogy SL; Albright CA; Tsang J; Balish EJ; Braun J; Huycke MM; Sartor RB Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology 2005, 128, 891–906. [DOI] [PubMed] [Google Scholar]
- 15.Ko CW; Singh S; Feuerstein JD; Falck-Ytter C; Falck-Ytter Y; Cross RK; Crockett S; Feuerstein J; Flamm S; Inadomi J; et al. AGA Clinical Practice Guidelines on the Management of Mild-to-Moderate Ulcerative Colitis. Gastroenterology 2019, 156, 748–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gomollón F; Dignass A; Annese V; Tilg H; Van Assche G; Lindsay JO; Peyrin-Biroulet L; Cullen GJ; Daperno M; Kucharzik T; et al. 3rd European Evidence-based Consensus on the Diagnosis and Management of Crohn’s Disease 2016: Part 1: Diagnosis and Medical Management. J. Crohns. Colitis 2017, 11, 3–25. [DOI] [PubMed] [Google Scholar]
- 17.Matsuoka K; Kobayashi T; Ueno F; Matsui T; Hirai F; Inoue N; Kato J; Kobayashi K; Kobayashi K; Koganei K; et al. Evidence-based clinical practice guidelines for inflammatory bowel disease. J. Gastroenterol 2018, 53, 305–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Laurell A; Sjöberg K Prebiotics and synbiotics in ulcerative colitis. Scand. J. Gastroenterol 2017, 52, 477–485. [DOI] [PubMed] [Google Scholar]
- 19.van Nood E; Vrieze A; Nieuwdorp M; Fuentes S; Zoetendal EG; de Vos WM; Visser CE; Kuijper EJ; Bartelsman JFWM; Tijssen JGP; et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med 2013, 368, 407–15. [DOI] [PubMed] [Google Scholar]
- 20.Garber K Drugging the gut microbiome. Nat. Biotechnol 2015, 33, 228–31. [DOI] [PubMed] [Google Scholar]
- 21.U. S. Food and Drug Administration Early clinical trials with live biotherapeutic products: chemistry, manufacturing, and control information; 2016;
- 22.Gill SR; Pop M; Deboy RT; Eckburg PB; Turnbaugh PJ; Samuel BS; Gordon JI; Relman DA; Fraser-Liggett CM; Nelson KE Metagenomic analysis of the human distal gut microbiome. Science 2006, 312, 1355–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Sekirov I; Russell SL; Antunes LCM; Finlay BB Gut microbiota in health and disease. Physiol. Rev 2010, 90, 859–904. [DOI] [PubMed] [Google Scholar]
- 24.Qin J; Li R; Raes J; Arumugam M; Burgdorf KS; Manichanh C; Nielsen T; Pons N; Levenez F; Yamada T; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tap J; Mondot S; Levenez F; Pelletier E; Caron C; Furet J-P; Ugarte E; Muñoz-Tamayo R; Paslier DLE; Nalin R; et al. Towards the human intestinal microbiota phylogenetic core. Environ. Microbiol 2009, 11, 2574–84. [DOI] [PubMed] [Google Scholar]
- 26.O’Hara AM; Shanahan F The gut flora as a forgotten organ. EMBO Rep. 2006, 7, 688–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lederberg J Infectious history. Science (80-. ). 2000, 288, 287–293. [DOI] [PubMed] [Google Scholar]
- 28.Sender R; Fuchs S; Milo R Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell 2016, 164, 337–40. [DOI] [PubMed] [Google Scholar]
- 29.Lozupone CA; Stombaugh JI; Gordon JI; Jansson JK; Knight R Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Gevers D; Kugathasan S; Denson LA; Vázquez-Baeza Y; Van Treuren W; Ren B; Schwager E; Knights D; Song SJ; Yassour M; et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 2014, 15, 382–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Norman JM; Handley SA; Baldridge MT; Droit L; Liu CY; Keller BC; Kambal A; Monaco CL; Zhao G; Fleshner P; et al. Disease-specific alterations in the enteric virome in inflammatory bowel disease. Cell 2015, 160, 447–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lewis JD; Chen EZ; Baldassano RN; Otley AR; Griffiths AM; Lee D; Bittinger K; Bailey A; Friedman ES; Hoffmann C; et al. Inflammation, Antibiotics, and Diet as Environmental Stressors of the Gut Microbiome in Pediatric Crohn’s Disease. Cell Host Microbe 2015, 18, 489–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zwolinska-Wcislo M; Brzozowski T; Budak A; Kwiecien S; Sliwowski Z; Drozdowicz D; Trojanowska D; Rudnicka-Sosin L; Mach T; Konturek SJ; et al. Effect of Candida colonization on human ulcerative colitis and the healing of inflammatory changes of the colon in the experimental model of Colitis ulcerosa. J. Physiol. Pharmacol 2009, 60, 107–118. [PubMed] [Google Scholar]
- 34.Manichanh C; Rigottier-Gois L; Bonnaud E; Gloux K; Pelletier E; Frangeul L; Nalin R; Jarrin C; Chardon P; Marteau P; et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 2006, 55, 205–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Baumgart M; Dogan B; Rishniw M; Weitzman G; Bosworth B; Yantiss R; Orsi RH; Wiedmann M; McDonough P; Kim SG; et al. Culture independent analysis of ileal mucosa reveals a selective increase in invasive Escherichia coli of novel phylogeny relative to depletion of Clostridiales in Crohn’s disease involving the ileum. ISME J. 2007, 1, 403–18. [DOI] [PubMed] [Google Scholar]
- 36.Giaffer MH; Holdsworth CD; Duerden BI The assessment of faecal flora in patients with inflammatory bowel disease by a simplified bacteriological technique. J. Med. Microbiol 1991, 35, 238–243. [DOI] [PubMed] [Google Scholar]
- 37.Mosca A; Leclerc M; Hugot JP Gut microbiota diversity and human diseases: Should we reintroduce key predators in our ecosystem? Front. Microbiol 2016, 7, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ott SJ; Musfeldt M; Wenderoth DF; Hampe J; Brant O; Fölsch UR; Timmis KN; Schreiber S Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut 2004, 53, 685–693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gophna U; Sommerfeld K; Gophna S; Doolittle WF; Veldhuyzen Van Zanten SJO Differences between tissue-associated intestinal microfloras of patients with Crohn’s disease and ulcerative colitis. J. Clin. Microbiol 2006, 44, 4136–4141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Xun Z; Zhang Q; Xu T; Chen N; Chen F Dysbiosis and ecotypes of the salivary microbiome associated with inflammatory bowel diseases and the assistance in diagnosis of diseases using oral bacterial profiles. Front. Microbiol 2018, 9, 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kelsen J; Bittinger K; Pauly-Hubbard H; Posivak L; Grunberg S; Baldassano R; Lewis JD; Wu GD; Bushman FD Alterations of the Subgingival Microbiota in Pediatric Crohn’s Disease Studied Longitudinally in Discovery and Validation Cohorts. Inflamm. Bowel Dis 2015, 21, 2797–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Atarashi K; Suda W; Luo C; Kawaguchi T; Motoo I; Narushima S; Kiguchi Y; Yasuma K; Watanabe E; Tanoue T; et al. Ectopic colonization of oral bacteria in the intestine drives TH1 cell induction and inflammation. Science 2017, 358, 359–365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Schmitz JM; Tonkonogy SL; Dogan B; Leblond A; Whitehead KJ; Kim SC; Simpson KW; Sartor RB Murine Adherent and Invasive E. coli Induces Chronic Inflammation and Immune Responses in the Small and Large Intestines of Monoassociated IL-10−/− Mice Independent of Long Polar Fimbriae Adhesin A. Inflamm. Bowel Dis 2019, 25, 875–885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Darfeuille-Michaud A; Boudeau J; Bulois P; Neut C; Glasser AL; Barnich N; Bringer MA; Swidsinski A; Beaugerie L; Colombel JF High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology 2004, 127, 412–421. [DOI] [PubMed] [Google Scholar]
