Abstract
Functional bowel disorders (i.e., constipation and diarrhea) are characterized by abdominal pain, bloating, distention, and/or bowel habit abnormalities in the absence of obvious anatomic or physiologic abnormalities on routine diagnostic tests. These symptoms are attributable to gastrointestinal sensorimotor dysfunctions resulting from peripheral and/or central mechanisms. Available drugs target the underlying bowel disturbance (ie, constipation, diarrhea, or both), supplemented when necessary by management of pain. Osmotic and stimulant laxatives, secretagogues, and serotonin 5-HT4 receptor agonists are approved for treating constipation. Loperamide, anticholinergic agents, rifaximin, bile-acid binding agents, eluxadoline, and clonidine are used to treat diarrhea. Several exciting new compounds, some of which have been evaluated in humans, are currently under development.
Introduction
Functional bowel disorders (FBD) comprise a spectrum of chronic gastrointestinal (GI) disorders characterized by abdominal pain, bloating, distention, and/or bowel habit abnormalities (e.g, constipation, diarrhea, or mixed constipation and diarrhea) in the absence of obvious anatomic or physiologic abnormalities on routine diagnostic tests. The pathophysiology of constipation and diarrhea are covered elsewhere.[1,2] This article reviews the current management, especially medications and emerging therapies that have been studied in the past 2 years (Table 1). Other and older investigational medications are discussed elsewhere.[3]
Table 1.
Unapproved and Recently Approved Drugs in Pre-clinical and Clinical Studies of IBS1
| Drug class | Examples2 | Mechanisms | Species and model3 | Effects3 |
|---|---|---|---|---|
| Drugs tested in humans and belonging to classes in which other drugs are approved for use in humans | ||||
| Antibiotics[16,19,21] | Rifaximin (approved), rifamycin | Poorly absorbable antibiotics with anti-inflammatory properties, potentially modulating gut microbiota | Placebo-controlled clinical phase 3 trials in IBS-D | Improved diarrhea, bloating, and abdominal pain |
| Serotonin 5HT3 antagonist [24,25,53–55] | Ramosetron (approved in Japan), cilansetron, ondansetron, and alosetron (approved) | 5HT3 receptor is expressed by peripheral and central neurons. Selective antagonism of peripheral 5HT3 receptors decreases gut transit, increases fluid absorption, and reduce visceral nociception | Placebo-controlled mechanistic studies in IBS-D | Reduced visceral sensation, slowed colonic transit, and reduced contractile and tonic responses to meal ingestion |
| Placebo-controlled phase 3 clinical trials in IBS-D | Improved stool consistency and abdominal pain | |||
| Opioids, cannabinoids [16,56,57] | Methyl-orvinol (μ- and κ-opioid receptor agonist, δ- opioid receptor antagonist) | Opiod receptors are present in ENS, ICC and smooth muscle cells | Mouse model of mustard oil-induced visceral hypersensitivity | Improved gastrointestinal hypermotility and reduced abdominal pain |
| Eluxadoline (μ- opioid receptor agonist, δ-opioid receptor antagonist; (approved) | Placebo-controlled phase 3 clinical trials in IBS-D | Inhibited colonic transit and reduced intestinal fluid/ion secretion | ||
| Palmitoyl- ethanolamide (PEA) | PEA is a fatty acid amide acting on peroxisome-activated receptor-γ (PPARγ) and cannabinoid-like G-protein- coupled receptors (GPCRs), potentially on mast cells | Placebo-controlled trial of 54 patients with IBS and 12 healthy volunteers | Reduced severity of abdominal pain without affecting mast cell counts | |
| Bile acid modulators [26–29] | Bile acid sequestrants (cholestyramine, colestipol, and colesevelam, all approved for other indications) Farnesoid X receptor (FXR) agonist (obeticholic acid) | Sequestrants bind to bile acids, increase fecal excretion of bile acids, and increase synthesis of new bile acids. Fecal bile acids bound to colesevelam are biologically inactive. By binding to ileal FXRs, obeticholic acid, a FXR agonist, increases the ileal production of fibroblast growth factor 19 which reduces the production of bile acids. | Uncontrolled studies of colsevelam (total of 36 patients), colestipol (27 patients), and obetichloic acid (28 patients) | Colsevelam, colestipol, and obetichloic acid reduced the frequency of bowel movements. The effects on stool consistency varied among studies. The effects of colesevelam on colonic transit were not significant. |
| Drugs tested in humans. No other drugs in these classes are approved for use in humans with functional bowel disorders | ||||
| Neurokinin-2 (NK2) receptor antagonist [31] | Ibodutant | NK2 is expressed by smooth muscle cells of the muscularis mucosae and propria and few inflammatory cells of the lamina propria. It is not involved in visceromotor function | Placebo controlled study in 559 patients with IBS-D | Improved stool consistency in women but not men |
