Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Dec 1.
Published in final edited form as: Mayo Clin Proc. 2018 Dec;93(12):1858–1872. doi: 10.1016/j.mayocp.2018.04.032

Management Options for Irritable Bowel Syndrome

Michael Camilleri 1,
PMCID: PMC6314474  NIHMSID: NIHMS1505842  PMID: 30522596

Abstract

Irritable bowel syndrome (IBS) is associated with diverse pathophysiological mechanisms. These include increased abnormal colonic motility or transit, intestinal or colorectal sensation, increased colonic bile acid concentration, superficial colonic mucosal inflammation, as well as epithelial barrier dysfunction, neurohormonal upregulation and activation of secretory processes in the epithelial layer. Novel approaches to treatment include lifestyle modification, changes in diet, probiotics and pharmacotherapy directed to the motility, sensation and intraluminal milieu of patients with IBS. Despite recent advances, there is a need for developing new treatments to relieve pain in IBS without deleterious central or other adverse effects.

Introduction

Irritable bowel syndrome (IBS) pain, diarrhea or constipation result from one or more pathophysiological mechanisms in each individual patient. IBS treatment typically addresses the predominant symptom experienced by the patient and targets the pathophysiology, such as accelerated transit or visceral hypersensitivity. There are still no effective disease-modifying treatments;1 however, research that has demonstrated and validated biomarkers based on the pathophysiology of IBS provides opportunities to direct effective treatments to correct those mechanisms, such as abnormalities of colonic transit or increased colonic concentrations of bile acids.2,3 Novel therapeutic approaches that have targeted these abnormalities in single-center, randomized, controlled trials (RCTs) using biomarker endpoints have correctly predicted therapeutic efficacy based on symptom-based endpoints in phase 2B or 3 multicenter RCTs.3

Visceral pain is a hallmark of IBS. Pain is transmitted to conscious perception in the brain via a three neuron chain, as with somatic pain; the main pathways are vagal, thoracolumbar and lumbosacral afferents that have both pro- and anti-nociceptive ion channels and receptors.4 There are several relevant neurotransmitters on the afferents conveying sensory signals to the central nervous system, including serotonin (5-HT) and neurokinins, as well as ion channels including transient receptor potential (TRP) channels that mediate activation of afferent nerves and detect thermal and chemical stimuli that produce acute or persistent pain.5

This article addresses the current approaches to treatment of IBS, including lifestyle modifications, changes in diet, alternative and herbal therapies, probiotics and pharmacotherapy (Figure 1)6 directed to the motility, sensation and intraluminal milieu of patients with IBS. There are recent, national societal guidelines for the management of IBS based on the available literature and systematic reviews and meta-analyses.7-10 With recently introduced medications, trials have used Food and Drug Administration (FDA)-recommended endpoints to judge efficacy. The level of evidence is weaker for more traditional therapies that were previously approved based on smaller, lower quality RCTs that involved heterogeneous patients or unvalidated endpoints.11 For each intervention discussed, the mechanisms, efficacy and safety (where available) are summarized.

Figure 1. Pharmacotherapy in Irritable Bowel Syndrome.

Figure 1.

From J Clin Med6with permission. Note: This is an open access article distributed under the Creative Commons Attribution License which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited (CC BY 4.0).

aCl = chloride; GC-C = guanylate cyclase C; NHE = sodium/hydrogen exchanger; TSPO = translocator protein

General Lifestyle Measures

Dietary Modifications

Many patients believe their IBS symptoms are due to food sensitivity.12

Mechanisms:

Food generates symptoms in patients with IBS,13 and four potential explanations are: prominent contractile14 and sensory15 responses of the colon to the ingestion of food (“gastrocolonic response”); alterations in the microbiome (which may occur quite rapidly after a change in diet);16 insoluble dietary fiber may exacerbate IBS symptoms;17 and dietary antigens may alter intestinal epithelial barrier.18 These putative mechanisms provide rationale for dietary modifications.

Efficacy:

The quality of published trials of dietary interventions in IBS is generally weak. A systematic review recommended that more evidence is needed.19 Specific diets, including low fructose, oligosaccharides, disaccharides, monosaccharides and polyols (FODMAPs) diet, are discussed below.

Elimination Diets

A sham-diet, RCT of 150 patients assessed a diet based on avoidance of foods to which patients had 3-fold elevated IgG antibody titers over background.20 Symptoms were substantially improved at 12 weeks. The results of the study showed that the number needed to treat (NNT) was 9 and reintroducting eliminated foods resulted in substantial worsening of symptoms. It is unclear whether the diet’s efficacy differed according to stool pattern. On the other hand, double-blind rechallenge to the dietary triggers resulted in reproducible symptoms in only ~25% of patients.21

Increased Dietary Fiber

Efficacy:

In the largest trial of the use of fiber in 275 patients with IBS (all subtypes) in the primary care setting, patients were randomized to 12 weeks of treatment with soluble fiber (psyllium), insoluble bran fiber, or placebo.22 Whereas bran was of no benefit, there was reduction in IBS symptom severity score (IBS-SSS) with psyllium over placebo at 12 weeks, and greater proportion of responders (>2 weeks’ adequate relief per month) with an NNT for psyllium of 4. Adverse event rates (overall) were not significantly more prevalent in either of the fiber groups compared to placebo.

In a systematic review and meta-analysis23 that included 906 patients in 14 RCTs (most of low methodological quality), there was only modest improvement of symptoms with fiber in IBS (NNT of 10), and the beneficial effect was limited to psyllium (which was tested in 499 patients in 7 studies) with an NNT of 7. The effect of bran was not significant. The rates of adverse events with psyllium, bran and placebo were not significantly different.

In contrast to weak benefits in IBS, mixed soluble and insoluble fiber and psyllium were equally efficacious in improving constipation in patients with chronic constipation.24

Mechanism:

The mechanism of benefit with psyllium is uncertain and is unlikely to relate to stool bulking alone, since bran has similar effects.25 On the other hand, increased production of short chain fatty acids, such as butyrate, with psyllium treatment may have anti-inflammatory effects on the colonic mucosa26 or alter the intestinal microbiota,27 which may conceivably contribute to reported benefits of psyllium.

Low FODMAP Diet

Mechanism:

FODMAPs are present in many commonly consumed foods (such as stone fruits and legumes), lactose-containing foods, and artificial sweeteners. As these chemical substances are poorly absorbed, they may induce osmotic effects (and fluid secretion) or fermentation (and distension) in the intestine,28,29 leading to increased colonic sensitivity from the distension.30 Sugar alcohols can induce dose-dependent symptoms of flatulence, abdominal discomfort, and laxative effects when consumed by both healthy volunteers and patients with IBS.31 A low FODMAP diet may lead to reduction in bacterial abundance32 and lower proportions of certain bacteria including Bifidobacteria.33 However, it is unclear whether the restriction of FODMAPs leads to long-term effects on the gut microbiota.

Efficacy:

The clinical benefits of a low FODMAPs diet remain indeterminate. For example, a crossover, RCT comparing a diet low in FODMAPs with a typical Australian diet in 30 patients with IBS of all subtypes,34 reported reduced global IBS symptoms, bloating, and pain while on the low FODMAP diet, with greater benefit in those with diarrhea-predominant IBS (IBS-D). Conversely, a larger parallel-group RCT of 67 IBS patients showed no difference in efficacy between low FODMAPs diet and conventional dietary recommendations,35 with both diets showing benefit relative to baseline.

Several meta-analyses suggest efficacy of low FODMAPs diet;36,37 however, analysis identified risk of bias, primarily due to lack of proper blinding and choice of control.38 Despite the weak evidence, the National Institute of Health and Care Excellence of the United Kingdom recommended, as a first line treatment, the use of low FODMAPs diet for patients with IBS in primary care.39 The IBS dietary algorithm from the British Dietetic Association was simplified to first-line healthy eating (provided by any healthcare professional) and second-line low FODMAP dietary advice.40

Gluten-free Diet

In the absence of markers of celiac disease, there is evidence that a subgroup of patients with IBS may benefit from use of gluten-free diet (GFD).

Mechanism:

The mechanism of the effect of gluten in IBS is unclear; gluten-containing diet affects small bowel epithelial mRNA expression of barrier protein and higher small bowel mucosal permeability in patients with IBS-D compared to GFD.41 There is also evidence that there are differences in expression of immune markers on gluten sensitivity without celiac disease in those who present with symptoms mimicking IBS-D, and increased expression of tolllike receptor 2 and reduced T-regulatory cell marker FOXP3 relative to controls and patients with celiac disease.42 In view of the fact that wheat contains high levels of fructans in addition to gluten, a trial examined the combination of low FODMAPs diet and GFD,43 and showed no additive effect of the GFD, suggesting that reduction in fructans may partly explain the effectiveness of a GFD in IBS.