- 45.Ohkusa T; Okayasu I; Ogihara T; Morita K; Ogawa M; Sato N Induction of experimental ulcerative colitis by Fusobacterium varium isolated from colonic mucosa of patients with ulcerative colitis. Gut 2003, 52, 79–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Van den Abbeele P; Belzer C; Goossens M; Kleerebezem M; De Vos WM; Thas O; De Weirdt R; Kerckhof F-M; Van de Wiele T Butyrate-producing Clostridium cluster XIVa species specifically colonize mucins in an in vitro gut model. ISME J. 2013, 7, 949–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Atarashi K; Tanoue T; Shima T; Imaoka A; Kuwahara T; Momose Y; Cheng G; Yamasaki S; Saito T; Ohba Y; et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2011, 331, 337–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Atarashi K; Tanoue T; Oshima K; Suda W; Nagano Y; Nishikawa H; Fukuda S; Saito T; Narushima S; Hase K; et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232–6. [DOI] [PubMed] [Google Scholar]
- 49.Hayashi A; Sato T; Kamada N; Mikami Y; Matsuoka K; Hisamatsu T; Hibi T; Roers A; Yagita H; Ohteki T; et al. A single strain of Clostridium butyricum induces intestinal IL-10-producing macrophages to suppress acute experimental colitis in mice. Cell Host Microbe 2013, 13, 711–722. [DOI] [PubMed] [Google Scholar]
- 50.Oka A; Mishima Y; Bongers G; Liu B; Herzog J; Baltus A; Marshall D; Hesse M; Atarashi K; Fukuda S; et al. Tu1844 - IL-10-Independent Protective Activities of Human-Derived Clostridium Strains in Experimental Colitis. Gastroenterology 2018, 154, S-1036. [Google Scholar]
- 51.Rossi O; van Berkel LA; Chain F; Tanweer Khan M; Taverne N; Sokol H; Duncan SH; Flint HJ; Harmsen HJM; Langella P; et al. Faecalibacterium prausnitzii A2–165 has a high capacity to induce IL-10 in human and murine dendritic cells and modulates T cell responses. Sci. Rep 2016, 6, 18507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Rigottier-Gois L Dysbiosis in inflammatory bowel diseases: the oxygen hypothesis. ISME J. 2013, 7, 1256–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Byndloss MX; Olsan EE; Rivera-Chávez F; Tiffany CR; Cevallos SA; Lokken KL; Torres TP; Byndloss AJ; Faber F; Gao Y; et al. Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017, 357, 570–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Rivera-Chávez F; Zhang LF; Faber F; Lopez CA; Byndloss MX; Olsan EE; Xu G; Velazquez EM; Lebrilla CB; Winter SE; et al. Depletion of Butyrate-Producing Clostridia from the Gut Microbiota Drives an Aerobic Luminal Expansion of Salmonella. Cell Host Microbe 2016, 19, 443–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Dubuquoy L; Jansson EA; Deeb S; Rakotobe S; Karoui M; Colombel J-F; Auwerx J; Pettersson S; Desreumaux P Impaired expression of peroxisome proliferator-activated receptor gamma in ulcerative colitis. Gastroenterology 2003, 124, 1265–76. [DOI] [PubMed] [Google Scholar]
- 56.Quinton JF; Sendid B; Reumaux D; Duthilleul P; Cortot A; Grandbastien B; Charrier G; Targan SR; Colombel JF; Poulain D Anti-Saccharomyces cerevisiae mannan antibodies combined with antineutrophil cytoplasmic autoantibodies in inflammatory bowel disease: prevalence and diagnostic role. Gut 1998, 42, 788–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Imdad A; Nicholson MR; Tanner-Smith EE; Zackular JP; Gomez-Duarte OG; Beaulieu DB; Acra S Fecal transplantation for treatment of inflammatory bowel disease. Cochrane Database Syst. Rev 2018, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Moayyedi P; Surette MG; Kim PT; Libertucci J; Wolfe M; Onischi C; Armstrong D; Marshall JK; Kassam Z; Reinisch W; et al. Fecal Microbiota Transplantation Induces Remission in Patients With Active Ulcerative Colitis in a Randomized Controlled Trial. Gastroenterology 2015, 149, 102–109.e6. [DOI] [PubMed] [Google Scholar]
- 59.Paramsothy S; Kamm MA; Kaakoush NO; Walsh AJ; van den Bogaerde J; Samuel D; Leong RWL; Connor S; Ng W; Paramsothy R; et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet 2017, 6736, 1–11. [DOI] [PubMed] [Google Scholar]
- 60.Rossen NG; Fuentes S; van der Spek MJ; Tijssen JG; Hartman JHA; Duflou A; Löwenberg M; van den Brink GR; Mathus-Vliegen EMH; de Vos WM; et al. Findings From a Randomized Controlled Trial of Fecal Transplantation for Patients With Ulcerative Colitis. Gastroenterology 2015, 149, 110–118.e4. [DOI] [PubMed] [Google Scholar]
- 61.Bazin T; Hooks KB; Barnetche T; Truchetet M-E; Enaud R; Richez C; Dougados M; Hubert C; Barré A; Nikolski M; et al. Microbiota Composition May Predict Anti-Tnf Alpha Response in Spondyloarthritis Patients: an Exploratory Study. Sci. Rep 2018, 8, 5446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Aden K; Rehman A; Waschina S; Pan W-H; Walker A; Lucio M; Nunez AM; Bharti R; Zimmerman J; Bethge J; et al. Metabolic Functions of Gut Microbes Associate With Efficacy of Tumor Necrosis Factor Antagonists in Patients With Inflammatory Bowel Diseases. Gastroenterology 2019, 157, 1279–1292.e11. [DOI] [PubMed] [Google Scholar]
- 63.Michail S; Durbin M; Turner D; Griffiths AM; Mack DR; Hyams J; Leleiko N; Kenche H; Stolfi A; Wine E Alterations in the gut microbiome of children with severe ulcerative colitis. Inflamm. Bowel Dis 2012, 18, 1799–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Routy B; Le Chatelier E; Derosa L; Duong CPM; Alou MT; Daillère R; Fluckiger A; Messaoudene M; Rauber C; Roberti MP; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [DOI] [PubMed] [Google Scholar]
- 65.Browne HP; Forster SC; Anonye BO; Kumar N; Neville BA; Stares MD; Goulding D; Lawley TD Culturing of “unculturable” human microbiota reveals novel taxa and extensive sporulation. Nature 2016, 533, 543–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.NIH Human Microbiome Project The Inflammatory Bowel Disease Multi’omics Database Available online: https://ibdmdb.org/.
- 67.Kirsner JB Historical Aspects of Inflammatory Bowel Disease. J. Clin. Gastroenterol 1988, 10, 286–297. [DOI] [PubMed] [Google Scholar]
- 68.Perencevich M; Burakoff R Use of antibiotics in the treatment of inflammatory bowel disease. Inflamm. Bowel Dis 2006, 12, 651–64. [DOI] [PubMed] [Google Scholar]
- 69.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. [DOI] [PubMed] [Google Scholar]
- 70.Gao J; Gillilland MG; Owyang C Rifaximin, gut microbes and mucosal inflammation: unraveling a complex relationship. Gut Microbes 2014, 5, 571–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sartor RB Review article: the potential mechanisms of action of rifaximin in the management of inflammatory bowel diseases. Aliment. Pharmacol. Ther 2016, 43 Suppl 1, 27–36. [DOI] [PubMed] [Google Scholar]
- 72.Wang S-L; Wang Z-R; Yang C-Q Meta-analysis of broad-spectrum antibiotic therapy in patients with active inflammatory bowel disease. Exp. Ther. Med 2012, 4, 1051–1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Khan KJ; Ullman TA; Ford AC; Abreu MT; Abadir A; Marshall JK; Talley NJ; Moayyedi P Antibiotic therapy in inflammatory bowel disease: A systematic review and meta-analysis. Am. J. Gastroenterol 2011, 106, 661–673. [DOI] [PubMed] [Google Scholar]
- 74.Ohkusa T; Kato K; Terao S; Chiba T; Mabe K; Murakami K; Mizokami Y; Sugiyama T; Yanaka A; Takeuchi Y; et al. Newly developed antibiotic combination therapy for ulcerative colitis: a double-blind placebo-controlled multicenter trial. Am. J. Gastroenterol 2010, 105, 1820–1829. [DOI] [PubMed] [Google Scholar]
- 75.Ohkusa T; Nomura T; Terai T; Miwa H; Kobayashi O; Hojo M; Takei Y; Ogihara T; Hirai S; Okayasu I; et al. Effectiveness of antibiotic combination therapy in patients with active ulcerative colitis: a randomized, controlled pilot trial with long-term follow-up. Scand. J. Gastroenterol 2005, 40, 1334–42. [DOI] [PubMed] [Google Scholar]
- 76.Nomura T; Ohkusa T; Okayasu I; Yoshida T; Sakamoto M; Hayashi H; Benno Y; Hirai S; Hojo M; Kobayashi O; et al. Mucosa-associated bacteria in ulcerative colitis before and after antibiotic combination therapy. Aliment. Pharmacol. Ther 2005, 21, 1017–1027. [DOI] [PubMed] [Google Scholar]