| Translocator protein 18 kDa (TSPO) antagonist [32,33] | ONO-2952 | TSPO is involved in steroidogenesis and transport of neurosteroids | Rat model with restraint stress | Suppressed restraint stress-induced defecation |
| Placebo-controlled study in 200 patients with IBS | Tendency to improve abdominal pain, stool consistency and stool frequency | |||
| Somatostatin analogs [34] | Lanreotide (approved for other indications) | Binds somatostatin receptors (types 2 and 5), which inhibits intestinal secretion and synthesis of growth hormone | 33 patients with idiopathic refractory diarrhea | Decreased stool frequency and improved quality of life |
| Histamine receptor antagonists [35,58] | Histamine receptor H1 (HRH1) antagonists (pyrilamine, ebastine, approved ebastine, approved for other indications in some countries) HRH1 and HRH4 antagonist (levocetirizine, JNJ7777120) |
Inhibition of neuronal expressed HRH1 prevents sensitization of transient receptor potential cation channel 1 TRPV1 channels HRH4s are expressed on mast cells and immune cells. HRH4s modulate neuronal sensitivity via indirect mechanisms. |
Human submucosal neurons and a placebo - controlled trial of 55 patients with IBS | Prevented sensitization of TRPV1 channels in human submucosal neurons. Reduced IBS symptoms and visceral pain perception in patients with IBS. |
| Rat model with 2,4,6- trinitrobenzenesulfonic acid (TNBS) colitis and post inflammatory visceral hypersensitivity | Normalizes post-inflammatory visceral hypersensitivity | |||
| TRPM8 agonist [38] | Pepper mint oil | TRPM8 couples to TRPV1 and A1 to inhibit downstream chemo- and mechanosensory actions | Rat model: TNBS colitis with post-inflammatory visceral hypersensitivity | Decreased post-inflammatory visceral hypersensitivity |
| Placebo-controlled trial in 72 patients with IBS | Decreased abdominal pain and increased quality of life | |||
| Protease-activated receptor (PAR)-targeting molecules and serine protease inhibitors [39,41,59] | PAR2 antagonist (ENMD-1068) PAR-4 agonist (PAR-4-AP, Cat-G, AYPGKF-NH2) Aprotinin, cathepsin-G inhibitor, soybean trypsin inhibitor | (epithelium derived) proteases are upregulated in IBS, they signal to enteric neurons leading to visceral hypersensitivity | Human submucosal neurons and mice receiving intracolonic instillation with IBS- supernatant | Decreased visceral hypersensitivity |
| Vitamin D3 [52] | Vitamin D inhibits T-cell proliferation and is capable of inhibiting the immune response | Placebo controlled trial in 90 patients with IBS | Improved abdominal pain and distention, flatulence, overall gastrointestinal symptoms (except dissatisfaction with bowel habits) and quality of life | |
| IgE blockade [42–44] | Omalizumab (approved for other indications) | Monoclonal antibody that binds IgE receptors on mast cells and basophils | Case reports in IBS-D patients | Almost complete resolution of symptoms in patients with IBS-D |
| Immunotherapy [46,47] | Serum-derived bovine immunoglobulin/pr otein isolate therapy (SBI, approved) | Intestinal co-culture | Immunoglobulins in SBI reduce antigen-associated inflammation through immune and steric exclusion mechanisms | |
| Uncontrolled study in 15 patients with IBS-D | Improved severity of abdominal pain and stool frequency | |||
| Drugs tested in animal models only | ||||
| G protein-coupled estrogen receptor (GPER) ligands [60] | G-1, a GPER selective agonist, and estradiol, a nonselective ER agonist | Inhibition of estrogen receptors dampens the cholinergic excitatory neuronal pathway | Human colonic muscle strips Mouse model with mustard oil-induced abdominal pain | G-1 and estradiol inhibit colonic motility and improve visceral pain in mouse model of visceral hypersensitivity |
Abbreviations: ENS: enteric nervous system, ICC: interstitial cells of Cajal, IBS: irritable bowel syndrome, IBS-D: diarrhea- predominant IBS
The emphasis is on drugs and drug classes that have been studied in the past 2 years.
Unless stated otherwise, approved refers to approval for bowel disorders by regulatory agencies in one or more countries.
If data from human studies are available, pre-clinical studies are not provided unless essential.
Constipation and Constipation-Predominant IBS (IBS-C)
Laxatives
Initially, treatment with increased dietary fiber intake and/or an inexpensive osmotic agent, such as milk of magnesia or polyethylene glycol, supplemented, if necessary, with stimulant laxatives (e.g., bisacodyl suppositories) is recommended.[2] Laxative non-responders should undergo anorectal tests to evaluate for a defecatory disorder, for which pelvic floor biofeedback therapy is required. Absent defecatory disorders, the next option is a secretagogue or prucalopride.
Secretagogues
Secretagogues (i.e., lubiprostone, linaclotide, and plecanatide) increase intestinal chloride secretion by activating channels on the apical enterocyte surface, causing net efflux of ions and water into the intestine. To maintain electroneutrality, sodium is also secreted into the intestine; water follows.