Efficacy:

Two RCTs demonstrate that response to a GFD is greater in patients who are HLA-DQ2 or –DQ8 positive.41,44 A third RCT showed similar improvement in IBS symptoms severity score; in addition, patients who were HLA-DQ2/8-positive had a greater reduction in depression score and increase in vitality score on the GFD, whereas patients who were HLA-DQ2 negative had improved bloating scores.45

Exercise

A few studies examined exercise as an approach to reduce symptoms in IBS. In a Swedish trial, initially over 12 weeks46 and, subsequently, with median follow-up of 5.2 (range: 3.8-6.2) years,47 20 to 60 minutes of moderate to vigorous physical activity (most commonly walking, aerobics and cycling) on 3 to 5 days per week was associated with significant improvement in symptom scores (IBS-SSS and psychological symptoms) over those in the control arm. It is unclear whether the effect of exercise differs according to IBS subtype.

Other movement-based, self-regulatory behavioral treatments have been shown to be beneficial for IBS patients, with yoga tending to reduce severity of IBS and somatic symptoms, and walking improving overall GI symptoms, negative affect, and anxiety.48

Alternative and Herbal Therapies

Prebiotics and Probiotics

Prebiotics include food ingredients such as fructo-oligosaccharides or inulin that remain undigested in the human gastrointestinal system and can promote the growth or activity of gut bacteria. In contrast, probiotics are live or attenuated micro-organisms that can affect the composition of the intestinal microorganisms. Probiotics may have anti-inflammatory and anti-nociceptive properties.49-52

Two trials of prebiotics in IBS were reported in the last three years. A RCT of 12 weeks duration compared 6g partially hydrolyzed guar gum, a prebiotic fiber, to placebo in 121 patients with IBS of all subtypes53 and demonstrated significant improvement in bloating; however, there was no benefit on global symptoms, abdominal pain, or quality of life. A second study tested the prebiotic, inulin (900mg), in children with IBS, and none of the 6 symptoms of IBS tested improved, in contrast to significant improvements with the synbiotic [5×109 colony forming units (CFU) of B. lactis B94 and 900 mg inulin] and the probiotic (5×109 CFU B. lactis B94) tested over 4 weeks.54

In a meta-analysis published in 2014, 35 trials of probiotics, involving 3452 patients with IBS55 showed that probiotics have a beneficial overall effect in IBS (NNT of 7) with the greatest impact on abdominal pain, bloating, and flatulence, but not on bowel urgency or bowel function. Mild adverse events were significantly more common with probiotics compared to placebo.

More recent meta-analyses of probiotics suggest benefit in IBS patients for overall symptoms treated with B. infantis (5 RCTs),56 S. cerevisiae CNCM I-3856 (2 trials),57 single probiotics at relatively lower dosage of organisms (<1010 CFU/day) and shorter duration (<8weeks).58 A meta-analysis of 15 trials that included 1793 patients showed improvement of general symptoms (7 trials), and of pain, distension, bloating, and flatulence each in 2 to 3 trials.59 With most trials of probiotics, few were of high methodological quality, and combining data from different probiotic species, strains, or combinations may not be valid.60

Other Herbal Therapies

The efficacy of other herbal therapies in IBS is unclear. Iberogast (STW-5) is a mixture of diverse extracts of flower, leaves, fruit, root, and herbs61 with antispasmodic effects on gastrointestinal smooth muscle62 through diverse mechanisms,63 and secretory effect on diverse chloride channels.64 In a RCT of 208 patients with IBS,65 there was improvement in global symptoms and abdominal pain scores with STW-5 compared to placebo.

The benefits of Chinese herbal medicines in IBS are inconsistent.66-68

MEDICATIONS FOR PAIN (Table 1)

Table 1. Summary of Current Pharmacological Treatments for IBS.

Mode of Action Therapy Efficacy Quality of
data
Adverse events Limitations of data
Smooth muscle relaxation Antispasmodic drugs +/− Low Dry mouth, dizziness, and blurred vision No high quality trials, heterogeneity between studies, possible publication bias, and only a small number of RCTsc assessing each individual antispasmodic
Peppermint oil + Moderate No increase in AEsa Heterogeneity between studies
Secretagogues Lubiprostone + Moderate Nausea commoner vs. placebo Only a modest benefit over placebo in published RCTsc
Linaclotide + High Diarrhea commoner vs. placebo None
Plecanatide + High Diarrhea commoner vs. placebo None
Tenapanor +/− Moderate Diarrhea commoner vs. placebo Awaiting phase 2B/3 trials
Neuromodulators Antidepressants + Moderate Dry mouth and drowsiness Few high quality trials, heterogeneity between studies, possible publication bias, and some atypical trials included
Neurokinin NK2 antagonist Promising in phase 2B RCTc Moderate No increase in AEsa Awaiting phase 3 trials
Histamine H1 antagonist Promising in single center trial Low No increase in AEsa Awaiting phase 2B trials
TSPOd inhibitor +/− Low Modest efficacy in a single proof of concept trial Awaiting phase 2B trials
Opioids Loperamide +/− Low Limited data Few RCTsc, with a small number of participants, not all of whom had IBSb
Eluxadoline + High Serious AEsa: acute pancreatitis and sphincter of Oddi spasm. Nausea and headache common vs. placebo Only a modest benefit over placebo in published RCTsc. No benefit over placebo in terms of abdominal pain
e5-HT3 receptor antagonists + High Serious AEa with alosetron: ischemic colitis; Constipation commoner vs. placebo. Fewer RCTsc of ramosetron and ondansetron. Ondansetron may have no benefit over placebo for abdominal pain.
e5-HT4 receptor agonists +/− high Diarrhea, cramping, and cardiovascular AEsa with “old generation” drugs in this class Data available for tegaserod and mosapride, not for “new generation” drugs in this class: prucalopride, naronapride, velusetrag, YKP10811
Bile acid sequestrants ? Low Limited data No published RCT sc
Rifaximin + Moderate No increase in AEsa Only a modest benefit over placebo in published RCTsc
a

AEs=adverse events

b

IBS=irritable bowel syndrome

c

RCTs=randomized, controlled trials

d

TSPO=translocator protein

e

5-HT=serotonin

Antispasmodic Drugs

Mechanism:

Antispasmodics inhibit the action of acetylcholine at muscarinic or tachykinin NK2 receptors, or block calcium channels on gastrointestinal smooth muscle, and alter gastrointestinal transit, contributing to relief of pain and disturbances in bowel habit.

Efficacy:

Systematic reviews documented weak evidence for the benefit of some antispasmodics for abdominal pain and global symptom relief,69 and significant improvement in abdominal pain in 7 of 9 studies, bowel symptoms in 2 of 9, and global symptom severity in 4 of 9 studies was reported.70

In a meta-analysis of 22 separate RCTs involving 1778 patients and 12 different antispasmodic drugs,71 antispasmodics were more effective than placebo, with an NNT of 5 overall, and slightly lower NNTs with hyoscine 3.5 (426 patients enrolled in 3 trials), otilonium 4.5 (435 patients enrolled in 4 trials,), cimetropium 3 (158 patients enrolled in 3 trials), and pinaverium 3 (188 patients enrolled in 3 trials), but confidence in these estimates is reduced because of significant heterogeneity, methodological weaknesses, possible publication bias and insufficient information on efficacy according to IBS subtype. In addition, most of the drugs studied are not approved by the Food and Drug Administration for the treatment of IBS in the United States, and promising data for those medications are reviewed elsewhere.6

Safety:

Antispasmodics cause more side effects than placebo, with the most common being dry mouth, dizziness, blurred vision, and constipation.

Peppermint Oil

Mechanism:

The major constituent of peppermint oil is menthol, which inhibits smooth muscle contractility in the gastrointestinal tract by blocking calcium influx.72,73 Menthol also induces analgesia by activating the temperature sensing ion channel, transient receptor potential cation channel subfamily M member 8 (TRPM8),74 which has anti-nociceptive properties in visceral afferents.

Efficacy:

Peppermint oil was more effective than placebo in a meta-analysis of four trials including 392 IBS patients with IBS (NNT of 2.5).71 The same methodological issues described for anti-spasmodics apply, and the estimate of efficacy (NNT) is likely inaccurate.

In a later meta-analysis of five RCTs of peppermint oil including 197 patients on active treatment and 195 on placebo, peppermint oil resulted in global improvement of IBS symptoms (5 studies) and abdominal pain (5 studies).75

A novel formulation with sustained release in the small intestine76 was tested in a 4-week trial in 72 patients with IBS-D or IBS-mixed (IBS-M), and there was no superiority over placebo for total IBS symptom score, though both treatment arms showed improvement from baseline.

Safety:

Peppermint oil can cause symptoms of gastroesophageal reflux, xerostomia, belching, a peppermint taste in the mouth, and a peppermint smell.

