Review
Copyright ©The Author(s) 2017.
World J Gastroenterol. Sep 28, 2017; 23(36): 6593-6627
Published online Sep 28, 2017. doi: 10.3748/wjg.v23.i36.6593
Table 1 Summary of the literature findings about anti-inflammatory therapies in irritable bowel syndrome
DrugRef.No. of patientsStudy designOutcome
Corticosteroids (prednisolone)Dunlop et al[24]29 patients with post-infectious irritable bowel syndromeRandomized, double-blind, placebo-controlled trial of 3 wk of oral prednisolone, 30 mg/dNot associated with any significant treatment-related improvement in abdominal pain, diarrhoea, frequency or urgency
Antibiotics (Rifaximin)Pimentel et al[29]623 IBS patients in TARGET 1 and 637 IBS in TARGET 2Phase 3 trials, 14 d with rifaximin 550 mg 3 times dailySignificantly increased the percentage of relief of global IBS symptoms and improved IBS-related bloating and abdominal pain, discomfort, and loose or watery stools, with regard to placebo for up to 10 wk post-treatment
Target 1 e 2
Antibiotics (norfloxacin)Ghoshal et al[32]80 IBS patients evaluate for SIBORandomized, double-blind, placebo-controlled trial; patients were randomized to 800 mg/d norfloxacin for 10 d or placeboAlthough norfloxacin was more effective at reducing the symptom score at 1 mo among patients with compared with those without SIBO but not placebo, the scores were comparable at 6 mo. Symptoms more often resolved to turn Rome III negative in SIBO patients treated with norfloxacin compared with placebo at 1 mo
Mast cell stabilizers (Ketotifen)Klooker et al[33]60 IBS patiensCase Control study; abarostat study to assess rectal sensitivity before and after 8 wk of treatment and, after the initial barostat, patients were randomised to receive ketotifen or placeboKetotifen but not placebo increased the threshold for discomfort in patients with IBS with visceral hypersensitivity, but this effect was not observed in normosensitive patients with IBS. Ketotifen significantly decreased abdominal pain and other IBS symptoms and improved quality of life
Mast cells stabilizers (DSCG)Lobo et al[34]Randomized, double-blind, placebo-controlled trial; with prolonged (6 mo) oral administration of DSCGInduces Mast Cell-Mediated Recovery of Healthy-Like Innate Immunity Genes Expression Profile in the Jejunal Mucosa
Mast cells stabilizers (ebastin)Wouters et al[35]65 IBS patientsDouble-blind placebo-controlled trial, after 2-wk run-in period, subjects were assigned randomly to groups ebastine (20 mg/d; n = 28) or placebo (n = 27) for 12 wkCompared with subjects given placebo, those given ebastine had reduced visceral hypersensitivity, increased symptom relief, and reduced abdominal pain scores
MesalazineBarbara et al[39]185 patients with IBSA phase 3, multicentre, tertiary setting, randomised, double-blind, placebo-controlled trial in patients with Rome III confirmed IBS. Patients were randomly assigned to either mesalazine, 800 mg, or placebo, three times daily for 12 wk, and were followed for additional 12 wkMesalazine treatment was not superior than placebo on the study primary endpoint, but a subgroup of patients with IBS showed a sustained therapy response and benefits from a mesalazine therapy
Lam et al[40]136 patients with IBS-DA double-blind, randomised placebo-controlled trial of 2 g mesalazine twice daily compared with placebo for 3 moThe authors concluded that mesalazine did not improve abdominal pain, stool consistency or percentage with satisfactory relief compared with placebo during the last 2 weeks’ follow-up, however a post hoc analysis in 13 post-infectious patients with IBS appeared to show benefit but this needs confirmation in a larger group[40]
Table 2 Summary of the literature findings about therapies restoring intestinal permeability in irritable bowel syndrome
DrugRef.No. of patientsStudy designOutcome
ProbioticsFord et al[61]Forty-three RCTs were eligible for inclusionMetanalysisProbiotics had beneficial effects on global IBS, abdominal pain, bloating, and flatulence scores. Data for prebiotics and synbiotics in IBS were sparse. Probiotics appeared to have beneficial effects in CIC (mean increase in number of stools per week = 1.49; 95%CI: 1.02-1.96), but there were only two RCTs. Synbiotics also appeared beneficial (RR of failure to respond to therapy = 0.78; 95%CI: 0.67-0.92). Again, trials for prebiotics were few in number, and no definite conclusions could be drawn