- 77.Burke DA; Axon AT; Clayden SA; Dixon MF; Johnston D; Lacey RW The efficacy of tobramycin in the treatment of ulcerative colitis. Aliment. Pharmacol. Ther 1990, 4, 123–9. [DOI] [PubMed] [Google Scholar]
- 78.Lobo AJ; Burke DA; Sobala GM; Axon AT Oral tobramycin in ulcerative colitis: effect on maintenance of remission. Aliment. Pharmacol. Ther 1993, 7, 155–8. [DOI] [PubMed] [Google Scholar]
- 79.Turunen UM; Färkkilä MA; Hakala K; Seppälä K; Sivonen A; Ogren M; Vuoristo M; Valtonen VV ; Miettinen TA Long-term treatment of ulcerative colitis with ciprofloxacin: a prospective, double-blind, placebo-controlled study. Gastroenterology 1998, 115, 1072–8. [DOI] [PubMed] [Google Scholar]
- 80.Singh S; Stroud AM; Holubar SD; Sandborn WJ; Pardi DS Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane database Syst. Rev 2015, CD001176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Segal JP; Ding NS; Worley G; Mclaughlin S; Preston S; Faiz OD; Clark SK; Hart AL Systematic review with meta-analysis: the management of chronic refractory pouchitis with an evidence-based treatment algorithm. Aliment. Pharmacol. Ther 2017, 45, 581–592. [DOI] [PubMed] [Google Scholar]
- 82.Holubar SD; Cima RR; Sandborn WJ; Pardi DS Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane database Syst. Rev 2010, 2019, CD001176. [DOI] [PubMed] [Google Scholar]
- 83.Gosselink MP; Schouten WR; Van Lieshout LMC; Hop WCJ; Laman JD; Ruseler-Van Embden JGH Eradication of pathogenic bacteria and restoration of normal pouch flora: Comparison of metronidazole and ciprofloxacin in the treatment of pouchitis. Dis. Colon Rectum 2004, 47, 1519–1525. [DOI] [PubMed] [Google Scholar]
- 84.Dubinsky V; Reshef L; Bar N; Keizer D; Golan N; Rabinowitz K; Godny L; Yadgar K; Zonensain K; Tulchinsky H; et al. Predominantly Antibiotic-resistant Intestinal Microbiome Persists in Patients With Pouchitis Who Respond to Antibiotic Therapy. Gastroenterology 2019. [DOI] [PubMed] [Google Scholar]
- 85.Singh S; Garg SK; Pardi DS; Wang Z; Murad MH; Loftus EV Comparative efficacy of pharmacologic interventions in preventing relapse of Crohn’s disease after surgery: a systematic review and network meta-analysis. Gastroenterology 2015, 148, 64–76.e2; quiz e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Townsend CM; Parker CE; MacDonald JK; Nguyen TM; Jairath V; Feagan BG; Khanna R Antibiotics for induction and maintenance of remission in Crohn’s disease. Cochrane database Syst. Rev 2019, 2, CD012730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Su JW; Ma JJ; Zhang HJ Use of antibiotics in patients with Crohn’s disease: a systematic review and meta-analysis. J. Dig. Dis 2015, 16, 58–66. [DOI] [PubMed] [Google Scholar]
- 88.Patton PH; Parker CE; MacDonald JK; Chande N Anti-tuberculous therapy for maintenance of remission in Crohn’s disease. Cochrane database Syst. Rev 2016, 7, CD000299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Prantera C; Lochs H; Campieri M; Scribano ML; Sturniolo GC; Castiglione F; Cottone M Antibiotic treatment of Crohn’s disease: results of a multicentre, double blind, randomized, placebo-controlled trial with rifaximin. Aliment. Pharmacol. Ther 2006, 23, 1117–25. [DOI] [PubMed] [Google Scholar]
- 90.Selby W; Pavli P; Crotty B; Florin T; Radford-Smith G; Gibson P; Mitchell B; Connell W; Read R; Merrett M; et al. Two-year combination antibiotic therapy with clarithromycin, rifabutin, and clofazimine for Crohn’s disease. Gastroenterology 2007, 132, 2313–9. [DOI] [PubMed] [Google Scholar]
- 91.Aberra FN; Brensinger CM; Bilker WB; Lichtenstein GR; Lewis JD Antibiotic use and the risk of flare of inflammatory bowel disease. Clin. Gastroenterol. Hepatol 2005, 3, 459–465. [DOI] [PubMed] [Google Scholar]
- 92.Keshaw H; Foong KS; Forbes A; Day RM Perianal fistulae in Crohn’s Disease: current and future approaches to treatment. Inflamm. Bowel Dis 2010, 16, 870–80. [DOI] [PubMed] [Google Scholar]
- 93.Tozer PJ; Burling D; Gupta A; Phillips RKS; Hart AL Review article: medical, surgical and radiological management of perianal Crohn’s fistulas. Aliment. Pharmacol. Ther 2011, 33, 5–22. [DOI] [PubMed] [Google Scholar]
- 94.Bernstein LH; Frank MS; Brandt LJ; Boley SJ Healing of perineal Crohn’s disease with metronidazole. Gastroenterology 1980, 79, 599. [PubMed] [Google Scholar]
- 95.Feagins LA; Holubar SD; Kane SV; Spechler SJ Current strategies in the management of intra-abdominal abscesses in Crohn’s disease. Clin. Gastroenterol. Hepatol 2011, 9, 842–50. [DOI] [PubMed] [Google Scholar]
- 96.Krook A; Lindström B; Kjellander J; Järnerot G; Bodin L Relation between concentrations of metronidazole and Bacteroides spp in faeces of patients with Crohn’s disease and healthy individuals. J. Clin. Pathol 1981, 34, 645–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Rafii F; Ruseler-Van Embden JG; van Lieshout LM Changes in bacterial enzymes and PCR profiles of fecal bacteria from a patient with ulcerative colitis before and after antimicrobial treatments. Dig. Dis. Sci 1999, 44, 637–42. [DOI] [PubMed] [Google Scholar]
- 98.Morikawa K; Watabe H; Araake M; Morikawa S Modulatory effect of antibiotics on cytokine production by human monocytes in vitro. Antimicrob. Agents Chemother 1996, 40, 1366–1370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wan YC; Li T; Han Y-D; Zhang H-Y; Lin H; Zhang B EFFECT OF PREGNANE XENOBIOTIC RECEPTOR ACTIVATION ON INFLAMMATORY BOWEL DISEASE TREATED WITH RIFAXIMIN. J. Biol. Regul. Homeost. Agents 2015, 29, 401–10. [PubMed] [Google Scholar]
- 100.Garrido-Mesa N; Camuesco D; Arribas B; Comalada M; Bailón E; Cueto-Sola M; Utrilla P; Nieto A; Zarzuelo A; Rodríguez-Cabezas ME; et al. The intestinal anti-inflammatory effect of minocycline in experimental colitis involves both its immunomodulatory and antimicrobial properties. Pharmacol. Res 2011, 63, 308–19. [DOI] [PubMed] [Google Scholar]
- 101.Shen B; Achkar JP; Lashner BA; Ormsby AH; Remzi FH; Brzezinski A; Bevins CL; Bambrick ML; Seidner DL; Fazio VW A randomized clinical trial of ciprofloxacin and metronidazole to treat acute pouchitis. Inflamm. Bowel Dis 2001, 7, 301–5. [DOI] [PubMed] [Google Scholar]
- 102.Leung W; Malhi G; Willey BM; McGeer AJ; Borgundvaag B; Thanabalan R; Gnanasuntharam P; Le B; Weizman AV; Croitoru K; et al. Prevalence and predictors of MRSA, ESBL, and VRE colonization in the ambulatory IBD population. J. Crohn’s Colitis 2012, 6, 743–749. [DOI] [PubMed] [Google Scholar]
- 103.Balram B; Battat R; Al-Khoury A; D’Aoust J; Afif W; Bitton A; Lakatos PL; Bessissow T Risk Factors Associated with Clostridium difficile infection in inflammatory bowel disease: A systematic review and meta-analysis. J. Crohn’s Colitis 2019, 13, 27–38. [DOI] [PubMed] [Google Scholar]
- 104.Ferreyra JA; Wu KJ; Hryckowian AJ; Bouley DM; Weimer BC; Sonnenburg JL Gut microbiota-produced succinate promotes C. difficile infection after antibiotic treatment or motility disturbance. Cell Host Microbe 2014, 16, 770–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Roy A; Lichtiger S Clostridium difficile Infection: A Rarity in Patients Receiving Chronic Antibiotic Treatment for Crohn’s Disease. Inflamm. Bowel Dis 2016, 22, 648–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Goldstein EJC; Johnson SJ; Maziade P; Evans CT; Sniffen JC; Millette M; Mcfarland LV Anaerobe Probiotics and prevention of Clostridium dif fi cile infection. 2017, 45, 114–119. [DOI] [PubMed] [Google Scholar]