Antidepressants

Mechanism:

The rationale for using antidepressants in IBS includes: the co-existence of psychological disorders in IBS;77 evidence that depression modifies the central nervous system response to painful stimuli;78 the benefits of antidepressants in chronic painful disorders;79,80 and correction of altered intestinal transit. Thus, tricyclic antidepressants (TCAs) prolong orocecal and intestinal transit times, whereas selective serotonin re-uptake inhibitors (SSRIs) decrease orocecal transit time.81 Based on this, TCAs are used in IBS-D and SSRIs are preferred in constipation-predominant IBS (IBS-C).

The mechanism of action of antidepressants in IBS is multifactorial, and may include reduced activation of pain centers in the anterior cingulate cortex and central pain processing,82 and peripheral mechanisms that have an effect on pain sensation such as colonic compliance and visceral afferent function.

Efficacy:

An updated systematic review and meta-analysis83 including 17 separate trials of antidepressants found overall beneficial effects of antidepressants on IBS symptoms (NNT of 4). However, low level of trial quality, inconsistencies of trial endpoints, questionable generalizability, uncharacteristic response levels of the placebo arm (14%)84 or the antidepressant (63%),85 and evidence of heterogeneity between studies and possible publication bias raise questions on the accuracy of the reported NNT.

In general, TCAs appear to have greater efficacy with an NNT of 4. In the meta-analysis, no heterogeneity was seen between the 11 studies that included TCAs compared with SSRIs, which also had an NNT of 4, but with significant heterogeneity among 7 trials. Seven RCTs reported benefit for abdominal pain, but there was substantial heterogeneity between studies. Effectiveness according to IBS subtype has been studied in only two RCTs.84,85 Three trials of antidepressants in IBS have showed no correlation between improvement in IBS symptoms and depression scores,86-88 and a fourth trial of a TCA showed greater benefit in non-depressed individuals.89

There are three open-label trials of the effects of duloxetine, a serotonin- and norepinephrine-reuptake inhibitor, that has shown efficacy in the treatment of IBS, such as in IBS-SSS (symptom severity), pain, bowel dysfunction, and quality of life.90-92 The SNRI class of medications is used in the treatment of pain; however, double-blind, placebo-controlled, randomized clinical trials for the relief of IBS, pain and the associated co-morbid depressive or generalized anxiety disorders are required.

Safety:

Side effects were significantly more common with TCAs; the most frequent side effects were drowsiness and dry mouth. Long-term use of some classes of psychotropic drugs for nonpsychiatric indications may be linked with dementia, based on population studies,93,94 although a cause and effect relationship has not been proven.

Drugs Acting on Opioid Receptors

Mechanism:

Opioid receptor agonists slow gut and colonic transit, increase fluid absorption, and reduce the sensation of pain.95

Efficacy:

Loperamide and diphenoxylate, μ-opioid agonists, are anti-diarrheal agents that have been in use for the treatment of chronic functional diarrhea for many years,96 based on limited evidence from small studies conducted around 30 years ago. One small trial of 21 patients with IBS-D, showed loperamide improved stool consistency, pain, as well as urgency.97 A second trial98 of 60 patients with either functional diarrhea or IBS-D confirmed reduction in stool frequency, improved consistency, as well as number of days with pain. A third trial99 of loperamide in unselected IBS patients showed improvement in stool frequency and consistency, and overall pain intensity, but was associated with increased abdominal pain during the night. Loperamide is the most useful agent for diarrhea or urgency.9

Eluxadoline is a novel κ- and μ-opioid receptor agonist and δ-opioid receptor antagonist. Based on 3 trials with ~3000 patients with IBS treated over 12 weeks, it was efficacious in the relief of diarrhea or the composite endpoint of diarrhea and pain.100,101 The recommended dose is 100 mg bid, unless not tolerated or if there is hepatic impairment, then the 75 mg bid dose should be used.

Safety:

Adverse events with eluxadoline are mainly nausea and headache. Rare cases of pancreatitis and sphincter of Oddi spasm have been reported. In accordance with FDA recommendation, eluxadoline should not be prescribed to patients with a history of biliary obstruction, cholecystectomy, pancreatitis, severe hepatic impairment, or severe constipation, or to those who consume more than three alcoholic drinks per day.

Off-Label Approaches for Visceral Pain

Histamine H1 receptor antagonist, ebastine

This drug is not approved in the U.S.

Mechanism:

Mast cells and their mediators, in particular, histamine, serotonin and proteases, contribute to the pathogenesis of IBS.102 Histamine released by colonic biopsies from patients with IBS can sensitize (via H1 histamine receptors) the TRPV1 on neurons in dorsal root ganglia (on afferent pathways) and on human submucosal neurons in rectal biopsies.103

Ebastine, a non-sedating antagonist of H1 receptors, has been shown to reduce visceral hypersensitivity, overall IBS symptoms, and abdominal pain in patients with IBS.103

Gamma-aminobutyric acid (GABA)ergic agents

GABAergic agents are α2δ ligands that reduce the release of many excitatory neurotransmitters involved in pain mechanisms including glutamate, noradrenaline, substance P, and calcitonin gene-related peptide (CGRP). Three studies assessed the effects of gabapentin and pregabalin on rectal and colonic sensation and compliance in IBS-D patients or healthy controls, and overall they suggest that these agents reduce senation without significantly affecting compliance. This suggests an effect exclusively on sensory mechanisms.104-106

A preliminary report of a randomized, controlled, 12-week clinical trial of pregabalin (dose escalation regimen to a maximum of 225 mg bid conducted at Mayo Clinic, Rochester, Minnesota in 85 patients with IBS showed lower average pain scores during weeks 9-12 and lower mean symptom severity scores with pregabalin compared with placebo.107

Future Approaches to Pain Relief in IBS

A new generation of peripherally active visceral analgesics,108 including opioid agents with no risk of respiratory depression or addiction potential, is being developed; these medications are eagerly awaited to address the significant unmet need of pain in IBS.

MEDICATIONS FOR DIARRHEA (Table 1)

Opioid agents and antidepressants (TCAs and SNRIs) may relieve diarrhea in addition to their effects on pain.

5-HT3 Receptor Antagonists

Mechanism:

Ninety percent of the total body serotonin (5-HT) is within intestinal enterochromaffin cells.109,110 5-HT is also a transmitter in the brain, and there are several different classes of 5-HT receptors in the brain and gut. Patients with IBS-D have increased and those with IBS-C have reduced postprandial 5-HT levels.111 5-HT3 receptor antagonists such as alosetron112 retard colonic transit. 5-HT3 receptors are also important mediators of visceral pain.113

Efficacy:

Alosetron is an effective agent based on the results of several meta-analyses of high quality, large RCTs which have all shown consistent results.114,115 In these studies, alosetron has an NNT of 8 for relief of abdominal pain, and an NNT of 4 for improvement in global symptoms. Alosetron is approved for use in women with severe IBS-D in the United States, but is regulated by an FDA prescribing program.

Ramosetron is efficacious and licensed for use in both male and female patients with IBS-D in Japan.116,117

In a crossover clinical trial118 in patients with IBS-D (n=122), ondansetron had significant effects on stool consistency, urgency, and frequency, and bloating, but no significant beneficial effect on pain.

Safety:

As a drug class, 5-HT3 antagonists can cause constipation butthis is usually manageable by adjusting the dose. Alosetron, unlike other drugs in this class, is associated with ischemic colitis (~1:800 treated patients).119

Bile Acid Sequestrants

Approximately 25% of patients with IBS-D have evidence of bile acid malabsorption (BAM).120,121 Although no RCTs of BA sequestrants in IBS have been reported, a Mayo Clinic open-label trial of colesevelam, 1875 mg twice daily for 10 days,122 in patients with IBS-D and increased fecal bile acid excretion demonstrated reduction in stool consistency and frequency. Another open-label study of colestipol, 1g b.i.d. for 8 weeks,123 showed improvements in IBS symptom severity scores, stool frequency, and adequate relief of symptoms in patients with IBS-D and BAM [a selenium-75-homocholic acid taurine (75SeHCAT) retention <20%].

Antibiotics

Rifaximin is a non-absorbable antibiotic that has shown improved global symptoms and bloating in IBS in two phase III RCTs.124,125 These trials included more than 1,200 patients with non-constipated IBS. Rifaximin, 550 mg TID for 2 weeks was associated with higher rates of adequate relief of global IBS symptoms and bloating (NNT of 9-12.5). The effect on symptoms lasted up to 10 weeks post-treatment. The NNT of 10 was confirmed for global symptoms and bloating in a meta-analysis of five RCTs of rifaximin,126 including 1803 patients. However, stool consistency, frequency of bowel movements, and urgency were not improved.