Lubiprostone, is a bicyclic fatty acid derivative derived from prostaglandin E1 that activates apical type 2 chloride channels (CIC-2). Lubiprostone also activates prostaglandin EP receptors and the apical cystic fibrosis transmembrane regulator (CFTR); the latter also mediates intestinal fluid secretion. Lubiprostone is approved by the Food and Drug Administration (FDA) and in Europe for treating chronic constipation and C-IBS. Women of childbearing age require a negative pregnancy test before starting and contraceptive measures during treatment.
Similar to the natriuretic peptides uroguanylin and guanylin and the heat-stable enterotoxin (ST peptide) produced by the pathogenic Escherichia coli (E. coli), linaclotide and plecanatide are peptide agonists at guanylate cyclase C (GC-C) receptors.[4] They stimulate intracellular production of cyclic guanosine monophosphate (cGMP), leading to activation of CFTR, with chloride and water secretion. Activation of GC-C receptors by uroguanylin and plecanatide is pH-dependent, regulated by mucosal acidity in the proximal intestine. By contrast, the effects of ST-peptide and linaclotide are pH-independent. The extent to which these differences in pH-dependency between linaclotide and plecanatide influence the response and side effect profile is unclear and controversial.[5] Both drugs have minimal oral bioavailability and are FDA approved for treating chronic constipation. Linaclotide is also approved for treating constipation-predominant IBS. Diarrhea is the main side effect.
Prucalopride
Prucalopride is a selective serotonin 5-HT4 receptor agonist, of a chemical class that differs from other older non-selective 5-HT4 receptor ligands like metoclopramide, cisapride and tegaserod.[6] Prucalopride stimulates gastric, small intestinal and colonic propulsive contractions, including colonic high amplitude propagated contractions, which precede defecation. Prucalopride improves bowel disturbances and abdominal symptoms such as bloating and pain.[7] Outside the United States, prucalopride is approved for treating laxative-unresponsive chronic constipation in men and women. The main, usually transient, side effects of prucalopride are headache, diarrhea and nausea.
Newer Agents in Clinical and Pre-Clinical studies
Elobixibat
Elobixibat is a first-in-class drug developed for treating chronic constipation and constipation-predominant irritable bowel syndrome (IBS-C).[8] By inhibiting the ileal bile acid transporter, hence its absorption, elobixibat allows bile to promote colonic secretion and contractions, thereby accelerating colonic transit. A placebo-controlled phase 2b study showed significant symptom improvement with 10 and 15 mg elobixibat daily. An international phase 3 program with elobixibat in chronic constipation (NCT01833065 and NCT01827592) was prematurely terminated because of distribution issues with the trial medication. Based on a positive phase 3 study, a new drug application was submitted in Japan in early 2017. The drug has not been evaluated in IBS.
Tenapanor
Tenapanor is a first-in class inhibitor of the enterocyte sodium/hydrogen exchanger isoform 3 (NHE3).[9] Tenapanor is minimally absorbed, acts from the lumen to inhibit sodium and phosphate absorption, which increases intestinal fluid volume and transit. In a 12-week phase 2 study in IBS-C, tenapanor (5–50 mg b.i.d.) dose-dependently improved constipation and abdominal symptoms; the 50 mg b.i.d. dose was significantly better than placebo.[10] A press release announced that the first phase 3 trial in IBS-C with tenapanor met its primary endpoint (combined pain and stool pattern responder rate 27% vs. 18.7% with placebo, p=0.02). Significance was met for the secondary endpoint of abdominal pain relief, but not for the complete spontaneous bowel movements endpoint.