- 107.Gupta V; Rodrigues R; Nguyen D; Sauk J; Khalili H; Yajnik V; Ananthakrishnan AN Adjuvant use of antibiotics with corticosteroids in inflammatory bowel disease exacerbations requiring hospitalisation: a retrospective cohort study and meta-analysis. Aliment. Pharmacol. Ther 2016, 43, 52–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Chang JC; Cohen RD Medical management of severe ulcerative colitis. Gastroenterol. Clin. North Am 2004, 33, 235–50, viii. [DOI] [PubMed] [Google Scholar]
- 109.Ledder O Antibiotics in inflammatory bowel diseases: do we know what we’re doing? Transl. Pediatr 2019, 8, 42–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Sanders ME; Guarner F; Guerrant R; Holt PR; Quigley EMM; Sartor RB; Sherman PM; Mayer EA An update on the use and investigation of probiotics in health and disease. Gut 2013, 62, 787–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Ganji-Arjenaki M; Rafieian-Kopaei M Probiotics are a good choice in remission of inflammatory bowel diseases: A meta analysis and systematic review. J. Cell. Physiol 2018, 233, 2091–2103. [DOI] [PubMed] [Google Scholar]
- 112.Peng L; Zhong Y; Wang A; Jiang Z Probiotics combined with aminosalicylic acid affiliates remission of ulcerative colitis: a meta-analysis of randomized controlled trial. Biosci. Rep 2019, 39, 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Astó E; Méndez I; Audivert S; Farran-Codina A; Espadaler J The Efficacy of Probiotics, Prebiotic Inulin-Type Fructans, and Synbiotics in Human Ulcerative Colitis: A Systematic Review and Meta-Analysis. Nutrients 2019, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Simmons S; Diao L; O’Brien E; Chafee M; Zhao J; Bernardo P; Cook D; Trucksis M; Henn MR Tu2019 - Engraftment of Ser-287, an Investigational Microbiome Therapeutic, is Related to Clinical Remission in a Placebo-Controlled, Double-Blind Randomized Trial (Seres-101) in Patients with Active Mild to Moderate Ulcerative Colitis (UC). Gastroenterology 2018, 154, S-1371–S-1372. [Google Scholar]
- 115.Bharat Misra, Curran John, Herfarth Hans H., Jagarlamudi Kiran, Oneto Caterina, Bhandari Bal R., Wiener Gregory, Kerman David H., Moss Alan C., Pomerantz Roger, Zhao Jeff, Bernardo Patricia, Simmons Sheri, Diao Liyang, O’Brien Edward, Henn Matthew R., M.T. 85 - SER-287, an Investigational Microbiome Therapeutic, Induces Remission and Endoscopic Improvement in a Placebo-Controlled, Double-Blind Randomized Trial in Patients with Active Mild-to-Moderate Ulcerative Colitis. Gastroenterology 2018, 154, S-85. [Google Scholar]
- 116.Diao L; Nnamani MC; OBrien E; Desjardins C; Martinez A; Simmons S; Litcofsky K; McGovern B; Weigand S; Cook D; et al. 623 – Ser-287, an Investigational Microbiome Therapeutic, Induces Widespread Transcriptional Changes Related to Clinical Remission in a Placebo-Controlled, Double-Blind Randomized Trial (Seres-101) in Patients with Active Mild-To-Moderate Ulcerative Colit. Gastroenterology 2019, 156, S-130. [Google Scholar]
- 117.Miele E; Pascarella F; Giannetti E; Quaglietta L; Baldassano RN; Staiano A Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am. J. Gastroenterol 2009, 104, 437–43. [DOI] [PubMed] [Google Scholar]
- 118.Oliva S; Di Nardo G; Ferrari F; Mallardo S; Rossi P; Patrizi G; Cucchiara S; Stronati L Randomised clinical trial: the effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis. Aliment. Pharmacol. Ther 2012, 35, 327–34. [DOI] [PubMed] [Google Scholar]
- 119.Palumbo VD; Romeo M; Marino Gammazza A; Carini F; Damiani P; Damiano G; Buscemi S; Lo Monte AI; Gerges-Geagea A; Jurjus A; et al. The long-term effects of probiotics in the therapy of ulcerative colitis: A clinical study. Biomed. Pap. Med. Fac. Univ. Palacky. Olomouc. Czech. Repub 2016, 160, 372–7. [DOI] [PubMed] [Google Scholar]
- 120.Ritchie ML; Romanuk TN A meta-analysis of probiotic efficacy for gastrointestinal diseases. PLoS One 2012, 7, e34938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Dong J; Teng G; Wei T; Gao W; Wang H Methodological Quality Assessment of Meta-Analyses and Systematic Reviews of Probiotics in Inflammatory Bowel Disease and Pouchitis. PLoS One 2016, 11, e0168785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Yasueda A; Mizushima T; Nezu R; Sumi R; Tanaka M; Nishimura J; Kai Y; Hirota M; Osawa H; Nakajima K; et al. The effect of Clostridium butyricum MIYAIRI on the prevention of pouchitis and alteration of the microbiota profile in patients with ulcerative colitis. Surg. Today 2016, 46, 939–949. [DOI] [PubMed] [Google Scholar]
- 123.Gionchetti P; Rizzello F; Venturi A; Brigidi P; Matteuzzi D; Bazzocchi G; Poggioli G; Miglioli M; Campieri M Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: A double-blind, placebo-controlled trial. Gastroenterology 2000, 119, 305–309. [DOI] [PubMed] [Google Scholar]
- 124.Shen B; Brzezinski A; Fazio VW; Remzi FH; Achkar JP; Bennett AE; Sherman K; Lashner BA Maintenance therapy with a probiotic in antibiotic-dependent pouchitis: Experience in clinical practice. Aliment. Pharmacol. Ther 2005, 22, 721–728. [DOI] [PubMed] [Google Scholar]
- 125.Gionchetti P; Rizzello F; Helwig U; Venturi A; Lammers KM; Brigidi P; Vitali B; Poggioli G; Miglioli M; Campieri M Prophylaxis of pouchitis onset with probiotic therapy: A double-blind, placebo-controlled trial. Gastroenterology 2003, 124, 1202–1209. [DOI] [PubMed] [Google Scholar]
- 126.Kühbacher T; Ott SJ; Helwig U; Mimura T; Rizzello F; Kleessen B; Gionchetti P; Blaut M; Campieri M; Fölsch UR; et al. Bacterial and fungal microbiota in relation to probiotic therapy (VSL#3) in pouchitis. Gut 2006, 55, 833–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Bengtsson J; Adlerberth I; Östblom A; Saksena P; Öresland T; Börjesson L Effect of probiotics (Lactobacillus plantarum 299 plus Bifidobacterium Cure21) in patients with poor ileal pouch function: A randomised controlled trial. Scand. J. Gastroenterol 2016, 51, 1087–1092. [DOI] [PubMed] [Google Scholar]
- 128.Derwa Y; Gracie DJ; Hamlin PJ; Ford AC Systematic review with meta-analysis: the efficacy of probiotics in inflammatory bowel disease. Aliment. Pharmacol. Ther 2017, 46, 389–400. [DOI] [PubMed] [Google Scholar]
- 129.Fedorak RN; Feagan BG; Hotte N; Leddin D; Dieleman LA; Petrunia DM; Enns R; Bitton A; Chiba N; Paré P; et al. The probiotic VSL#3 has anti-inflammatory effects and could reduce endoscopic recurrence after surgery for Crohn’s disease. Clin. Gastroenterol. Hepatol 2015, 13, 928–35.e2. [DOI] [PubMed] [Google Scholar]
- 130.Campieri M; Rizzello F; Venturi A; Poggioli G; Ugolini F Combination of antibiotic and probiotic treatment is efficacious in prophylaxis of post-operative recurrence of Crohn’s disease: A randomized controlled study VS mesalamine. Gastroenterology 2000, 118, A781. [Google Scholar]
- 131.Whelan K; Quigley EMM Probiotics in the management of irritable bowel syndrome and inflammatory bowel disease. Curr. Opin. Gastroenterol 2013, 29, 184–9. [DOI] [PubMed] [Google Scholar]
- 132.Lorea Baroja M; Kirjavainen PV; Hekmat S; Reid G Anti-inflammatory effects of probiotic yogurt in inflammatory bowel disease patients. Clin. Exp. Immunol 2007, 149, 470–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Steed H; Macfarlane GT; Blackett KL; Bahrami B; Reynolds N; Walsh SV; Cummings JH; Macfarlane S Clinical trial: the microbiological and immunological effects of synbiotic consumption - a randomized double-blind placebo-controlled study in active Crohn’s disease. Aliment. Pharmacol. Ther 2010, 32, 872–83. [DOI] [PubMed] [Google Scholar]
- 134.Joeres-Nguyen-Xuan TH; Boehm SK; Joeres L; Schulze J; Kruis W Survival of the probiotic Escherichia coli Nissle 1917 (EcN) in the gastrointestinal tract given in combination with oral mesalamine to healthy volunteers. Inflamm. Bowel Dis 2010, 16, 256–62. [DOI] [PubMed] [Google Scholar]