With repeat courses of rifaximin separated by 10 weeks, 550mg t.i.d. daily for 2 weeks in each course, there was significant benefit for urgency, bloating and combined abdominal pain and stool consistency with each of 2 repeat treatment courses compared to placebo. Rifaximin is approved for IBS-D patients with up to two repeat treatments in case of symptom recurrence. It is worth noting that rifaximin127 accelerated ascending colon emptying and overall colonic transit at 48 hours in IBS-D, although it had no effects on intestinal mucosal permeability, stool microbiome, or stool bile acids. The acceleration of colonic transit would seem deleterious for patients with IBS-D; on the other hand, it may explain the reported improvement in IBS-C (bloating, constipation, and straining) with combined neomycin plus rifaximin compared to neomycin with placebo.128

Importantly, there is no evidence of adverse effects with rifaximin compared to placebo, and no increased risk of Clostridium difficile.

MEDICATIONS FOR CONSTIPATION (Table 1)

Intestinal Secretagogues

Chloride channel-related

Lubiprostone is a prostaglandin derivative that acts on chloride channels on the apical membrane of the intestinal enterocyte to induce chloride secretion, which is then followed by the passive movement of sodium ions and water into the lumen. As a result, stools become looser and gastrointestinal transit is accelerated.129 The drug is approved at a dose of 8 μg twice daily for women with IBS-C130 and 24 μg bid for men and women with chronic constipation.131,132 There are general improvements in abdominal pain scores that parallel the improved straining and stool consistency. Nausea is the most common side effect, experienced by 8% of patients, but it is generally relatively mild and self-limited.133

Linaclotide is a minimally absorbed guanylate cyclase C receptor (GC-C) agonist that causes secretion of chloride and bicarbonate into the intestinal lumen via the cystic fibrosis transmembrane regulator (CFTR). This results in parallel and sodium and water secretion. The activation of CFTR results from increase in intracellular cyclic guanosine monophosphate (cGMP), which has also been shown to affect sensory afferent neurons, leading to inhibition of pain. In clinical trials conducted in patients with IBS-C (as well as others in chronic constipation), linaclotide relieved constipation and significantly improved abdominal discomfort and bloating.134-136 Three dose levels are approved: 72, 145, and 290 mcg per day: 72 mcg and 145 mcg doses are for chronic idiopathic constipation, and the 290 mcg dose for IBS-C (men and women). Dose can be titrated to observe benefit and reduce the risk of diarrhea, which may occur in up to 20% of patients administered the highest dose.

Plecanatide is a peptide analog of uroguanylin, which is an endogenous GC-C agonist137 released into the intestine from goblet cells. Plecanatide, 3 or 6mg per day, is also efficacious in treating chronic idiopathic constipation138 and IBS-C139, including relief of worst abdominal pain. Plecanatide, 3 mg dose, is now FDA approved for both indications. It is reported to be associated with lower risk of diarrhea than linaclotide, although the availability of 3 doses of linaclotide provides opportunity to titrate its dose to avoid diarrhea.

5-HT4 Receptor Agonists

As a class, 5-HT4 receptor agonists have demonstrated efficacy in patients with IBS-C. Tegaserod (available in come countries, but not in the U.S.) is an effective treatment for IBS-C, relieving overall and multiple individual IBS-C symptoms (abdominal pain/discomfort, bloating, and constipation) in placebo-controlled as well as open-label trials.140 Repeat treatments with tegaserod are effective, and tegaserod is associated with improvements in quality of life and work productivity.140

Adverse effects associated with tegaserod are diarrhea, cramping, and rare cardiovascular events, with the latter attributed to off-target effects on other 5-HT receptors (e.g. 5-HT2A and 2B antagonist). However, in a matched cohort study conducted within a large U.S. health insurance database involving 52,229 patients, there was no increased risk of cardiovascular ischemic events.141

Mosapride (approved in some countries, but not in the U.S.) improved symptoms in patients with IBS-C in a pilot study (n=10).142 However, a 12-month study in a larger cohort (n=69 patients) showed no significant improvements in overall or specific IBS symptoms (pain, bloating, stool frequency or consistency), or quality of life with mosapride over placebo.143

Cognitive Behavioral Therapy and Hypnotherapy

Where available, cognitive behavioral therapy (CBT) and hypnotherapy may be used in the management of patients with IBS. Recent systematic reviews show psychological interventions are efficacious including long-term benefits, and that the gains are not dependent on the number of sessions. Indeed, CBT and hypnosis appear efficacious in minimal-contact formats, as well as various technologies (e.g., internet, telephone, smartphone apps), self-help interventions and engaging trained nonprofessional mental health providers to deliver interventions.144

Conclusions

At the present time, the treatment of IBS remains focused on treating the patient’s most troublesome symptom. These treatments are quite efficacious for bowel dysfunction, though the treatment of pain without use of opioids or centrally acting agents is suboptimal. Hence, lifestyle interventions including diet, cognitive behavioral therapy and hypnotherapy (the latter not discussed here) should be considered to relieve symptoms in a holistic approach to the patient’s symptoms. Pharmacotherapies have not been shown to alter the long-term or natural history of the disorder. There is still considerable unmet need; new treatments that target some of the important actionable biomarkers of IBS, as well as the ability to conduct high quality, randomized, controlled trials augur well for the development of treatments that will impact patients’ symptoms and hopefully the natural history of IBS in the future. In particular, the new generation of peripherally active visceral analgesics, including opioid agents with no risk of respiratory depression or addiction potential, is eagerly awaited to address the significant unmet need of pain in IBS.

Acknowledgments

Funding: NIH R01-DK92179

Abbreviations:

IBS

irritable bowel syndrome

RCTs

randomized, controlled trials

5-HT

serotonin

TRP

transient receptor potential

FDA

Food and Drug Administration

FODMAPs

fructose, oligosaccharides, disaccharides, monosaccharides and polyols

NNT

number needed to treat

IBS-SSS

IBS symptom severity score

IBS-D

diarrhea-predominant IBS

GFD

gluten-free diet

CFU

colony forming units

STW-5

iberogast

TRPM8

transient receptor potential cation channel subfamily M member 8

IBS-M

IBS-mixed

TCAs

tricyclic antidepressants (TCAs)

SSRIs

selective serotonin re-uptake inhibitors

IBS-C

constipation-predominant IBS

GABA

amma-aminobutyric acid

CGRP

calcitonin gene-related peptide

BAM

bile acid malabsorption

75SeHCAT

selenium-75-homocholic acid taurine

GC-C

guanylate cyclase C

CFTR

cystic fibrosis transmembrane regulator

cGMP

cyclic guanosine monophosphate

CBT

cognitive behavioral therapy

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: Dr. Camilleri has received research funding regarding this topic from Novartis, Allergan and Entera Health. He has done consulting regarding this topic (with the fee going to his employer, Mayo Clinic) for Elobix AB, Shire, and Takeda.