Diarrhea and Diarrhea-Predominant IBS (IBS-D)
Dietary modifications
In IBS-D, diets with a low content of fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) provide greater symptom relief than placebo but not when compared to a common sense diet.[11] Also, these diets have profound, potentially deleterious, effects on the colonic microbiome.[12] Oligosaccharides with an alpha-galactosidic linkage have been implicated in the generation of symptoms after carbohydrate rich meals. In a twelve week placebo-controlled trial of alpha-galactosidase 400 IU capsules t.i.d. in 125 IBS patients, the supplement was no better than placebo and the withdrawal rate was higher in the active group.[13]
Antispasmodics and antidiarrheal agents
Pooled analyses suggest that antispasmodics (e.g., hyoscyamine, dicyclomine, peppermint oil) and antidepressants (e.g., serotonin reuptake inhibitors [SSRIs] and tricyclic antidepressants [TCAs]) improve abdominal pain and other IBS symptoms.[11] The non-absorbed μ opioid agonist loperamide (starting dose of 2 mg given 30 minutes before meals, maximum dose 16 mg daily), is an inexpensive initial approach for patients with diarrhea and a prominent gastrocolonic response. In some patients, finding the right balance between diarrhea and constipation, which is a side effect of loperamide, is challenging. In loperamide nonresponders, eluxadoline, rifaximin, and clonidine are options in men and women, and alosetron in women.[11,14,15] Except for clonidine, these drugs have been approved by the FDA; eluxadoline and rifaximin were approved in May 2015. Because alosetron was associated with ischemic colitis, prescriptions for this drug in the United states require additional precautions detailed elsewhere.[11]
Eluxadoline
Eluxadoline is a μ-opioid receptor agonist and δ-opioid receptor antagonist that decreases bowel contractions, inhibits colonic transit, and reduces fluid/ion secretion. Oral bioavailability is 1.0%.[16] In phase 3 studies, the improvement in abdominal pain and diarrhea over 26 weeks was significantly higher for eluxadoline 100 mg b.i.d. than placebo (i.e., 29.3% vs. 19.0%, respectively in study 1 and 32.7% vs. 20.2% in study 2). Even in loperamide non-responders, eluxadoline was more effective than placebo.[17] The 75 mg twice-daily dose was also better versus placebo in both studies but differences were only statistically significant in study 2. Constipation, nausea, and abdominal pain were the most common adverse events. Sphincter of Oddi spasm, pancreatitis or elevated hepatic enzymes are less common but more serious side effects. In clinical trials, this adverse event occurred during the first week of treatment and resolved with drug discontinuation. In the initial FDA approval, eluxadoline was contraindicated in patients who had a history of pancreatitis or who consumed >3 alcoholic drinks per day; the lower dose (75 mg twice daily) was recommended in patients who had a cholecystectomy. Thereafter, that is between May 2015 and February 2017, 120 serious cases of pancreatitis, of which 76 required hospitalization and 2 died, were reported to the FDA database.[18] These serious side effects occurred early (e.g., after one or two doses) or with prolonged use. Of 68 cases that reported gallbladder status, 56 cases of pancreatitis or death occurred in patients who do not have a gallbladder. Hence, the FDA forbids the use of eluxadoline in patients who have had a cholecystectomy.
Rifaximin
Rifaximin is a nonsystemic antibiotic. In two identical, 12-week, phase 3 trials (TARGET 1 and 2, pooled N = 1260), in IBS-D patients treated with rifaximin (550 mg t.i.d. for 2 weeks) were more likely than placebo to report adequate relief of global IBS symptoms for 2 weeks of the first 4-week post-treatment period (i.e., 40.7% vs. 31.7%) and also at 10 weeks post-treatment.[19] Adequate relief was defined by an affirmative response to the question: “In regard to all your symptoms of IBS, as compared with the way you felt before you started the study medication, have you, in the past 7 days, had adequate relief of your IBS symptoms?” Subsequently, the TARGET 3 trial evaluated a second course of rifaximin in patients who relapsed after a treatment with rifaximin. Of 1074 patients (44.1%) who responded to open-label rifaximin, 692 (64.4%) relapsed within 18 weeks thereafter; 636 were randomized to the same regimen of rifaximin (n = 328) or placebo (n = 308) as in TARGET 1 and 2. During the 4 week follow-up period, response rates, as defined by improved abdominal pain and stool consistency, was significantly greater with rifaximin than placebo (38.1% vs 31.5%). Six weeks after the first retreatment, all patients were retreated again; again, the response to rifaximin was greater than placebo.[16] Adverse events, primarily headache, upper respiratory tract infection, and abdominal pain, were uncommon and similar between groups. No cases of Clostridium difficile infection were reported in these or subsequent studies but patients were not routinely tested. In summary, rifaximin is safe, modestly effective, and expensive (approximate cost of $50 daily). The FDA has only approved 3 courses of rifaximin. However, in practice, many patients are treated indefinitely, which is of concern. Antimicrobial effects probably mediate the response to rifaximin. The impetus to trials with rifaximin originated from studies which suggested that IBS-D was associated with small intestinal bacterial overgrowth (SIBO).[20] However, the tests used in routine clinical practice to diagnose SIBO lack standardization and validity, hence the contribution of SIBO to IBS is debatable. Moreover, trials of rifaximin in IBS did not evaluate for the presence of SIBO.
Similar to rifaximin, the efficacy of 2 weeks treatment with the poorly absorbable antibiotic rifamycin 600 mg b.i.d. or t.i.d. is being evaluated in IBS-D.[21]
Serotonin 5HT3 antagonists
The 5HT3-antagonist alosetron reduces abdominal pain and diarrhea in IBS-D patients.[22] Because it can cause, albeit rarely, severe constipation and ischemic colitis, it is approved for limited use, including the United States. However, the experience in clinical practice is largely reassuring.[23] Another 5HT3-antagonist, ramosetron, with mainly peripheral effects, is efficacious in IBS-D, has a low incidence of constipation, does not cause ischemic colitis, and is approved in several Asian countries. In a phase III study in 296 male patients with IBS-D, overall IBS symptom relief (31.3 vs. 9.5%), improvement of stool consistency and abdominal pain relief were greater for ramosetron than placebo.[24] Similar effects were observed in a phase III study of 576 female IBS-D patients.[25]
Bile acid modulators
Bile acids stimulate colonic secretion and motility causing diarrhea in up to one third of patients with IBS-D. Uncontrolled studies suggest that bile acid sequestrants (e.g., cholestyramine, colestipol, and colesevelam) reduce colonic transit and improve stool consistency in these patients.[26–28] Obeticholic acid (or 6-ethyl chenodeoxycholic acid), a farnesoid X receptor agonist, increases enterocyte synthesis of the hormone fibroblast growth factor (FGF)-19 and upregulates the feedback inhibition of hepatic bile acid synthesis.[29] In a single-center, pilot study, obeticholic acid was well tolerated, significantly reduced bile acid synthesis, and improved stool form and frequency.