- 135.Skelly AN; Sato Y; Kearney S; Honda K Mining the microbiota for microbial and metabolite-based immunotherapies. Nat. Rev. Immunol 2019, 19, 305–323. [DOI] [PubMed] [Google Scholar]
- 136.Sassone-Corsi M; Raffatellu M No Vacancy: How Beneficial Microbes Cooperate with Immunity To Provide Colonization Resistance to Pathogens. J. Immunol 2015, 194, 4081–4087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Round JL; Mazmanian SK Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. U. S. A 2010, 107, 12204–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Böhmig GA; Krieger PM; Säemann MD; Wenhardt C; Pohanka E; Zlabinger GJ n-butyrate downregulates the stimulatory function of peripheral blood-derived antigen-presenting cells: a potential mechanism for modulating T-cell responses by short-chain fatty acids. Immunology 1997, 92, 234–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.D’Incà R; Barollo M; Scarpa M; Grillo AR; Brun P; Vettorato MG; Castagliuolo I; Sturniolo GC Rectal administration of Lactobacillus casei DG modifies flora composition and Toll-like receptor expression in colonic mucosa of patients with mild ulcerative colitis. Dig. Dis. Sci 2011, 56, 1178–87. [DOI] [PubMed] [Google Scholar]
- 140.Kruis W; Frič P; Pokrotnieks J; Lukáš M; Fixa B; Kaščák M; Kamm MA; Weismueller J; Beglinger C; Stolte M; et al. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut 2004, 53, 1617–1623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Neutra MR; Frey A; Kraehenbuhl JP Epithelial M cells: gateways for mucosal infection and immunization. Cell 1996, 86, 345–8. [DOI] [PubMed] [Google Scholar]
- 142.Barnich N; Carvalho FA; Glasser A; Darcha C; Jantscheff P; Allez M; Peeters H; Bommelaer G; Desreumaux P; Colombel J-F; et al. CEACAM6 acts as a receptor for adherent-invasive E. coli, supporting ileal mucosa colonization in Crohn disease. J. Clin. Invest 2007, 117, 1566–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Bernet-Camard MF; Liévin V; Brassart D; Neeser JR; Servin AL; Hudault S The human Lactobacillus acidophilus strain LA1 secretes a nonbacteriocin antibacterial substance(s) active in vitro and in vivo. Appl. Environ. Microbiol 1997, 63, 2747–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Fukuda S; Toh H; Hase K; Oshima K; Nakanishi Y; Yoshimura K; Tobe T; Clarke JM; Topping DL; Suzuki T; et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature 2011, 469, 543–547. [DOI] [PubMed] [Google Scholar]
- 145.Crost EH; Pujol A; Ladiré M; Dabard J; Raibaud P; Carlier JP; Fons M Production of an antibacterial substance in the digestive tract involved in colonization-resistance against Clostridium perfringens. Anaerobe 2010, 16, 597–603. [DOI] [PubMed] [Google Scholar]
- 146.Buffie CG; Bucci V; Stein RR; McKenney PT; Ling L; Gobourne A; No D; Liu H; Kinnebrew M; Viale A; et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 2015, 517, 205–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Marion-Letellier R; Déchelotte P; Iacucci M; Ghosh S Dietary modulation of peroxisome proliferator-activated receptor gamma. Gut 2009, 58, 586–93. [DOI] [PubMed] [Google Scholar]
- 148.Petrof EO Probiotics and Gastrointestinal Disease: Clinical Evidence and Basic Science. Antiinflamm. Antiallergy. Agents Med. Chem 2009, 8, 260–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Cui HH; Chen CL; De Wang J ; Yang YJ; Cun Y; Wu JB; Liu YH; Dan HL; Jian YT; Chen XQ Effects of probiotic on intestinal mucosa of patients with ulcerative colitis. World J. Gastroenterol 2004, 10, 1521–1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Duranti S; Gaiani F; Mancabelli L; Milani C; Grandi A; Bolchi A; Santoni A; Lugli GA; Ferrario C; Mangifesta M; et al. Elucidating the gut microbiome of ulcerative colitis: bifidobacteria as novel microbial biomarkers. FEMS Microbiol. Ecol 2016, 92, 1–30. [DOI] [PubMed] [Google Scholar]
- 151.Furusawa Y; Obata Y; Fukuda S; Endo T. a ; Nakato G; Takahashi D; Nakanishi Y; Uetake C; Kato K; Kato T; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–50. [DOI] [PubMed] [Google Scholar]
- 152.Smith PM; Howitt MR; Panikov N; Michaud M; Gallini CA; Bohlooly-Y M; Glickman JN; Garrett WS The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Yoshimatsu Y; Yamada A; Furukawa R; Sono K; Osamura A; Nakamura K; Aoki H; Tsuda Y; Hosoe N; Takada N; et al. Effectiveness of probiotic therapy for the prevention of relapse in patients with inactive ulcerative colitis. World J. Gastroenterol 2015, 21, 5985–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Mishima Y; Oka A; Liu B; Herzog JW; Eun CS; Fan T-J; Bulik-Sullivan E; Carroll IM; Hansen JJ; Chen L; et al. Microbiota maintain colonic homeostasis by activating TLR2/MyD88/PI3K signaling in IL-10-producing regulatory B cells. J. Clin. Invest 2019, 130, 3702–3716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Pronio A; Montesani C; Butteroni C; Vecchione S; Mumolo G; Vestri AR; Vitolo D; Boirivant M Probiotic administration in patients with ileal pouch-anal anastomosis for ulcerative colitis is associated with expansion of mucosal regulatory cells. Inflamm. Bowel Dis 2008, 14, 662–668. [DOI] [PubMed] [Google Scholar]
- 156.Hiramatsu Y; Hosono A; Takahashi K; Kaminogawa S Bifidobacterium components have immunomodulatory characteristics dependent on the method of preparation. Cytotechnology 2007, 55, 79–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Jia K; Tong X; Wang R; Song X The clinical effects of probiotics for inflammatory bowel disease: A meta-analysis. Medicine (Baltimore). 2018, 97, e13792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Snydman DR The safety of probiotics. Clin. Infect. Dis 2008, 46 Suppl 2, S104–11; discussion S144–51. [DOI] [PubMed] [Google Scholar]
- 159.Charteris WP; Kelly PM; Morelli L; Collins JK Development and application of an in vitro methodology to determine the transit tolerance of potentially probiotic Lactobacillus and Bifidobacterium species in the upper human gastrointestinal tract. J. Appl. Microbiol 1998, 84, 759–68. [DOI] [PubMed] [Google Scholar]
- 160.Kuisma J; Mentula S; Jarvinen H; Kahri A; Saxelin M; Farkkila M Effect of Lactobacillus rhamnosus GG on ileal pouch inflammation and microbial flora. Aliment. Pharmacol. Ther 2003, 17, 509–15. [DOI] [PubMed] [Google Scholar]
- 161.Bezkorovainy A Probiotics: determinants of survival and growth in the gut. Am. J. Clin. Nutr 2001, 73, 399S–405S. [DOI] [PubMed] [Google Scholar]
- 162.Matthes H; Krummenerl T; Giensch M; Wolff C; Schulze J Clinical trial: probiotic treatment of acute distal ulcerative colitis with rectally administered Escherichia coli Nissle 1917 (EcN). BMC Complement. Altern. Med 2010, 10, 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Gibson GR; Roberfroid MB Dietary modulation of the human colonic microbiota: introducing the concept of prebiotics. J. Nutr 1995, 125, 1401–12. [DOI] [PubMed] [Google Scholar]
- 164.Hallert C; Kaldma M; Petersson BG Ispaghula husk may relieve gastrointestinal symptoms in ulcerative colitis in remission. Scand. J. Gastroenterol 1991, 26, 747–50. [DOI] [PubMed] [Google Scholar]
- 165.Kanauchi O; Fujiyama Y; Mitsuyama K; Araki Y; Ishii T; Nakamura T; Hitomi Y; Agata K; Saiki T; Andoh A; et al. Increased growth of Bifidobacterium and Eubacterium by germinated barley foodstuff, accompanied by enhanced butyrate production in healthy volunteers. Int. J. Mol. Med 1999, 3, 175–9. [DOI] [PubMed] [Google Scholar]
- 166.Fujimori S; Gudis K; Mitsui K; Seo T; Yonezawa M; Tanaka S; Tatsuguchi A; Sakamoto C A randomized controlled trial on the efficacy of synbiotic versus probiotic or prebiotic treatment to improve the quality of life in patients with ulcerative colitis. Nutrition 2009, 25, 520–5. [DOI] [PubMed] [Google Scholar]