Mazurak et al[67]Fifty-six papersMetanalysisThe heterogeneity of the studies of probiotics in IBS questions the value of meta-analyses and the use of different bacterial strains and different mixtures of these strains, as well as different dosages, are the main contributors to this heterogeneity
GlutamineAkobeng et al[73]Two randomized trialCochrane analysisNot significant difference in the permeability and no effect in the clinical remission
Larazotide acetateLeffler et al[78]342 adults with celiac disease who had been on a gluten free diet (GFD) for 12 mo or longer and maintained their current GFD during the studyRandomized, double-blind, placebo-controlled study assessed larazotide acetate 0.5, 1, or 2 mg 3 times dailyReduce signs and symptoms in celiac disease patients on a GFD better than a GFD alone
Table 3 Summary of the literature findings about irritable bowel syndrome-C therapies
DrugRef.No. of patientsStudy designOutcome
LinaclotideAndresen et al[82]36 women with IBS-CPhase IIa randomized, double-blind, placebo- controlled trial. Patients were randomized in a 1:1:1 fashion to placebo, linaclotide 100 μg, and linaclotide 1000 μg and was evaluated the effect of 5 dNo treatment effects were seen for gastric emptying or colonic filling with linaclotide. Significant treatment effects were found for ascending colon emptying t½ times (P = 0.015) and overall total colonic transit times at 48 h (P = 0.02), for the 1000 μg dose (P = 0.004) but not the 100 μg dose, as well as overall treatment effects on increased stool frequency, decreased stool consistency, improved ease of passage, and acceleration of time to first bowel movement (P < 0.001)[82]
Johnston et al[83]420 patients with IBS-CPhase IIb randomized, double-blind, parallel-group, multicenter, placebo-controlled trial evaluate 12 wk of linaclotide at a daily dose range of 75-600 μgCompared with placebo, all doses of linaclotide significantly improved bowel habits, including frequency of SBMs and CSBMs, severity of straining, stool consistency, as well as abdominal pain scores. Abdominal discomfort, bloating, and global IBS-C measures were also improved, for all doses except for the 75 μg (abdominal discomfort) and 150 μg dose (bloating). Effects were present for the first week, and sustained throughout the 12 wk of treatment
Chey et al[84]804 adults with IBS-CPhase III trials randomized, double-blind, placebo-controlled to receive linaclotide 290 lg or placebo daily for 26 wk, with change-from-baseline end points measuredat 12 and 26 wkOver 12 wk, the FDA combined primary end point was achieved by 33.7% of patients receiving linaclotide compared with 13.9% of patients receiving placebo (P < 0.0001)
Videlock et al[109]7 trials of linaclotide in patients with IBS-C or CCA meta-analysis from MEDLINE, EMBASE, and the Cochrane central register of controlled trials were searched for randomized, placebo-controlled trialsThe NNT for the primary endpoint of these trials (3 SCBMs/wk and an increase of C1 SCBM/wk, for 75% of weeks) was 7 (95%CI: 5-8)
Rao et al[86]803 adults with IBS-CPhase III trials randomized, double-blind, placebo-controlled to receive linaclotide 290 lg or placebo once/d for 12 wkLinaclotide demonstrated statistically significant improvements in all primary and secondary efficacy end points compared with placebo Severity of straining, constipation, and stool consistency also improved in the linaclotide group compared with the placebo group
PlecanatideMiner et al[92]951 patients with CICPhase III, randomized, double-blind trial, received plecanatide 0.3, 1 or 3 mg, or placebo once/d for 12 wkThe proportion of overall responders was significantly greater with plecanatide 3 mg compared with placebo (19% vs 10.7%, P = 0.009); weekly responder rates were also significantly greater for plecanatide 3 mg than placebo for weeks 1-12. Improvements in stool frequency, consistency, straining, and quality of life were also noted with the 3-mg dose vs placebo. Data for other plecanatide doses were not reported
PrucaloprideQuigley et al[88]620 patients with CCA double-blind, placebo-controlled study. Patients receiving 2 or 4 mg of prucalopride for 12 wkIncreased one or more CSBMs per week compared to patients in the control group