Tachykinins
Tachykinins, a family of neuropeptides including substance P, neurokinin (NK) A and B, are important mediators of gastrointestinal motility and nociception.[30] Ibodutant, a selective neurokinin-2 receptor antagonist (1,3 and 10 mg daily), was evaluated in a placebo-controlled phase 2 study in 559 IBS-D patients. In the overall population, the 10 mg dose tended to show benefit over placebo (39.6% vs. 27.5% responder rate, p=0.032, but not after correction for multiple testing). In a pre-specified analysis by gender, there was a dose-response effect in females; ibodutant (10 mg) was superior than placebo (46.8% vs. 24.4% response rate, p= 0.003, significant after correction for multiple testing).[31] A phase 3 program with ibodutant (10 mg) in women with IBS-D is being conducted in Europe and in the U.S.A.
Translocator protein antagonists
ONO-2952, which is a novel and selective high affinity antagonist for the Translocator Protein 18 kDa (TSPO), influences neuronal excitability. It decreased visceral hypersensitivity in animal models.[32] In a 4-week study in IBS-D with approximately 65 patients per arm, ONO-2952 (60 mg) tended to improve stool consistency and diarrhea, but effects were not statistically significant versus placebo.[33]
Somatostatin
Somatostatin reduces diarrhea by inhibiting gastrointestinal motility and secretion. In a study of 33 patients with chronic idiopathic diarrhea (daily stool volume greater than 200 grams) treated with the long-acting somatostastin analogue lanreotide 120 mg s.c. every 4 weeks for 3 months, lanreotide significantly improved stool frequency and consistency and quality of life; 42% of patients had a more than 50% reduction in stool frequency after 1 month.[34]
Histamine receptor antagonists and other agents acting on TRP (Transient Receptor Potential) channels
Histamine sensitizes the nociceptor transient receptor potential cation channel V1 (TRPV1) and contributes to visceral hypersensitivity in preclinical models. In IBS patients, there is sensitization of TRPV1 channels, which is mediated by histamine and/or its metabolite imidazole acetaldehyde, in the rectal submucosal plexus.[35,36] In a 12-week, double-blind, placebo-controlled study, the HRH1 receptor antagonist ebastine (20mg once daily) reduced visceral hypersensitivity, and improved symptoms, including abdominal pain, versus placebo.[35] (Table 1). A large multicenter study is in progress.
TRPM8 agonists couple to TRPV1 and A1, inhibit their downstream chemo- and mechanosensory actions, and reduce post inflammatory visceral hyperalgesia in rats.[37] In a short (4 weeks) study in 72 patients, peppermint oil, a TRPM8 agonist, was more effective than placebo in IBS-D and IBS-M; the unconventional primary endpoint was the change from baseline in the Total IBS Symptom Score (TISS) after 4 weeks of treatment.[38]
Protease activating receptor 2 (PAR2) antagonists
In animal models of visceral hypersensitivity, PAR-2 antagonists and a PAR-4 agonist reversed colonic visceral hypersensitivity induced by supernatants from colonic biopsies from IBS patients.[39,40] Perhaps proteases such as trypsin 3 released from the epithelium of IBS patients activated sensory nerve endings.[41] Human studies are awaited.