References

  • 1.Ford AC, Forman D, Bailey AG, Axon ATR, Moayyedi P. Irritable bowel syndrome: A 10-year natural history of symptoms, and factors that influence consultation behavior. Am J Gastroenterol. 2008;103(5): 1229–1239. [DOI] [PubMed] [Google Scholar]
  • 2.Camilleri M, Shin A, Busciglio I, et al. Validating biomarkers of treatable mechanisms in irritable bowel syndrome. Neurogastroenterol Motil. 2014;26(12):1677–1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Camilleri M. Review Article: Biomarkers and personalized therapy in lower functional gastrointestinal disorders. Aliment Pharmacol Ther. 2015;42(7): 818–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Brierley SM, Linden DR. Neuroplasticity and dysfunction after gastrointestinal inflammation. Nat Rev Gastroenterol Hepatol. 2014;11(10):611–627. [DOI] [PubMed] [Google Scholar]
  • 5.Julius D. TRP channels and pain. Annu Rev Cell Dev Biol. 2013;29(Oct.):355–384. [DOI] [PubMed] [Google Scholar]
  • 6.Camilleri M, Ford AC. Invited Review: Pharmacotherapy for irritable bowel syndrome. J Clin Med. 2017;6(11):E101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.American College of Gastroenterology Task Force on Irritable Bowel Syndrome, Brandt LJ, Chey WD, et al. An evidence-based position statement on the management of irritable bowel syndrome. Am J Gastroenterol. 2009;104(Jan., Suppl. 1):S8–S35. [DOI] [PubMed] [Google Scholar]
  • 8.Chang L, Lembo A, Sultan S. American Gastroenterological Association Institute Technical Review on the pharmacological management of irritable bowel syndrome. Gastroenterology 2014;147(5): 1149–1172, e2. [DOI] [PubMed] [Google Scholar]
  • 9.Ford AC, Moayyedi P, Lacy BE, et al. American College of Gastroenterology monograph on the management of irritable bowel syndrome and chronic idiopathic constipation. Am J Gastroenterol. 2014;109 (Aug., Suppl. 1):S2–S26. [DOI] [PubMed] [Google Scholar]
  • 10.Spiller R, Aziz Q, Creed FEA, et al. Clinical Services Committee of The British Society of Gastroenterology guidelines on the irritable bowel syndrome: mechanisms and practical management. Gut 2007;56(12):1770–1798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Camilleri M American College of Gastroenterology monograph on the management of irritable bowel syndrome. Expert Opin Pharmacother. 2015;16(5):629–632. [DOI] [PubMed] [Google Scholar]
  • 12.Bohn L, Storsrud S, Tornblom H, Bengtsson U, Simren M. Self-reported food-related gastrointestinal symptoms in IBS are common and associated with more severe symptoms and reduced quality of life. Am J Gastroenterol. 2013;108(5):634–641. [DOI] [PubMed] [Google Scholar]
  • 13.Ragnarsson G, Bodemar G. Pain is temporally related to eating but not to defaecation in the irritable bowel syndrome (IBS). Patients’ description of diarrhea, constipation and symptom variation during a prospective 6-week study. Eur J Gastroenterol Hepatol. 1998;10(5):415–421. [DOI] [PubMed] [Google Scholar]
  • 14.Sullivan MA, Cohen S, Snape WJ Jr. Colonic myoelectrical activity in irritable-bowel syndrome. Effect of eating and anticholinergics. N Engl J Med. 1978;298(16):878–283. [DOI] [PubMed] [Google Scholar]
  • 15.Simrén M, Abrahamsson H, Björnsson ES. An exaggerated sensory component of the gastrocolonic response in patients with irritable bowel syndrome. Gut 2001;48(1):20–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505(7484):559–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Francis CY, Whorwell PJ. Bran and irritable bowel syndrome: time for reappraisal. Lancet 1994;344(8914):39–40. [DOI] [PubMed] [Google Scholar]
  • 18.Fritscher-Ravens A, Schuppan D, Ellrichmann M, et al. Confocal endomicroscopy shows food-associated changes in the intestinal mucosa of patients with irritable bowel syndrome. Gastroenterology 2014;147(5):1012–1020, e4. [DOI] [PubMed] [Google Scholar]
  • 19.Moayyedi P, Quigley EM, Lacy BE, et al. The effect of dietary intervention on irritable bowel syndrome: A systematic review. Clin Transl Gastroenterol. 2015;6(Aug. 20):e107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Atkinson W, Sheldon TA, Shaath N, Whorwell PJ. Food elimination based on IgG antibodies in irritable bowel syndrome: A randomised controlled trial. Gut 2004;53(10):1459–1464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Young E, Stoneham MD, Petruckevitch A, Barton J, Rona R. A population study of food intolerance. Lancet 1994;343(8906):1127–1130. [DOI] [PubMed] [Google Scholar]
  • 22.Bijkerk CJ, de Wit NJ, Muris JW, Whorwell PJ, Knottnerus JA, Hoes AW. Soluble or insoluble fibre in irritable bowel syndrome in primary care? Randomised placebo controlled trial. Br Med J. 2009;339(Aug. 27): b3154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Moayyedi P, Quigley EM, Lacy BE, et al. The effect of fiber supplementation on irritable bowel syndrome: A systematic review and meta-analysis. Am J Gastroenterol. 2014;109(9): 1367–1374. [DOI] [PubMed] [Google Scholar]
  • 24.Erdogan A, Rao SS, Thiruvaiyaru D, et al. Randomised clinical trial: mixed soluble/insoluble fibre vs. psyllium for chronic constipation. Aliment Pharmacol Ther. 2016;44(1): 35–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tomlin J, Read NW. Laxative properties of indigestible plastic particles. Br Med J. 1988;297(6657): 1175–1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Zimmerman MA, Singh N, Martin PM, et al. Butyrate suppresses colonic inflammation through HDAC1-dependent Fas upregulation and Fas-mediated apoptosis of T cells. Am J Physiol Gastrointest Liver Physiol. 2012;302(12):G1405–G1415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lee SM, Han HW, Yim SY. Beneficial effects of soy milk and fiber on high cholesterol diet-induced alteration of gut microbiota and inflammatory gene expression in rats. Food Funct. 2015;6(2):492–500. [DOI] [PubMed] [Google Scholar]
  • 28.Shepherd SJ, Parker FC, Muir JG, Gibson PR. Dietary triggers of abdominal symptoms in patients with irritable bowel syndrome: Randomized placebo-controlled evidence. Clin Gastroenterol Hepatol. 2008;6(7):765–771. [DOI] [PubMed] [Google Scholar]
  • 29.Undseth R, Berstad A, Klow NE, Arnljot K, Moi KS, Valeur J. Abnormal accumulation of intestinal fluid following ingestion of an unabsorbable carbohydrate in patients with irritable bowel syndrome: An MRI study. Neurogastroenterol Motil. 2014;26(12):1686–1693. [DOI] [PubMed] [Google Scholar]
  • 30.Major G, Pritchard S, Murray K, et al. Colon hypersensitivity to distension, rather than excessive gas production, produces carbohydrate-related symptoms in individuals with irritable bowel syndrome. Gastroenterology 2017;152(1):124–133. [DOI] [PubMed] [Google Scholar]
  • 31.Lenhart A, Chey WD. A systematic review of the effects of polyols on gastrointestinal ealth and irritable bowel syndrome. Adv Nutr. 2017;8(4):587–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Halmos EP, Christophersen CT, Bird AR, Shepherd SJ, Gibson PR, Muir JG. Diets that differ in their FODMAP content alter the colonic luminal microenvironment. Gut 2015;64(1):93–100. [DOI] [PubMed] [Google Scholar]
  • 33.Staudacher HM, Lomer MC, Anderson JL, et al. Fermentable carbohydrate restriction reduces luminal bifidobacteria and gastrointestinal symptoms in patients with irritable bowel syndrome. J Nutr. 2012;142(8):1510–1518. [DOI] [PubMed] [Google Scholar]
  • 34.Halmos EP, Power VA, Shepherd SJ, Gibson PR, Muir JG. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology 2014;146(1):67–75. [DOI] [PubMed] [Google Scholar]
  • 35.Bohn L, Storsrud S, Liljebo T, et al. Diet low in FODMAPs reduces symptoms of irritable bowel syndrome as well as traditional dietary advice: A randomized controlled trial. Gastroenterology 2015;149(6):1399–1407. [DOI] [PubMed] [Google Scholar]
  • 36.Varjú P, Farkas N, Hegyi P, et al. Low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet improves symptoms in adults suffering from irritable bowel syndrome (IBS) compared to standard IBS diet: A meta-analysis of clinical studies. PLoS One 2017;12(8):e0182942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Altobelli E, Del Negro V, Angeletti PM, Latella G. Low-FODMAP diet improves irritable bowel syndrome symptoms: a meta-analysis. Nutrients 2017;9(9). pii: E940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Krogsgaard LR, Lyngesen M, Bytzer P. Systematic review: quality of trials on the symptomatic effects of the low FODMAP diet for irritable bowel syndrome. Aliment Pharmacol Ther. 2017;45(12):1506–1513. [DOI] [PubMed] [Google Scholar]