Camilleri et al[98]713 patients with CCA double-blind, placebo-controlled study. Patients receiving 2 or 4 mg of prucalopride for 12 wkIncreased frequency of three or more CSBMs per week, and improved evacuation completeness, perceived disease severity, and quality of life
Müller-Lissner et al[97]Elderly patients aged 65 years and older with CCA double-blind, placebo-controlled studyNo changes in electrocardiogram or corrected QT (QTc) interval were reported, indicating its safety for the treatment of CC in the elderly
Patients receiving 2 or 4 mg of prucalopride for 12 wk
Ke et al[102]4 Randomized, Placebo-controlled StudiesA Pooled AnalysisSafe and well-tolerated It was also effective in improving the abdominal symptoms of CC such as abdominal discomfort, bloating, straining, and painful bowel movements
YKP10811Shin et al[114]55 patientsA single-center, randomized, parallel-group, double-blinded, placebo-controlled study were assigned randomly to groups given YKP10811 10 mg (n = 15), 20 mg (n = 16), 30 mg (n = 15), or placebo (n = 11) daily for 8 dEnhanced gastrointestinal and colonic transit and improved bowel function during an 8-d treatment trial. In general, the 10-mg and 20-mg doses were the most effective in accelerating colonic transit. No serious adverse events were observed
RenzaprideCamilleri et al[101]46 women with IBS-CIn a phase II studyRenzapride 4 mg q.d. accelerated colonic transit and increased ascending colon emptying vs placebo
George et al[130]510 patientsMulticentre, randomized, placebo-controlled, double-blind study men and women were randomized to placebo or renzapride (1, 2 or 4 mg/d) for 12 wk4 mg renzapride q.d. in terms of improving frequency of bowel movements and stool consistency
Ford et al[131]29 RCTs were eligible for inclusionMeta-analysis of placebo-controlled clinicalRenzapride and cisapride were not more effective than placebo in IBS patients
Mozaffari et al[133]2528 C-IBS and non C-, non D-IBS patientsMeta-analysis from randomized placebo-controlled clinical trialsRenzapride has no significant advantage over placebo in relieving symptoms in IBS patients
VelusetragManini et al[115]60 healthy volunteersPhase II clinical trials, pt were randomly assigned, in double-blind fashion, to placebo, 5, 15, 30 or 50 mg velusetrag, with transit measurements after single and 6-d dosingSingle doses of velusetrag (30 and 50 mg), but not placebo, accelerated colonic transit, as measured by colonic filling at 6 h and geometric center at 24 h
Goldberg et al[140]401 subjects with CCIn a Phase II randomized, double-blind, placebo-controlled trialShort bowel movement (SBM) frequency, complete SBM and other associated symptoms with CC were significantly improved in comparison with placebo in patients who received velusetrag for 4 wk
NaronaprideDennis et al[149]210 patients with CC.Phase II, randomized, double-blind, placebo-controlled, dose definition study (orally 20, 40, 80 and 120 mg twice a day) to evaluate the clinical effects of 9 days’There were borderline effects on gastric emptying at half-time; however, ATI-7505 accelerated colonic transit at 24 h and ascending colonic emptying
Chenodeoxycolic acidOdunsi-Shiyanbade et al[155]60 healthy volunteersRandomized controlled trial, CDCA 500 mg and 1000 mg given for 4 dSignificant increases in stool frequency, decreases in stool consistency, and improvements in ease of stool passage were reported with CDCA
Rao et al[153]36 female patientsDouble-blind placebo-controlled studyAccelerated colonic transit and improved bowel function
ElobixibatSimrén et al[162]30 patientsDose-finding randomized trial five dose-levels (range: 0.1-10 mg/d) or to placeboIncreased C4, reduced LDL cholesterol and increased colonic transit from 3 to 1.9 d, and increased the number of SBM and CSBM/W in patients with CIC
Chey et al[157]190 patientsWere randomized to 5, 10, or 15 mg A3309 or placebo once daily. 8-wk, multicenter, randomized, double-blind, placebo-controlled, parallel group, phase IIb study,A3309 increased stool frequency and improved constipation-related symptoms in CIC; effects were maintained over 8 wk of treatment
LubiprostoneDrossman et al[173]1171 patients in totalTwo double-blind, randomized, multicenter, placebo-controlled phase III clinical trials,, a 12-wk randomized 2:1 to receive either lubiprostone 8 lg or matching placebo twice/d with food; a 36-wk open-label extension studyLubiprostone was superior to placebo in the primary end point of overall responders, greater improvements in all secondary outcome measures compared with placebo