- 167.Hafer A; Krämer S; Duncker S; Krüger M; Manns MP; Bischoff SC Effect of oral lactulose on clinical and immunohistochemical parameters in patients with inflammatory bowel disease: a pilot study. BMC Gastroenterol. 2007, 7, 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Casellas F; Borruel N; Torrejón A; Varela E; Antolin M; Guarner F; Malagelada J-R Oral oligofructose-enriched inulin supplementation in acute ulcerative colitis is well tolerated and associated with lowered faecal calprotectin. Aliment. Pharmacol. Ther 2007, 25, 1061–7. [DOI] [PubMed] [Google Scholar]
- 169.Hanai H; Kanauchi O; Mitsuyama K; Andoh A; Takeuchi K; Takayuki I; Araki Y; Fujiyama Y; Toyonaga A; Sata M; et al. Germinated barley foodstuff prolongs remission in patients with ulcerative colitis. Int. J. Mol. Med 2004, 13, 643–7. [PubMed] [Google Scholar]
- 170.Hallert C; Björck I; Nyman M; Pousette A; Grännö C; Svensson H Increasing fecal butyrate in ulcerative colitis patients by diet: controlled pilot study. Inflamm. Bowel Dis 2003, 9, 116–21. [DOI] [PubMed] [Google Scholar]
- 171.Fernández-Bañares F; Hinojosa J; Sánchez-Lombraña JL; Navarro E; Martínez-Salmerón JF; García-Pugés A; González-Huix F; Riera J; González-Lara V; Domínguez-Abascal F; et al. Randomized clinical trial of Plantago ovata seeds (dietary fiber) as compared with mesalamine in maintaining remission in ulcerative colitis. Spanish Group for the Study of Crohn’s Disease and Ulcerative Colitis (GETECCU). Am. J. Gastroenterol 1999, 94, 427–33. [DOI] [PubMed] [Google Scholar]
- 172.Faghfoori Z; Shakerhosseini R; Navai L; Somi MH; Nikniaz Z; Abadi A Effects of an Oral Supplementation of Germinated Barley Foodstuff on Serum CRP Level and Clinical Signs in Patients with Ulcerative Colitis. Heal. Promot. Perspect 2014, 4, 116–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Kanauchi O; Mitsuyama K; Homma T; Takahama K; Fujiyama Y; Andoh A; Araki Y; Suga T; Hibi T; Naganuma M; et al. Treatment of ulcerative colitis patients by long-term administration of germinated barley foodstuff: multi-center open trial. Int. J. Mol. Med 2003, 12, 701–704. [PubMed] [Google Scholar]
- 174.Ghiamati Yazdi F; Soleimanian-Zad S; van den Worm E; Folkerts G Turmeric Extract: Potential Use as a Prebiotic and Anti-Inflammatory Compound? Plant Foods Hum. Nutr 2019, 74, 293–299. [DOI] [PubMed] [Google Scholar]
- 175.Hanai H; Iida T; Takeuchi K; Watanabe F; Maruyama Y; Andoh A; Tsujikawa T; Fujiyama Y; Mitsuyama K; Sata M; et al. Curcumin maintenance therapy for ulcerative colitis: randomized, multicenter, double-blind, placebo-controlled trial. Clin. Gastroenterol. Hepatol 2006, 4, 1502–6. [DOI] [PubMed] [Google Scholar]
- 176.Salli K; Anglenius H; Hirvonen J; Saarinen MT; Tiihonen K; Maukonen J; Ouwehand AC; Hibberd AA; Ahonen I The effect of 2’-fucosyllactose on simulated infant gut microbiome and metabolites; a pilot study in comparison to GOS and lactose. Sci. Rep 2019, 9, 13232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Sawin EA; De Wolfe TJ; Aktas B; Stroup BM; Murali SG; Steele JL; Ney DM Glycomacropeptide is a prebiotic that reduces Desulfovibrio bacteria, increases cecal short-chain fatty acids, and is anti-inflammatory in mice. Am. J. Physiol. Gastrointest. Liver Physiol 2015, 309, G590–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Welters CFM; Heineman E; Thunnissen FBJM; van den Bogaard AEJM; Soeters PB; Baeten CGMI Effect of dietary inulin supplementation on inflammation of pouch mucosa in patients with an ileal pouch-anal anastomosis. Dis. Colon Rectum 2002, 45, 621–7. [DOI] [PubMed] [Google Scholar]
- 179.Meijer HP; Welters CF; Heineman E; Salomons GS; Büller HA; Dekker J; Einerhand AW Enteral inulin does not affect epithelial gene expression and cell turnover within the ileoanal pouch. Dis. Colon Rectum 2000, 43, 1427–34. [DOI] [PubMed] [Google Scholar]
- 180.Roberfroid MB; Van Loo JAE; Gibson GR The Bifidogenic Nature of Chicory Inulin and Its Hydrolysis Products. J. Nutr 1998, 128, 11–19. [DOI] [PubMed] [Google Scholar]
- 181.Levenstein S; Prantera C; Luzi C; D’Ubaldi A Low residue or normal diet in Crohn’s disease: a prospective controlled study in Italian patients. Gut 1985, 26, 989–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Heaton KW; Thornton JR; Emmett PM Treatment of Crohn’s disease with an unrefined-carbohydrate, fibre-rich diet. Br. Med. J 1979, 2, 764–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Jones VA; Dickinson RJ; Workman E; Wilson AJ; Freeman AH; Hunter JO Crohn’s disease: maintenance of remission by diet. Lancet (London, England) 1985, 2, 177–80. [DOI] [PubMed] [Google Scholar]
- 184.De Preter V; Joossens M; Ballet V; Shkedy Z; Rutgeerts P; Vermeire S; Verbeke Phd K Metabolic profiling of the impact of oligofructose-enriched inulin in Crohn’s disease patients: a double-blinded randomized controlled trial. Clin. Transl. Gastroenterol 2013, 4, e30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.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. [DOI] [PubMed] [Google Scholar]
- 186.Ito M; Kimura M; Deguchi Y; Miyamori-Watabe A; Yajima T; Kan T Effects of transgalactosylated disaccharides on the human intestinal microflora and their metabolism. J. Nutr. Sci. Vitaminol. (Tokyo). 1993, 39, 279–88. [DOI] [PubMed] [Google Scholar]
- 187.Ramirez-Farias C; Slezak K; Fuller Z; Duncan A; Holtrop G; Louis P Effect of inulin on the human gut microbiota: stimulation of Bifidobacterium adolescentis and Faecalibacterium prausnitzii. Br. J. Nutr 2009, 101, 541–50. [DOI] [PubMed] [Google Scholar]
- 188.Moens F; De Vuyst L Inulin-type fructan degradation capacity of Clostridium cluster IV and XIVa butyrate-producing colon bacteria and their associated metabolic outcomes. Benef. Microbes 2017, 8, 473–490. [DOI] [PubMed] [Google Scholar]
- 189.Schroeder BO; Birchenough GMH; Ståhlman M; Arike L; Johansson MEV ; Hansson GC; Bäckhed F Bifidobacteria or Fiber Protects against Diet-Induced Microbiota-Mediated Colonic Mucus Deterioration. Cell Host Microbe 2018, 23, 27–40.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Hosono A; Ozawa A; Kato R; Ohnishi Y; Nakanishi Y; Kimura T; Nakamura R Dietary fructooligosaccharides induce immunoregulation of intestinal IgA secretion by murine Peyer’s patch cells. Biosci. Biotechnol. Biochem 2003, 67, 758–64. [DOI] [PubMed] [Google Scholar]
- 191.Gerber M Fiber and breast cancer: another piece of the puzzle--but still an incomplete picture. J. Natl. Cancer Inst 1996, 88, 857–858. [DOI] [PubMed] [Google Scholar]
- 192.Psyllium P Plantago ovata (psyllium). Altern. Med. Rev 2002, 7, 155–159. [PubMed] [Google Scholar]
- 193.Pittler MH; Schmidt K; Ernst E Adverse events of herbal food supplements for body weight reduction: systematic review. Obes. Rev 2005, 6, 93–111. [DOI] [PubMed] [Google Scholar]
- 194.Lee D; Albenberg L; Compher C; Baldassano R; Piccoli D; Lewis JD; Wu GD Diet in the pathogenesis and treatment of inflammatory bowel diseases. Gastroenterology 2015, 148, 1087–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Wu GD; Chen J; Hoffmann C; Bittinger K; Chen Y-Y; Keilbaugh SA; Bewtra M; Knights D; Walters WA; Knight R; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.David LA; Maurice CF; Carmody RN; Gootenberg DB; Button JE; Wolfe BE; Ling AV ; Devlin AS; Varma Y; Fischbach MA; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Levine A; Wine E; Assa A; Sigall Boneh R; Shaoul R; Kori M; Cohen S; Peleg S; Shamaly H; On A; et al. Crohn’s Disease Exclusion Diet Plus Partial Enteral Nutrition Induces Sustained Remission in a Randomized Controlled Trial. Gastroenterology 2019, 157, 440–450.e8. [DOI] [PubMed] [Google Scholar]