  • 39.Diagnosis and management of irritable bowel syndrome in adults in primary care: summary of NICE guidance, 2015. Br Med J. 2015;350(Mar. 3):h1216. [DOI] [PubMed] [Google Scholar]
  • 40.McKenzie YA, Bowyer RK, Leach H, et al. British Dietetic Association systematic review and evidence-based practice guidelines for the dietary management of irritable bowel syndrome in adults (2016 update). J Hum Nutr Diet. 2016;29(5):549–575. [DOI] [PubMed] [Google Scholar]
  • 41.Vazquez Roque MI, Camilleri M, Smyrk T, et al. A controlled trial of gluten-free diet in patients with irritable bowel syndrome-diarrhea: Effects on bowel frequency and intestinal function. Gastroenterology 2013;144(5):903–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sapone A, Lammers KM, Casolaro V, et al. Divergence of gut permeability and mucosal immune gene expression in two gluten-associated conditions: celiac disease and gluten sensitivity. BMC Med. 2011;9(Mar. 9):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Biesiekierski JR, Peters SL, Newnham ED, Rosella O, Muir JG, Gibson GR. No effects of gluten in patients with self-reported non-celiac gluten sensitivity after dietary reduction of fermentable, poorly absorbed, short-chain carbohydrates. Gastroenterology 2013;145(2):320–328. [DOI] [PubMed] [Google Scholar]
  • 44.Wahnschaffe U, Schulzke JD, Zeitz M, et al. Predictors of clinical response to gluten-free diet in patients diagnosed with diarrhea predominant irritable bowel syndrome. Clin Gastroenterol Hepatol. 2007;5(7):844–850. [DOI] [PubMed] [Google Scholar]
  • 45.Aziz I, Trott N, Briggs R, North JR, Hadjivassiliou M, Sanders DS. Efficacy of a gluten-free diet in subjects with irritable bowel syndrome-diarrhea unaware of their HLA-DQ2/8 genotype. Clin Gastroenterol Hepatol. 2016;14(5):696–703, e1. [DOI] [PubMed] [Google Scholar]
  • 46.Johannesson E, Simren M, Strid H, Bajor A, Sadik R. Physical activity improves symptoms in irritable bowel syndrome: A randomized controlled trial. Am J Gastroenterol. 2011;106(5):915–922. [DOI] [PubMed] [Google Scholar]
  • 47.Johannesson E, Ringström G, Abrahamsson H, Sadik R. Intervention to increase physical activity in irritable bowel syndrome shows long-term positive effects. World J Gastroenterol. 2015;21(2):600–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Shahabi L, Naliboff BD, Shapiro D. Self-regulation evaluation of therapeutic yoga and walking for patients with irritable bowel syndrome: a pilot study. Psychol Health Med. 2016;21(2): 176–188. [DOI] [PubMed] [Google Scholar]
  • 49.Kamiya T, Wang L, Forsythe P, et al. Inhibitory effects of Lactobacillus reuteri on visceral pain induced by colorectal distension in Sprague-Dawley rats. Gut 2006;55(2):191–196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.O'Mahony L, McCarthy J, Kelly P, et al. Lactobacillus and bifidobacterium in irritable bowel syndrome: Symptom responses and relationship to cytokine profiles. Gastroenterology 2005;128(3):541–551. [DOI] [PubMed] [Google Scholar]
  • 51.Verdu EF, Bercik P, Bergonzelli GE, et al. Lactobacillus paracasei normalizes muscle hypercontractility in a murine model of postinfective gut dysfunction. Gastroenterology 2004;127(3):826–837. [DOI] [PubMed] [Google Scholar]
  • 52.Verdu EF, Bercik P, Verma-Gandhu M, et al. Specific probiotic therapy attenuates antibiotic induced visceral hypersensitivity in mice. Gut 2006;55(2):182–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Niv E, Halak A, Tiommny E, et al. Randomized clinical study: Partially hydrolyzed guar gum (PHGG) versus placebo in the treatment of patients with irritable bowel syndrome. Nutr Metab (Lond). 2016. February 6;13:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Baştürk A1, Artan R, Yılmaz A. Efficacy of synbiotic, probiotic, and prebiotic treatments for irritable bowel syndrome in children: A randomized controlled trial. Turk J Gastroenterol. 2016;27(5):439–443. [DOI] [PubMed] [Google Scholar]
  • 55.Ford AC, Quigley EM, Lacy BE, et al. Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: systematic review and meta-analysis. Am J Gastroenterol. 2014;109(10):1547–1561. [DOI] [PubMed] [Google Scholar]
  • 56.Yuan F, Ni H, Asche CV, Kim M, Walayat S, Ren J. Efficacy of Bifidobacterium infantis 35624 in patients with irritable bowel syndrome: a meta-analysis. Curr Med Res Opin. 2017;33(7): 1191–1197. [DOI] [PubMed] [Google Scholar]
  • 57.Cayzeele-Decherf A, Pelerin F, Leuillet S, et al. Saccharomyces cerevisiae CNCM I-3856 in irritable bowel syndrome: An individual subject meta-analysis. World J Gastroenterol. 2017;23(2):336–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zhang Y, Li L, Guo C, et al. Effects of probiotic type, dose and treatment duration on irritable bowel syndrome diagnosed by Rome III criteria: a meta-analysis. BMC Gastroenterol. 2016;16(1):62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Didari T, Mozaffari S, Nikfar S, Abdollahi M. Effectiveness of probiotics in irritable bowel syndrome: Updated systematic review with meta-analysis. World J Gastroenterol. 2015;21(10):3072–3084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Whelan K. Editorial: The importance of systematic reviews and meta-analyses of probiotics and prebiotics. Am J Gastroenterol. 2014;109(10):1563–1565. [DOI] [PubMed] [Google Scholar]
  • 61.Madisch A, Melderis H, Mayr G, Sassin I, Hotz J. A plant extract and its modified preparation in functional dyspepsia. Results of a double-blind placebo controlled comparative study. Z Gastroenterol. 2001;39(7):511–517. [DOI] [PubMed] [Google Scholar]
  • 62.Ammon HP, Kelber O, Okpanyi SN. Spasmolytic and tonic effect of iberogast (STW 5) in intestinal smooth muscle. Phytomedicine 2006;13 (Nov. 24, Suppl. 5):67–74. [DOI] [PubMed] [Google Scholar]
  • 63.Simmen U, Kelber O, Okpanyi SN, Jaeggi R, Bueter B, Weiser D. Binding of STW 5 (Iberogast) and its components to intestinal 5-HT, muscarinic M3, and opioid receptors. Phytomedicine 2006;13 (Nov. 24, Suppl. 5):51–55. [DOI] [PubMed] [Google Scholar]
  • 64.Krueger D, Gruber L, Buhner S, et al. The multi-herbal drug STW 5 (Iberogast) has prosecretory action in the human intestine. Neurogastroenterol Motil. 2009;21(11):1203–e110. [DOI] [PubMed] [Google Scholar]
  • 65.Madisch A, Holtmann G, Plein K, Hotz J. Treatment of irritable bowel syndrome with herbal preparations: Results of a double-blind, randomized, placebo-controlled, multi¬centre trial. Aliment Pharmacol Ther. 2004;19(3):271–279. [DOI] [PubMed] [Google Scholar]
  • 66.Bensoussan A, Talley NJ, Hing M, Menzies R, Guo A, Ngu M. Treatment of irritable bowel syndrome with Chinese herbal medicine: A randomized controlled trial. JAMA. 1998;280(18): 1585–1589. [DOI] [PubMed] [Google Scholar]
  • 67.Bensoussan A, Kellow JE, Bourchier SJ, et al. Efficacy of a Chinese herbal medicine in providing adequate relief of constipation-predominant irritable bowel syndrome: a randomized controlled trial. Clin Gastroenterol Hepatol. 2015;13(11):1946–1954, e1. [DOI] [PubMed] [Google Scholar]
  • 68.Leung WK, Wu JC, Liang SM, et al. Treatment of diarrhea-predominant irritable bowel syndrome with traditional Chinese herbal medicine: A randomized placebo-controlled trial. Am J Gastroenterol. 2006;101(7):1574–1580. [DOI] [PubMed] [Google Scholar]
  • 69.Quartero AO, Meineche-Schmidt V, Muris J, Rubin G, de Wit N. Bulking agents, antispasmodic and antidepressant medication for the treatment of irritable bowel syndrome. Cochrane Database Syst Rev. 2005. April 18 (2) CD003460. [DOI] [PubMed] [Google Scholar]
  • 70.Tack J, Fried M, Houghton LA, Spicak J, Fisher G. Systematic review: the efficacy of treatments for irritable bowel syndrome--a European perspective. Aliment Pharmacol Ther. 2006;24(2): 183–205. [DOI] [PubMed] [Google Scholar]
  • 71.Ford AC, Talley NJ, Spiegel BMR, et al. Effect of fibre, antispasmodics, and peppermint oil in irritable bowel syndrome: systematic review and meta-analysis. Br Med J. 2008;337(Nov. 13):a2313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hawthorn M, Ferrante J, Luchowski E, Rutledge A, Wei XY, Triggle DJ. The actions of peppermint oil and menthol on calcium channel dependent processes in intestinal, neuronal and cardiac preparations. Aliment Pharmacol Ther. 1988;2(2):101–118. [DOI] [PubMed] [Google Scholar]
  • 73.Amato A, Liotta R, Mule F. Effects of menthol on circular smooth muscle of human colon: Analysis of the mechanism of action. Eur J Pharmacol. 2014;740(Oct. 5): 295–301. [DOI] [PubMed] [Google Scholar]