Chey et al[174]
Johanson et al[175]479 patients in totalTwo 4-wk phase III, randomized, double-blind, placebo-controlled, multicenter clinical trialsPatients treated with lubiprostone had a statistical higher frequency of SBMs during the first week of treatment compared with placebo
Barish et al[82]
Hyman et al[176]127 pediatric patients with CIC (mean age 10.2 yr)An open-label 4-wk clinical trialDemonstrated that lubiprostone was efficacious and well tolerated at total daily doses of 12-48 lg
AZD1722Rosenbaum et al[185]181 patients with IBS-CPhase IIa In a IIa double-blind, randomized placebo-controlled study, Tenapanor was given orally at the doses of 10, 30 and 100 mg once daily for 4 consecutive weeks with 2 wk follow-upThe primary end point [change in complete spontaneous bowel movements (CSBM) from baseline to week 4] was not met in this study and the incidence of diarrhea was comparable with placebo group. However, improvement in bloating and abdominal pain was noted in IBS-C patients
Rosenbaum et al[186]371 IBS-C patientsPhase IIb In a IIb randomized, double-blind, placebo-controlled, multicenter studyThe overall responder was met in 50.0% of tenapanor-treated group (50 mg) vs 23.6% for placebo (after 12 wk). After 12 wk, adequate relief in IBS-C symptoms was observed in 63.1% of tenapanor-treated group (50 mg twice daily) vs 39.3% in placebo
Rosenbaum et al[187]356 patientsA double-blind, placebocontrolled, randomized phase 2b trial 12-wk dose-ranging study evaluating tenapanor 5 mg, 20 mg or 50 mg b.i.d. vs placebo (1/2)Tenapanor 50 mg b.i.d. significantly improved CSBM responder rate (primary endpoint) compared with placebo in patients with IBS-C. Tenapanor 50 mg b.i.d. also improved key secondary endpoints compared with placebo, including overall responder rate, abdominal pain responder rate and stool frequency. In addition, improvements were observed in several exploratory endpoints addressing a range of symptoms in patients with IBS-C. Tenapanor was generally well tolerated and had minimal systemic availability. Tenapanor shows promise as a future treatment option for patients with IBS-C
Neu p11 (piromalatine)Camilleri et al[202]40 IBS patientsIn double blind placebo controlled study were randomly assigned to receive either melatonin 3 mg (n = 20) or matching placebo (n = 20) at bedtime for two weeksMelatonin 3 mg at bedtime for two weeks significantly attenuated abdominal pain and reduced rectal pain sensitivity without improvements in sleep disturbance or psychological distress. The findings suggest that the beneficial effects of melatonin On abdominal pain in IBS patients with sleep disturbances are independent of its action on sleep disturbances or psychological profiles
Table 4 Summary of the literature findings about irritable bowel syndrome-D therapies
DrugRef.No. of ptStudy designOutcomes
RamosetronLee et al[213]343 male ptA multicenter, randomized, open-label trial male patients with IBS-D; pt were randomized to either a 4-wk treatment of ramosetron 5 mg once daily ,or a 4-wk treatment of mebeverine 135 mg three times dailyGlobal IBS symptoms, abdominal pain/discomfort and abnormal bowel habits in the ramosetron and mebeverine groups significantly increased during the treatment period. The severity scores of abdominal pain/discomfort and urgency, the stool form score, and the stool frequency in both treatment arms were significantly reduced, compared with the baselines
Fukudo et al[222]296 male ptA randomized, double-blind, placebo-controlled trial in male patients with IBS-D Patients were given 5 mg oral ramosetron (n = 147) or placebo (n = 149) once daily for 12 wk after a 1-wk baseline periodImproving stool consistency in the first month. The ramosetron group had significantly higher monthly rates of relief of overall IBS symptoms and IBS-related quality of life than the placebo group. Adverse events occurring in 46.9% and 51.7% of ramosetron and placebo patients, respectively