- 198.Yu J; Guo H; Xie J; Luo J; Li Y; Liu L; Ou S; Zhang G; Peng X The Alternate Consumption of Quercetin and Alliin in the Traditional Asian Diet Reshaped Microbiota and Altered Gene Expression of Colonic Epithelial Cells in Rats. J. Food Sci 2019, 84, 678–686. [DOI] [PubMed] [Google Scholar]
- 199.Benno Y; Endo K; Miyoshi H; Okuda T; Koishi H; Mitsuoka T Effect of rice fiber on human fecal microflora. Microbiol. Immunol 1989, 33, 435–40. [DOI] [PubMed] [Google Scholar]
- 200.Chiba M; Abe T; Tsuda H; Sugawara T; Tsuda S; Tozawa H; Fujiwara K; Imai H Lifestyle-related disease in Crohn’s disease: relapse prevention by a semi-vegetarian diet. World J. Gastroenterol 2010, 16, 2484–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Obih C; Wahbeh G; Lee D; Braly K; Giefer M; Shaffer ML; Nielson H; Suskind DL Specific carbohydrate diet for pediatric inflammatory bowel disease in clinical practice within an academic IBD center. Nutrition 2016, 32, 418–25. [DOI] [PubMed] [Google Scholar]
- 202.Ishikawa H; Matsumoto S; Ohashi Y; Imaoka A; Setoyama H; Umesaki Y; Tanaka R; Otani T Beneficial effects of probiotic bifidobacterium and galacto-oligosaccharide in patients with ulcerative colitis: a randomized controlled study. Digestion 2011, 84, 128–33. [DOI] [PubMed] [Google Scholar]
- 203.Furrie E; Macfarlane S; Kennedy A; Cummings JH; Walsh SV; O’Neil DA; Macfarlane GT Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: A randomised controlled pilot trial. Gut 2005, 54, 242–249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Paramsothy S; Paramsothy R; Rubin DT; Kamm MA; Kaakoush NO; Mitchell HM; Castaño-Rodríguez N Faecal Microbiota Transplantation for Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. J. Crohns. Colitis 2017, 11, 1180–1199. [DOI] [PubMed] [Google Scholar]
- 205.Fang H; Fu L; Wang J Protocol for Fecal Microbiota Transplantation in Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Biomed Res. Int 2018, 2018, 8941340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Bennet JD; Brinkman M Treatment of ulcerative colitis by implantation of normal colonic flora. Lancet (London, England) 1989, 1, 164. [DOI] [PubMed] [Google Scholar]
- 207.Costello SP; Hughes PA; Waters O; Bryant RV; Vincent AD; Blatchford P; Katsikeros R; Makanyanga J; Campaniello MA; Mavrangelos C; et al. Effect of Fecal Microbiota Transplantation on 8-Week Remission in Patients With Ulcerative Colitis: A Randomized Clinical Trial. JAMA 2019, 321, 156–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Ishikawa D; Sasaki T; Osada T; Kuwahara-Arai K; Haga K; Shibuya T; Hiramatsu K; Watanabe S Changes in Intestinal Microbiota Following Combination Therapy with Fecal Microbial Transplantation and Antibiotics for Ulcerative Colitis. Inflamm. Bowel Dis 2017, 23, 116–125. [DOI] [PubMed] [Google Scholar]
- 209.Lee SM; Donaldson GP; Mikulski Z; Boyajian S; Ley K; Mazmanian SK Bacterial colonization factors control specificity and stability of the gut microbiota. Nature 2013, 501, 426–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Wei Y; Gong J; Zhu W; Tian H; Ding C; Gu L; Li N; Li J Pectin enhances the effect of fecal microbiota transplantation in ulcerative colitis by delaying the loss of diversity of gut flora. BMC Microbiol. 2016, 16, 255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Damman CJ; Brittnacher MJ; Westerhoff M; Hayden HS; Radey M; Hager KR; Marquis SR; Miller SI; Zisman TL Low Level Engraftment and Improvement following a Single Colonoscopic Administration of Fecal Microbiota to Patients with Ulcerative Colitis. PLoS One 2015, 10, e0133925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Vermeire S; Joossens M; Verbeke K; Wang J; Machiels K; Sabino J; Ferrante M; Van Assche G; Rutgeerts P; Raes J Donor Species Richness Determines Faecal Microbiota Transplantation Success in Inflammatory Bowel Disease. J. Crohns. Colitis 2016, 10, 387–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Herfarth H; Barnes EL; Long MD; Isaacs KL; Leith T; Silverstein M; Gerardin Y; Kassam Z Combined Endoscopic and Oral Fecal Microbiota Transplantation in Patients with Antibiotic-Dependent Pouchitis: Low Clinical Efficacy due to Low Donor Microbial Engraftment. Inflamm. Intest. Dis 2019, 4, 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Stallmach A; Lange K; Buening J; Sina C; Vital M; Pieper DH Fecal Microbiota Transfer in Patients With Chronic Antibiotic-Refractory Pouchitis. Am. J. Gastroenterol 2016, 111, 441–3. [DOI] [PubMed] [Google Scholar]
- 215.Wei Y; Zhu W; Gong J; Guo D; Gu L; Li N; Li J Fecal Microbiota Transplantation Improves the Quality of Life in Patients with Inflammatory Bowel Disease. Gastroenterol. Res. Pract 2015, 2015, 517597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Vaughn BP; Vatanen T; Allegretti JR; Bai A; Xavier RJ; Korzenik J; Gevers D; Ting A; Robson SC; Moss AC Increased Intestinal Microbial Diversity Following Fecal Microbiota Transplant for Active Crohn’s Disease. Inflamm. Bowel Dis 2016, 22, 2182–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Cui B; Feng Q; Wang H; Wang M; Peng Z; Li P; Huang G; Liu Z; Wu P; Fan Z; et al. Fecal microbiota transplantation through mid-gut for refractory Crohn’s disease: safety, feasibility, and efficacy trial results. J. Gastroenterol. Hepatol 2015, 30, 51–8. [DOI] [PubMed] [Google Scholar]
- 218.Suskind DL; Brittnacher MJ; Wahbeh G; Shaffer ML; Hayden HS; Qin X; Singh N; Damman CJ; Hager KR; Nielson H; et al. Fecal microbial transplant effect on clinical outcomes and fecal microbiome in active Crohn’s disease. Inflamm. Bowel Dis 2015, 21, 556–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Goyal A; Yeh A; Bush BR; Firek BA; Siebold LM; Rogers MB; Kufen AD; Morowitz MJ Safety, Clinical Response, and Microbiome Findings Following Fecal Microbiota Transplant in Children With Inflammatory Bowel Disease. Inflamm. Bowel Dis 2018, 24, 410–421. [DOI] [PubMed] [Google Scholar]
- 220.Li P; Zhang T; Xiao Y; Tian L; Cui B; Ji G; Liu Y-Y; Zhang F 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, 349–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Li SS; Zhu A; Benes V; Costea PI; Hercog R; Hildebrand F; Huerta-Cepas J; Nieuwdorp M; Salojärvi J; Voigt AY; et al. Durable coexistence of donor and recipient strains after fecal microbiota transplantation. Science 2016, 352, 586–9. [DOI] [PubMed] [Google Scholar]
- 222.Ott SJ; Waetzig GH; Rehman A; Moltzau-Anderson J; Bharti R; Grasis JA; Cassidy L; Tholey A; Fickenscher H; Seegert D; et al. Efficacy of Sterile Fecal Filtrate Transfer for Treating Patients With Clostridium difficile Infection. Gastroenterology 2017, 152, 799–811.e7. [DOI] [PubMed] [Google Scholar]
- 223.Ding X; Li Q; Li P; Zhang T; Cui B; Ji G; Lu X; Zhang F Long-Term Safety and Efficacy of Fecal Microbiota Transplant in Active Ulcerative Colitis. Drug Saf. 2019, 42, 869–880. [DOI] [PubMed] [Google Scholar]
- 224.Goyal Alka, Chu Andrew, Calabro Kathleen, Firek Brian, Bush Brian, M.M. 663 SAFETY AND EFFICACY OF FECAL MICROBIOTA TRANSPLANT IN CHILDREN WITH INFLAMMATORY BOWEL DISEASE. J. Pediatr. Gastroenterol. Nutr 2016, 63, S212. [Google Scholar]
- 225.Uygun A; Ozturk K; Demirci H; Oger C; Avci IY; Turker T; Gulsen M Fecal microbiota transplantation is a rescue treatment modality for refractory ulcerative colitis. Medicine (Baltimore). 2017, 96, e6479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.DeFilipp Z; Bloom PP; Torres Soto M; Mansour MK; Sater MRA; Huntley MH; Turbett S; Chung RT; Chen Y-B; Hohmann EL Drug-Resistant E. coli Bacteremia Transmitted by Fecal Microbiota Transplant. N. Engl. J. Med 2019, NEJMoa1910437. [DOI] [PubMed] [Google Scholar]