  • 74.Liu B, Fan L, Balakrishna S, Sui A, Morris JB, Jordt SE. TRPM8 is the principal mediator of menthol-induced analgesia of acute and inflammatory pain. Pain 2013;154(10):2169–2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Khanna R, MacDonald JK, Levesque BG. Peppermint oil for the treatment of irritable bowel syndrome: a systematic review and meta-analysis. J Clin Gastroenterol. 2014;48(6):505–512. [DOI] [PubMed] [Google Scholar]
  • 76.Cash BD, Epstein MS, Shah SM. A novel delivery system of peppermint oil is an effective therapy for irritable bowel syndrome symptoms. Dig Dis Sci. 2016;61(2):560–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Henningsen P, Zimmermann T, Sattel H. Medically unexplained physical symptoms, anxiety and depression: A meta-analytic review. Psychosom Med. 2003;65(4):528–533. [DOI] [PubMed] [Google Scholar]
  • 78.Schmid J, Langhorst J, Gass F, et al. Placebo analgesia in patients with functional and organic abdominal pain: A fMRI study in IBS, UC and healthy volunteers. Gut 2015;64(3):418–427. [DOI] [PubMed] [Google Scholar]
  • 79.McQuay HJ, Tramer M, Nye BA, Carroll D, Wiffen PJ, Moore RA. A systematic review of antidepressants in neuropathic pain. Pain 1996;68(2–3):217–227. [DOI] [PubMed] [Google Scholar]
  • 80.Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev. 2007. October 17 (4) CD005454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gorard DA, Libby GW, Farthing MJ. Influence of antidepressants on whole gut orocaecal transit times in health and irritable bowel syndrome. Aliment Pharmacol Ther. 1994;8(2):159–166. [DOI] [PubMed] [Google Scholar]
  • 82.Morgan V, Pickens D, Gautam S, Kessler R, Mertz H. Amitriptyline reduces rectal pain related activation of the anterior cingulate cortex in patients with irritable bowel syndrome. Gut 2005;54(5):601–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ford AC, Quigley EM, Lacy BE, et al. Effect of antidepressants and psychological therapies, including hypnotherapy, in irritable bowel syndrome: systematic review and meta-analysis. Am J Gastroenterol. 2014;109(9):1350–1365. [DOI] [PubMed] [Google Scholar]
  • 84.Vahedi H, Merat S, Rashidioon A, Ghoddoosi A, Malekzadeh R. The effect of fluoxetine in patients with pain and constipation-predominant irritable bowel syndrome: A double-blind randomized-controlled study. Aliment Pharmacol Ther. 2005;22(5):381–385. [DOI] [PubMed] [Google Scholar]
  • 85.Vahedi H, Merat S, Momtahen S, et al. Clinical trial: The effect of amitriptyline in patients with diarrhea-predominant irritable bowel syndrome. Aliment Pharmacol Ther. 2008;27(8):678–684. [DOI] [PubMed] [Google Scholar]
  • 86.Tabas G, Beaves M, Wang J, Friday P, Mardini H, Arnold G. Paroxetine to treat irritable bowel syndrome not responding to high fiber diet: A double-blind placebo-controlled trial. Am J Gastroenterol. 2004;99(5):914–920. [DOI] [PubMed] [Google Scholar]
  • 87.Tack J, Broekaert D, Fischler B, van Oudenhove L, Gevers AM, Janssens J. A controlled crossover study of the selective serotonin reuptake inhibitor citalopram in irritable bowel syndrome. Gut 2006;55(8):1095–1103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Vij JC, Jiloha RC, Kumar N, Madhu SV, Malika V, Anand BS. Effect of antidepressant drug (doxepin) on irritable bowel syndrome patients. Indian J Psychiatr. 1991;33(4):243–246. [Google Scholar]
  • 89.Drossman DA, Toner BB, Whitehead WE, et al. Cognitive-behavioral therapy versus education and desipramine versus placebo for moderate to severe functional bowel disorders. Gastroenterology 2003;125(1):19–31. [DOI] [PubMed] [Google Scholar]
  • 90.Brennan BP, Fogarty KV, Roberts JL, Reynolds KA, Pope HG Jr, Hudson JI. Duloxetine in the treatment of irritable bowel syndrome: an open-label pilot study. Hum Psychopharmacol 2009;24(5):423–428. [DOI] [PubMed] [Google Scholar]
  • 91.Kaplan A, Franzen MD, Nickell PV, Ransom D, Lebovitz PJ. An open-label trial of duloxetine in patients with irritable bowel syndrome and comorbid generalized anxiety disorder. Int J Psychiatry Clin Pract 2014;18(1):11–15. [DOI] [PubMed] [Google Scholar]
  • 92.Lewis-Fernández R, Lam P, Lucak S, et al. An open-label pilot study of duloxetine in patients with irritable bowel syndrome and comorbid major depressive disorder. J Clin Psychopharmacol 2016;36(6):710–715. [DOI] [PubMed] [Google Scholar]
  • 93.Lee CW, Lin CL, Sung FC, Liang JA, Kao CH. Antidepressant treatment and risk of dementia: a population-based, retrospective case-control study. J Clin Psychiatr. 2016;77(1):117–122. [DOI] [PubMed] [Google Scholar]
  • 94.Breining A, Bonnet-Zamponi D, Zerah L, et al. Exposure to psychotropics in the French older population living with dementia: a nationwide population-based study. Int J Geriatr Psychiatr. 2017;32(7):750–760. [DOI] [PubMed] [Google Scholar]
  • 95.Camilleri M, Lembo A, Katzka DA. Opioids in gastroenterology: treating adverse effects and creating therapeutic benefits. Clin Gastroenterol Hepatol. 2017;15(9):1338–1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kasich AM. Treatment of diarrhea in irritable colon, including preliminary observations with a new antidiarrheal agent, diphenoxylate hydrochloride (Lomotil). Am J Gastroenterol. 1961;35(Jan.): 46–49. [PubMed] [Google Scholar]
  • 97.Lavo B, Stenstam M, Nielsen AL. Loperamide in treatment of irritable bowel syndrome - A double-blind placebo controlled study. Scand J Gastroenterol Suppl. 1987;130:77–80. [DOI] [PubMed] [Google Scholar]
  • 98.Hovdenak N Loperamide treatment of the irritable bowel syndrome. Scand J Gastroenterol Suppl. 1987;130:81–84. [DOI] [PubMed] [Google Scholar]
  • 99.Efskind PS, Bernklev T, Vatn MH. A double-blind placebo-controlled trial with loperamide in irritable bowel syndrome. Scand J Gastroenterol. 1996;31(5):463–468. [DOI] [PubMed] [Google Scholar]
  • 100.Dove LS, Lembo A, Randall CW, et al. Eluxadoline benefits patients with irritable bowel syndrome with diarrhea in a phase 2 study. Gastroenterology 2013;145(2):329–338, e1. [DOI] [PubMed] [Google Scholar]
  • 101.Lembo AJ, Lacy BE, Zuckerman MJ, et al. Eluxadoline for irritable bowel syndrome with diarrhea. N Engl J Med. 2016;374(3):242–253. [DOI] [PubMed] [Google Scholar]
  • 102.Wouters MM, Vicario M, Santos J. The role of mast cells in functional GI disorders. Gut 2016;65(1):155–168. [DOI] [PubMed] [Google Scholar]
  • 103.Wouters MM, Balemans D, Van Wanrooy S, et al. Histamine receptor H1-mediated sensitization of TRPV1 mediates visceral hypersensitivity and symptoms in patients with irritable bowel syndrome. Gastroenterology 2016;150(4):875–887, e9. [DOI] [PubMed] [Google Scholar]
  • 104.Lee KJ, Kim JH, Cho SW. Gabapentin reduces rectal mechanosensitivity and increases rectal compliance in patients with diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther. 2005;22(10):981–988. [DOI] [PubMed] [Google Scholar]
  • 105.Iturrino J, Camilleri M, Busciglio I, Burton D, Zinsmeister AR. Effect of the α2δ ligand, pregabalin, on colonic sensory and motor functions in healthy adults. Am J Physiol. 2011;301(2):G377–G378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Houghton LA, Fell C, Whorwell PJ, Jones I, Sudworth DP, Gale JD. Effect of a second- generation alpha2delta ligand (pregabalin) on visceral sensation in hypersensitive patients with irritable bowel syndrome. Gut 2007;56(9):1218–1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Saito YA, Almazar AE, Tilkes K, et al. A placebo-controlled trial of pregabalin for irritable bowel syndrome. Am J Gastroenterol. 2016;111(9):S236 (abstract).27685295 [Google Scholar]
  • 108.Camilleri M Towards an effective peripheral visceral analgesic: responding to the national opioid crisis. Am J Physiol Gastrointest Liver Physiol. 2018. February 22. doi: 10.1152/ajpgi.00013.2018. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Berger M, Gray JA, Roth BL. The expanded biology of serotonin. Annu Rev Med. 2009;60(Feb.):355–366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Gershon MD, Wade PR, Kirchgessner AL, Tamir H. 5-HT receptor subtypes outside the central nervous system. Roles in the physiology of the gut. Neuropsychopharmacology 1990;3(5–6):385–395. [PubMed] [Google Scholar]
  • 111.Atkinson W, Lockhart S, Whorwell PJ, Keevil B, Houghton LA. Altered 5-hydroxytryptamine signaling in patients with constipation- and diarrhea-predominant irritable bowel syndrome. Gastroenterology 2006;130(1):34–43. [DOI] [PubMed] [Google Scholar]