Fukudo et al[223]576 female ptA randomized, double-blind, placebo-controlled trial. The subjects received either 2.5 μg ramosetron or placebo once daily for 12 wk.Global improvement, an increased stool consistency a significant reductions in abdominal pain and discomfort and greater improvement in QOL compared with placebo
Lx1031Brown et al[229]155 patientsA phase-II multicenter, randomized, double-blind, placebo-controlled, the subjects were assigned randomly in a double-blind fashion to 1 of 2 doses of LX1031 (250 mg 4 times/d or 1000 mg 4 times/d) or placebo, taken daily during the 28-d treatment periodImproved significantly in patients given 1000 mg LX1031 compared with those given placebo, at week 1, together with nonsignificant improvements at weeks 2, 3 and 4. Adverse Effects reported were generally mild, self-limited, and evenly distributed across the placebo and both LX1031 treatment arms
ASP-7147Lembo et al[230]64 patientsRCT performed on during a 4-wkDemonstrating improvement in multiple symptoms of IBS-D. The persistence of treatment effect suggests the possibility of retained efficacy with less frequent dosing in follow-on trials
JNJ-27018966[232]807 patientsA randomized, controlled, double-blind study, 25, 100, and 200 mg twice daily to placeboThe composite of diarrhea and pain was significantly improved in the JNJ-27018966 25 and 200 mg twice-daily groups compared to placebo
ROSE-010Hellström et al[233]160 patientsA randomized placebo-controlled trial. Patients were randomized to ROSE-010 100 μg once daily, 300 μg once daily or placeboTreatment with ROSE-010 resulted in a two fold greater response to abdominal pain compared to placebo and significantly greater patient-reported satisfaction with ROSE-010. The most common treatment-related adverse effect was nausea
AST-120Tack et al[95]115 non-constipation-related IBS patientsA randomized, double-blind, controlled studyAST-120 2 g three times daily significantly improved the proportion of patients with at least a 50% reduction in the number of days with abdominal pain compared to placebo. AST-120 resulted in significantly improved bloating and numerically improved stool consistency compared to placebo. The safety profile AST-120 was similar to placebo
IbodutantTrinkley et al[231]559 IBS-D patientsA randomized, double-blind, controlled trialImproved abdominal pain, satisfactory relief of overall symptoms, and quality of life compared to placebo. All three doses of ibodutant (1, 3, 10 mg once daily) were superior to placebo, but 10 mg once daily was most effective and females responded better than males
AsimadolineTrinkley et al[231]596 IBS-D patientsA randomized, controlled, double-blind trial compared asimadoline 0.15, 0.5 and 1 mg twice daily to placeboAsimadoline 0.5 mg twice daily significantly improved by two fold the total number of months with adequate relief of IBS pain, pain scores, urgency and frequency
ColesevelamOdunsi-Shiyanbade et al[155]12 IBS-D patientsSingle center trialColesevelam modestly affected overall colonic transit (emptying of the ascending colon took an average 4 h longer in patients given colesevelam compared to placebo). Furthermore, colesevelam was associated with greater ease of stool passage and somewhat firmer stool consistency. No effects on mucosal permeability or safety were identified
Camilleri et al[240]12 IBS-D patientsA 10-d single-center, unblinded, single-dose trialColesevelam increases delivery of BAs to stool while improving stool consistency, and increases hepatic BA synthesis, avoiding steatorrhea in patients with IBS-D
SolinefacinFukushima et al[249]20 IBS-D patientsAn open-label trial. After a 2-wk observation period, all participants received solifenacin for 6 wk. Subsequently, the administration of solifenacin was discontinued and ramosetron, a serotonin 3 receptor antagonist, was administered for 4 wkThe efficacy of solifenacin in the treatment of IBS with diarrhea was not inferior to that of ramosetron
TiropamideLee et al[251]287 patientsA multicenter, randomized, non-inferiority Patients randomly allocated to either tiropramide 100 mg or octylonium 20 mg t.i.d (means 3 times a day) for 4 wkTiropamide led to symptom improvement in terms of total symptom scores for 4 wk, compared with 3 wk in the placebo group; in addition, at week 4 abdominal pain was only improved in the tiropramide group. The incidence of adverse events was similar in the 2 groups, and no severe adverse events involving either drug were observed