IgE blockade
Case reports suggest that omalizumab improved IBS symptoms in IBS-D.[42–44] This non-anaphylactogenic anti-IgE antibody prevents free serum IgE from binding to cellular receptors on mast cells and basophils. While FGID patients are not allergic to food allergens, some have increased jejunal mucosal humoral responses. [45]
Other agents
A serum-derived bovine immunoglobulin/protein isolate (SBI) reduced stool frequency and abdominal pain but did not affect small bowel markers of immunity or inflammation in an uncontrolled study of 15 IBS-D patients.[46] The FDA regards SBI as a safe prescription medical food product. In vitro studies demonstrated that immunoglobulins in SBI reduce antigen-associated inflammation through immune and steric exclusion mechanisms.[47]
Palmitoylethanolamide (PEA) is a agonist at cannabinoid-2 and peroxisome proliferator-activated receptor-α (PPAR-α)- receptors, and stabilizes mast cells. A 12-week trial in 54 IBS patients observed that a food additive containing PEA with polydatin (200 mg/20 mg), which may also stabilize mast cells, improved abdominal pain more than placebo.[48] Whether these effects are mediated via mast cells or PPAR-alpha-receptors on enteric glial cells, which have also been implicated in animal models of postoperative ileus and IBS is unclear.[49,50] Indeed, activation of PPAR-alpha receptors in enteric glia by PEA improves colon inflammation through enteric glia/toll like receptor 4-dependent PPAR-alpha activation.[51]
In a placebo-controlled trial from Iran in 90 IBS patients, vitamin D3 (50000 IU every fortnight) improved abdominal symptoms (e.g., pain and distention) and quality of life, but not dissatisfaction with bowel habits. Vitamin D levels stayed in the normal range in the treatment group.[52] At baseline, two thirds of the patients had vitamin D levels below 20 ng/ml. Future studies to confirm and determine the mechanism of action, perhaps immune modulatory or antidepressive, are warranted.
Summary
Several drugs which target an expanding repertoire of pharmacological targets have been approved or are under development for treating functional bowel disorders in humans. These agents target the underlying bowel disturbance (ie, constipation, diarrhea, or both), supplemented when necessary by management of pain. As new agents are developed, their incremental utility over existing compounds should also be evaluated.
Figure 1.

Receptors and cellular mechanisms of drugs approved for treating functional bowel disorders.
Highlights.
For constipation and diarrhea, stepwise therapy is recommended
-
When initial therapy is unsuccessful:
-
○
Secretagogues and 5-HT4 receptor agonists are used for chronic constipation
-
○
Bile-acid binding agents, eluxadoline, and rifaximin are used to treat diarrhea
-
○
Several newer compounds are being developed and tested in clinical trials
Acknowledgments
This study was supported in part by USPHS NIH Grants R01 DK78924 to AEB, by research grant G.0699.10N from the Fund for Scientific Research (FWO) Flanders (Belgium) to MMW.
Dr. Tack has given Scientific advice to Abide Therapeutics, AlfaWassermann, Allergan, Christian Hansen, Danone, Ironwood, Janssen, Kiowa Kirin, Menarini, Mylan, Novartis, Nutricia, Rhythm, Shionogi, Shire, SK Life Sciences, Takeda, Theravance, Tsumura, Yuhan, Zealand and Zeria pharmaceuticals, has received research grants or support from Abide Therapeutics, Shire, Tsumura, Zeria and has served on the Speaker bureau for Abbott, Allergan, AstraZeneca, Janssen, Kiowa Kirin, Menarini, Mylan, Novartis, Shire, Takeda and Zeria pharmaceuticals.
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.
Conflict of interest: Dr. Bharucha serves as a consultant for Allergan Pharmaceuticals. Dr. Wouters has no conflict of interest.
References
- 1.Camilleri M. Peripheral mechanisms in irritable bowel syndrome. New England Journal of Medicine. 2013;368:578–579. doi: 10.1056/NEJMc1214185. [DOI] [PubMed] [Google Scholar]
- 2.Bharucha AE, Locke GR, Pemberton JH. AGA Practice Guideline on Constipation:Technical Review. Gastroenterology. 2013;144:218–238. doi: 10.1053/j.gastro.2012.10.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wadhwa A, Camilleri M, Grover M. New and Investigational Agents for Irritable Bowel Syndrome. Current Gastroenterology Reports. 2015;17:46. doi: 10.1007/s11894-015-0473-x. [DOI] [PubMed] [Google Scholar]