  • 112.Viramontes BE, Camilleri M, McKinzie S, Pardi DS, Burton D, Thomforde GM. Gender-related differences in slowing colonic transit by a 5-HT3 antagonist in subjects with diarrhea-predominant irritable bowel syndrome. Am J Gastroenterol. 2001;96(9):2671–2676. [DOI] [PubMed] [Google Scholar]
  • 113.Hicks GA, Coldwell JR, Schindler M, et al. Excitation of rat colonic afferent fibres by 5-HT(3) receptors. J Physiol. 2002;544(Pt 3):861–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Andresen V, Montori VM, Keller J, West CP, Layer P, Camilleri M. Effects of 5-hydroxytryptamine (serotonin) type 3 antagonists on symptom relief and constipation in nonconstipated irritable bowel syndrome: A systematic review and meta-analysis of randomised controlled trials. Clin Gastroenterol Hepatol. 2008;6(5):545–555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Ford AC, Brandt LJ, Young C, Chey WD, Foxx-Orenstein AE, Moayyedi P. Efficacy of 5-HT-3 antagonists and 5-HT-4 agonists in irritable bowel syndrome: Systematic review and meta-analysis. Am J Gastroenterol. 2009;104(7):1831–1843. [DOI] [PubMed] [Google Scholar]
  • 116.Fukudo S, Kinoshita Y, Okumura T, et al. Ramosetron reduces symptoms of irritable bowel syndrome with diarrhea and improves quality of life in women. Gastroenterology 2016;150(2):358–366. [DOI] [PubMed] [Google Scholar]
  • 117.Matsueda K, Harasawa S, Hongo M, Hiwatashi N, Sasaki D. A randomized, double-blind, placebo-controlled clinical trial of the effectiveness of the novel serotonin type 3 receptor antagonist ramosetron in both male and female Japanese patients with diarrhea-predominant irritable bowel syndrome. Scand J Gastroenterol. 2008;43(10):1202–1211. [DOI] [PubMed] [Google Scholar]
  • 118.Garsed K, Chernova J, Hastings M, et al. A randomised trial of ondansetron for the treatment of irritable bowel syndrome with diarrhoea. Gut 2014;63(10):1617–1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chang L, Chey WD, Harris L, Olden K, Surawicz C, Schoenfeld P. Incidence of ischemic colitis and serious complications of constipation among patients using alosetron: Systematic review of clinical trials and post-surveillance marketing data. Am J Gastroenterol. 2006; 101(5): 1069–1079. [DOI] [PubMed] [Google Scholar]
  • 120.Aziz I, Mumtaz S, Bholah H, Chowdhury FU, Sanders DS, Ford AC. High prevalence of idiopathic bile acid diarrhea among patients with diarrhea-predominant irritable bowel syndrome based on Rome III criteria. Clin Gastroenterol Hepatol. 2015;13(9):1650–1655. [DOI] [PubMed] [Google Scholar]
  • 121.Valentin N, Camilleri M, Altayar O, et al. Biomarkers for bile acid diarrhea in functional bowel disorder with diarrhea: a systematic review and meta-analysis. Gut 2016;65(12):1951–1959. [DOI] [PubMed] [Google Scholar]
  • 122.Camilleri M, Acosta A, Busciglio I, et al. Effect of colesevelam on faecal bile acids and bowel functions in diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther. 2015;41(5):438–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bajor A, Tornblom H, Rudling M, Ung KA, Simren M. Increased colonic bile acid exposure: A relevant factor for symptoms and treatment in IBS. Gut 2015;64(1):84–92. [DOI] [PubMed] [Google Scholar]
  • 124.Pimentel M, Lembo A, Chey WD, et al. Rifaximin therapy for patients with irritable bowel syndrome without constipation. N Engl J Med. 2011;364(1):22–32. [DOI] [PubMed] [Google Scholar]
  • 125.Pimentel M, Chow EJ, Lin HC. Eradication of small intestinal bacterial overgrowth reduces symptoms of irritable bowel syndrome. Am J Gastroenterol. 2000;95(12):3503–3506. [DOI] [PubMed] [Google Scholar]
  • 126.Menees SB, Maneerattanaporn M, Kim HM, Chey WD. The efficacy and safety of rifaximin for the irritable bowel syndrome: A systematic review and meta-analysis. Am J Gastroenterol. 2012;107(1):28–35. [DOI] [PubMed] [Google Scholar]
  • 127.Acosta A, Camilleri M, Shin A, et al. Effects of rifaximin on transit, permeability, fecal microbiome, and organic acid excretion in irritable bowel syndrome. Gastroenterology 2016;150(Suppl. 1):S948–S949 (abstract). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Pimentel M, Chang C, Chua KS, et al. Antibiotic treatment of constipation-predominant irritable bowel syndrome. Dig Dis Sci. 2014;59(6):1278–1285. [DOI] [PubMed] [Google Scholar]
  • 129.Camilleri M, Bharucha AE, Ueno R, et al. Effect of a selective chloride channel activator, lubiprostone, on gastrointestinal transit, gastric sensory, and motor functions in healthy volunteers. Am J Physiol Gastrointest Liver Physiol. 2006;290(5):G942–G947. [DOI] [PubMed] [Google Scholar]
  • 130.Drossman DA, Chey WD, Johanson JF, et al. Clinical trial: lubiprostone in patients with constipation-associated irritable bowel syndrome - results of two randomized, placebo-controlled studies. Aliment Pharmacol Ther. 2009;29(3):329–341. [DOI] [PubMed] [Google Scholar]
  • 131.Johanson JF, Morton D, Geenen J, Ueno R. Multicenter, 4-week, double-blind, randomized, placebo-controlled trial of lubiprostone, a locally-acting type-2 chloride channel activator, in patients with chronic constipation. Am J Gastroenterol. 2008;103(1):170–177. [DOI] [PubMed] [Google Scholar]
  • 132.Fukudo S, Hongo M, Kaneko H, Takano M, Ueno R. Lubiprostone increases spontaneous bowel movement frequency and quality of life in patients with chronic idiopathic constipation. Clin Gastroenterol Hepatol. 2015;13(2):294–301. [DOI] [PubMed] [Google Scholar]
  • 133.Cryer B, Drossman D, Chey W, Webster L, Habibi S, Wang M. Analysis of nausea in clinical studies of lubiprostone for the treatment of constipation disorders. Dig Dis Sci. 2017;62(12):3568–3578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Lembo AJ, Schneier HA, Shiff SJ, et al. Two randomized trials of linaclotide for chronic constipation. N Engl J Med. 2011;365(6):527–536. [DOI] [PubMed] [Google Scholar]
  • 135.Chey WD, Lembo AJ, Lavins BJ, et al. Linaclotide for irritable bowel syndrome with constipation: A 26-week, randomized, double-blind, placebo-controlled trial to evaluate efficacy and safety. Am J Gastroenterol. 2012;107(11):1702–1712. [DOI] [PubMed] [Google Scholar]
  • 136.Rao S, Lembo AJ, Shiff SJ, et al. 12-week, randomized, controlled trial with a 4-week randomized withdrawal period to evaluate the efficacy and safety of linaclotide in irritable bowel syndrome with constipation. Am J Gastroenterol. 2012;107(11): 1714–1724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Shailubhai K, Barrow L, Talluto C, et al. Plecanatide, a guanylate cyclase C agonist improves bowel habits and symptoms associated with chronic constipation in a phase IIa clinical study. Am J Gastroenterol. 2011;106 (Suppl. 2s):S502 (abstract). [Google Scholar]
  • 138.Miner PB Jr, Koltun WD, Wiener GJ, et al. A randomized phase III clinical trial of plecanatide, a uroguanylin analog, in patients with chronic idiopathic constipation. Am J Gastroenterol. 2017;112(4): 613–621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Fogel R, Dorn SD, Krause R, et al. Efficacy and safety of plecanatide in patients with irritable bowel syndrome with constipation: results from 2 randomized, double-blind, placebo-controlled clinical trials. Gastroenterology 2017;152 (Suppl. 1):S1309–S1310 (abstract). [Google Scholar]
  • 140.Layer P, Keller J, Loeffler H, Kreiss A. Tegaserod in the treatment of irritable bowel syndrome (IBS) with constipation as the prime symptom. Ther Clin Risk Manag. 2007;3(1):107–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Loughlin J, Quinn S, Rivero E, et al. Tegaserod and the risk of cardiovascular ischemic events: an observational cohort study. J Cardiovasc Pharmacol Ther. 2010; 15(2): 151–157. [DOI] [PubMed] [Google Scholar]
  • 142.Nakamura M, Ohmiya N, Miyahara R, et al. Are symptomatic changes in irritable bowel syndrome correlated with the capsule endoscopy transit time? A pilot study using the 5-HT4 receptor agonist mosapride. Hepatogastroenterology 2011;58(106):453–458. [PubMed] [Google Scholar]
  • 143.Mansour NM, Ghaith O, El-Halabi M, Sharara AI. A prospective randomized trial of mosapride vs. placebo in constipation-predominant irritable bowel syndrome. Am J Gastroenterol. 2012;107(5):792–793. [DOI] [PubMed] [Google Scholar]
  • 144.Radziwon CD, Lackner JM. Cognitive behavioral therapy for IBS: How useful, how often, and how does it work? Curr Gastroenterol Rep. 2017;19(10):49. [DOI] [PubMed] [Google Scholar]

RESOURCES