- 227.Jacob V; Crawford C; Cohen-Mekelburg S; Viladomiu M; Putzel GG; Schneider Y; Chabouni F; OʼNeil S; Bosworth B; Woo V; et al. Single Delivery of High-Diversity Fecal Microbiota Preparation by Colonoscopy Is Safe and Effective in Increasing Microbial Diversity in Active Ulcerative Colitis. Inflamm. Bowel Dis 2017, 23, 903–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Turnbaugh PJ; Hamady M; Yatsunenko T; Cantarel BL; Ley RE; Sogin ML; Jones WJ; Roe B. a ; Jason P; Egholm M; et al. A core gut microbiome between lean and obesity twins. Nature 2009, 457, 222–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Hedin C; van der Gast CJ; Rogers GB; Cuthbertson L; McCartney S; Stagg AJ; Lindsay JO; Whelan K Siblings of patients with Crohn’s disease exhibit a biologically relevant dysbiosis in mucosal microbial metacommunities. Gut 2016, 65, 944–53. [DOI] [PubMed] [Google Scholar]
- 230.Siegmund B Is intensity the solution for FMT in ulcerative colitis? Lancet (London, England) 2017, 389, 1170–1172. [DOI] [PubMed] [Google Scholar]
- 231.Lupp C; Robertson ML; Wickham ME; Sekirov I; Champion OL; Gaynor EC; Finlay BB Host-mediated inflammation disrupts the intestinal microbiota and promotes the overgrowth of Enterobacteriaceae. Cell Host Microbe 2007, 2, 119–29. [DOI] [PubMed] [Google Scholar]
- 232.Ratner M Microbial cocktails join fecal transplants in IBD treatment trials. Nat. Biotechnol 2015, 33, 787–788. [DOI] [PubMed] [Google Scholar]
- 233.Faith JJ; Ahern PP; Ridaura VK; Cheng J; Gordon JI Identifying gut microbe-host phenotype relationships using combinatorial communities in gnotobiotic mice. Sci. Transl. Med 2014, 6, 220ra11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Foligne B; Nutten S; Grangette C; Dennin V; Goudercourt D; Poiret S; Dewulf J; Brassart D; Mercenier A; Pot B Correlation between in vitro and in vivo immunomodulatory properties of lactic acid bacteria. World J. Gastroenterol 2007, 13, 236–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Peña JA; Rogers AB; Ge Z; Ng V; Li SY; Fox JG; Versalovic J Probiotic Lactobacillus spp. diminish Helicobacter hepaticus-induced inflammatory bowel disease in interleukin-10-deficient mice. Infect. Immun 2005, 73, 912–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Peran L; Camuesco D; Comalada M; Nieto A; Concha A; Diaz-Ropero MP; Olivares M; Xaus J; Zarzuelo A; Galvez J Preventative effects of a probiotic, Lactobacillus salivarius ssp. salivarius, in the TNBS model of rat colitis. World J. Gastroenterol 2005, 11, 5185–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Alard J; Peucelle V; Boutillier D; Breton J; Kuylle S; Pot B; Holowacz S; Grangette C New probiotic strains for inflammatory bowel disease management identified by combining in vitro and in vivo approaches. Benef. Microbes 2018, 9, 317–331. [DOI] [PubMed] [Google Scholar]
- 238.Yan F; Cao H; Cover TL; Washington MK; Shi Y; Liu L; Chaturvedi R; Peek RM; Wilson KT; Polk DB Colon-specific delivery of a probiotic-derived soluble protein ameliorates intestinal inflammation in mice through an EGFR-dependent mechanism. J. Clin. Invest 2011, 121, 2242–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Segawa S; Fujiya M; Konishi H; Ueno N; Kobayashi N; Shigyo T; Kohgo Y Probiotic-derived polyphosphate enhances the epithelial barrier function and maintains intestinal homeostasis through integrin-p38 MAPK pathway. PLoS One 2011, 6, e23278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.von Schillde M-A; Hörmannsperger G; Weiher M; Alpert C-A; Hahne H; Bäuerl C; van Huynegem K; Steidler L; Hrncir T; Pérez-Martínez G; et al. Lactocepin Secreted By Lactobacillus Exerts Anti-Inflammatory Effects By Selectively Degrading Proinflammatory Chemokines. Cell Host Microbe 2012, 11, 387–396. [DOI] [PubMed] [Google Scholar]
- 241.Quévrain E; Maubert MA; Michon C; Chain F; Marquant R; Tailhades J; Miquel S; Carlier L; Bermúdez-Humarán LG; Pigneur B; et al. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut 2016, 65, 415–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Li HB; Chen MY; Qiu ZW; Cai QQ; Li DT; Tang HM; Chen XL Efficacy and safety of Kangfuxin liquid combined with aminosalicylic acid for the treatment of ulcerative colitis: A systematic review and meta-analysis. Med. (United States) 2018, 97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Sham HP; Bazett M; Bosiljcic M; Yang H; Luk B; Law HT; Morampudi V; Yu HB; Pankovich J; Sutcliffe S; et al. Immune Stimulation Using a Gut Microbe-Based Immunotherapy Reduces Disease Pathology and Improves Barrier Function in Ulcerative Colitis. Front. Immunol 2018, 9, 2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Bressler B; Bethel KP; Kleef R; Reynolds SL; Sutcliffe S; Mullins DW; Gunn H Site-Specific Immunomodulator: A Novel Treatment for Crohn’s Disease. Gastroenterol. Res. Pract 2015, 2015, 231243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Sutcliffe S; Kalyan S; Pankovich J; Chen JMH; Gluck R; Thompson D; Bosiljcic M; Bazett M; Fedorak RN; Panaccione R; et al. Novel Microbial-Based Immunotherapy Approach for Crohn’s Disease. Front. Med 2019, 6, 170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Zhu W; Winter MG; Byndloss MX; Spiga L; Duerkop BA; Hughes ER; Büttner L; de Lima Romão E; Behrendt CL; Lopez CA; et al. Precision editing of the gut microbiota ameliorates colitis. Nature 2018, 553, 208–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Kernbauer E; Ding Y; Cadwell K An enteric virus can replace the beneficial function of commensal bacteria. Nature 2014, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Gogokhia L; Buhrke K; Bell R; Hoffman B; Brown DG; Hanke-Gogokhia C; Ajami NJ; Wong MC; Ghazaryan A; Valentine JF; et al. Expansion of Bacteriophages Is Linked to Aggravated Intestinal Inflammation and Colitis. Cell Host Microbe 2019, 25, 285–299.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Charlet R; Pruvost Y; Tumba G; Istel F; Poulain D; Kuchler K; Sendid B; Jawhara S Remodeling of the Candida glabrata cell wall in the gastrointestinal tract affects the gut microbiota and the immune response. Sci. Rep 2018, 8, 3316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Kunyeit L; Kurrey NK; Anu-Appaiah KA; Rao RP Probiotic Yeasts Inhibit Virulence of Non-albicans Candida Species. MBio 2019, 10, 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Mohamadzadeh M; Pfeiler EA; Brown JB; Zadeh M; Gramarossa M; Managlia E; Bere P; Sarraj B; Khan MW; Pakanati KC; et al. Regulation of induced colonic inflammation by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc. Natl. Acad. Sci. U. S. A 2011, 108 Suppl, 4623–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Zhang B; Liu Y; Lan X; Xu X; Zhang X; Li X; Zhao Y; Li G; Du C; Lu S; et al. Oral Escherichia coli expressing IL-35 meliorates experimental colitis in mice. J. Transl. Med 2018, 16, 71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Riglar DT; Giessen TW; Baym M; Kerns SJ; Niederhuber MJ; Bronson RT; Kotula JW; Gerber GK; Way JC; Silver PA Engineered bacteria can function in the mammalian gut long-term as live diagnostics of inflammation. Nat. Biotechnol 2017, 35, 653–658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Wells JM; Mercenier A Mucosal delivery of therapeutic and prophylactic molecules using lactic acid bacteria. Nat. Rev. Microbiol 2008, 6, 349–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Mackie RI; Sghir A; Gaskins HR Developmental microbial ecology of the neonatal gastrointestinal tract. Am. J. Clin. Nutr 1999, 69, 1035S–1045S. [DOI] [PubMed] [Google Scholar]