- 4.Bharucha AE, Waldman SA. Taking a lesson from microbial diarrheagenesis in the management of chronic constipation. Gastroenterology. 2010;138:813–817. doi: 10.1053/j.gastro.2010.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Busby RW, Ortiz S. Clarification of linaclotide pharmacology presented in a recent clinical study of plecanatide. Digestive Diseases & Sciences. 2014;59:1066–1067. doi: 10.1007/s10620-013-2953-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Tack J, Camilleri M, Chang L, Chey WD, Galligan JJ, Lacy BE, Muller-Lissner S, Quigley EM, Schuurkes J, De Maeyer JH, et al. Systematic review: cardiovascular safety profile of 5-HT(4) agonists developed for gastrointestinal disorders. Alimentary Pharmacology & Therapeutics. 2012;35:745–767. doi: 10.1111/j.1365-2036.2012.05011.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tack J, Stanghellini V, Dubois D, Joseph A, Vandeplassche L, Kerstens R. Effect of prucalopride on symptoms of chronic constipation. Neurogastroenterology & Motility. 2014;26:21–27. doi: 10.1111/nmo.12217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chey WD, Camilleri M, Chang L, Rikner L, Graffner H. A randomized placebo-controlled phase IIb trial of a3309, a bile acid transporter inhibitor, for chronic idiopathic constipation. American Journal of Gastroenterology. 2011;106:1803–1812. doi: 10.1038/ajg.2011.162. [Erratum appears in Am J Gastroenterol 2014 May;109(5):782] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ardelyx: Ardelyx Reports Successful Phase 3 T3MPO-1 Trial of Tenapanor in Patients with IBS-C: Study achieves primary endpoint; tenapanor well-tolerated in treated patients. Edited by; 2017 May 12
- 10.Chey WD, Lembo AJ, Rosenbaum DP. Tenapanor Treatment of Patients With Constipation-Predominant Irritable Bowel Syndrome: A Phase 2, Randomized, Placebo-Controlled Efficacy and Safety Trial. American Journal of Gastroenterology. 2017;112:763–774. doi: 10.1038/ajg.2017.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lacy BE, Moreau JC. Diarrhea-predominant irritable bowel syndrome: Diagnosis, etiology, and new treatment considerations. Journal of the American Association of Nurse Practitioners. 2016;28:393–404. doi: 10.1002/2327-6924.12387. [DOI] [PubMed] [Google Scholar]
- 12.McIntosh K, Reed DE, Schneider T, Dang F, Keshteli AH, De Palma G, Madsen K, Bercik P, Vanner S. FODMAPs alter symptoms and the metabolome of patients with IBS: a randomised controlled trial. Gut. 2016;14:14. doi: 10.1136/gutjnl-2015-311339. [DOI] [PubMed] [Google Scholar]
- 13.Hillila M, Farkkila MA, Sipponen T, Rajala J, Koskenpato J. Does oral alpha-galactosidase relieve irritable bowel symptoms? Scandinavian Journal of Gastroenterology. 2016;51:16–21. doi: 10.3109/00365521.2015.1063156. [DOI] [PubMed] [Google Scholar]
- 14.Camilleri M, Kim D-Y, McKinzie S, Kim H-J, Thomforde G, Burton D, Low PA, Zinsmeister AR. A randomized, controlled exploratory study of clonidine in diarrhea-predominant irritable bowel syndrome. Clinical Gastroenterology & Hepatology. 2003;1:111–121. doi: 10.1053/cgh.2003.50019. [DOI] [PubMed] [Google Scholar]
- 15.Bharucha AE, Fletcher JG, Camilleri M, Edge J, Carlson P, Zinsmeister AR. Effects of clonidine in women with fecal incontinence. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2014;12:843–851 e842. doi: 10.1016/j.cgh.2013.06.035. quiz e844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lembo AJ, Lacy BE, Zuckerman MJ, Schey R, Dove LS, Andrae DA, Davenport JM, McIntyre G, Lopez R, Turner L, et al. Eluxadoline for Irritable Bowel Syndrome with Diarrhea. New England Journal of Medicine. 2016;374:242–253. doi: 10.1056/NEJMoa1505180. [DOI] [PubMed] [Google Scholar]
- 17.Lacy BE, Chey WD, Cash BD, Lembo AJ, Dove LS, Covington PS. Eluxadoline Efficacy in IBS-D Patients Who Report Prior Loperamide Use. American Journal of Gastroenterology. 2017;18:18. doi: 10.1038/ajg.2017.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Administration USFaD. FDA Drug Safety Communication: FDA warns about increased risk of serious pancreatitis with irritable bowel drug Viberzi (eluxadoline) in patients without a gallbladder. Edited by; 2017 March 15. [Google Scholar]
- 19.Pimentel M, Lembo A, Chey WD, Zakko S, Ringel Y, Yu J, Mareya SM, Shaw AL, Bortey E, Forbes WP, et al. Rifaximin therapy for patients with irritable bowel syndrome without constipation. New England Journal of Medicine. 2011;364:22–32. doi: 10.1056/NEJMoa1004409. [DOI] [PubMed] [Google Scholar]
- 20.Aziz I, Tornblom H, Simren M. Small intestinal bacterial overgrowth as a cause for irritable bowel syndrome: guilty or not guilty? Current Opinion in Gastroenterology. 2017;33:196–202. doi: 10.1097/MOG.0000000000000348. [DOI] [PubMed] [Google Scholar]
- 21.Cosmo Technologies Ltd. Rifamycin SV-MMX® 600 mg Tablets Administered Three or Two Times Daily to Patients With IBS-D. Edited by; 2017 March 14. [Google Scholar]
- 22.Ford AC, Brandt LJ, Young C, Chey WD, Foxx-Orenstein AE, Moayyedi P. Efficacy of 5-HT3 antagonists and 5-HT4 agonists in irritable bowel syndrome: systematic review and meta-analysis. American Journal of Gastroenterology. 2009;104:1831–1843. doi: 10.1038/ajg.2009.223. quiz 1844. [DOI] [PubMed] [Google Scholar]
- 23.Chang L, Tong K, Ameen V. Ischemic colitis and complications of constipation associated with the use of alosetron under a risk management plan: clinical characteristics, outcomes, and incidences. American Journal of Gastroenterology. 2010;105:866–875. doi: 10.1038/ajg.2010.25. [DOI] [PubMed] [Google Scholar]
- 24.Fukudo S, Ida M, Akiho H, Nakashima Y, Matsueda K. Effect of ramosetron on stool consistency in male patients with irritable bowel syndrome with diarrhea. Clinical Gastroenterology & Hepatology. 2014;12:953–959.e954. doi: 10.1016/j.cgh.2013.11.024. [DOI] [PubMed] [Google Scholar]
- 25.Fukudo S, Kinoshita Y, Okumura T, Ida M, Akiho H, Nakashima Y, Nishida A, Haruma K. Ramosetron Reduces Symptoms of Irritable Bowel Syndrome With Diarrhea and Improves Quality of Life in Women. Gastroenterology. 2016;150:358–366.e358. doi: 10.1053/j.gastro.2015.10.047. [DOI] [PubMed] [Google Scholar]
- 26.Odunsi-Shiyanbade ST, Camilleri M, McKinzie S, Burton D, Carlson P, Busciglio IA, Lamsam J, Singh R, Zinsmeister AR. Effects of chenodeoxycholate and a bile acid sequestrant, colesevelam, on intestinal transit and bowel function. Clinical Gastroenterology & Hepatology. 2010;8:159–165. doi: 10.1016/j.cgh.2009.10.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Camilleri M, Acosta A, Busciglio I, Boldingh A, Dyer RB, Zinsmeister AR, Lueke A, Gray A, Donato LJ. Effect of colesevelam on faecal bile acids and bowel functions in diarrhoea-predominant irritable bowel syndrome. Alimentary Pharmacology & Therapeutics. 2015;41:438–448. doi: 10.1111/apt.13065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.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;64:84–92. doi: 10.1136/gutjnl-2013-305965. [DOI] [PubMed] [Google Scholar]
- 29.Walters JR, Johnston IM, Nolan JD, Vassie C, Pruzanski ME, Shapiro DA. The response of patients with bile acid diarrhoea to the farnesoid X receptor agonist obeticholic acid. Alimentary Pharmacology & Therapeutics. 2015;41:54–64. doi: 10.1111/apt.12999. [DOI] [PubMed] [Google Scholar]
- 30.Corsetti M, Akyuz F, Tack J. Targeting tachykinin receptors for the treatment of functional gastrointestinal disorders with a focus on irritable bowel syndrome. Neurogastroenterology & Motility. 2015;27:1354–1370. doi: 10.1111/nmo.12616. [DOI] [PubMed] [Google Scholar]
- 31.Tack J, Schumacher K, Tonini G, Scartoni S, Capriati A, Maggi CA, the Iris i The neurokinin-2 receptor antagonist ibodutant improves overall symptoms, abdominal pain and stool pattern in female patients in a phase II study of diarrhoea-predominant IBS. Gut. 2016;15:15. doi: 10.1136/gutjnl-2015-310683. [DOI] [PubMed] [Google Scholar]
- 32.Mitsui K, Niwa T, Kawahara Y, Morimoto N, Ohmoto K, Kato M, Yamaura Y, Yoshimoto N, Suna H, Katsumata S. Anti-stress effects of ONO-2952, a novel translocator protein 18 kDa antagonist, in rats. Neuropharmacology. 2015;99:51–66. doi: 10.1016/j.neuropharm.2015.07.011. [DOI] [PubMed] [Google Scholar]
- 33.Whitehead WE, Duffy K, Sharpe J, Nabata T, Bruce M. Randomised clinical trial: exploratory phase 2 study of ONO-2952 in diarrhoea-predominant irritable bowel syndrome. Alimentary Pharmacology & Therapeutics. 2017;45:14–26. doi: 10.1111/apt.13839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bisschops R, De Ruyter V, Demolin G, Baert D, Moreels T, Pattyn P, Verhelst H, Lepoutre L, Arts J, Caenepeel P, et al. Lanreotide Autogel in the Treatment of Idiopathic Refractory Diarrhea: Results of an Exploratory, Controlled, Before and After, Open-label, Multicenter, Prospective Clinical Trial. Clinical Therapeutics. 2016;38:1902–1911.e1902. doi: 10.1016/j.clinthera.2016.06.012. [DOI] [PubMed] [Google Scholar]
- 35.Wouters MM, Balemans D, Van Wanrooy S, Dooley J, Cibert-Goton V, Alpizar YA, Valdez-Morales EE, Nasser Y, Van Veldhoven PP, Vanbrabant W, et al. Histamine Receptor H1-Mediated Sensitization of TRPV1 Mediates Visceral Hypersensitivity and Symptoms in Patients With Irritable Bowel Syndrome. Gastroenterology. 2016;150:875–887.e879. doi: 10.1053/j.gastro.2015.12.034. [DOI] [PubMed] [Google Scholar]
- 36.Balemans D, Aguilera-Lizarraga J, Florens M, Theofanous S, Perna E, Van Der Merwe S, Wouters MM, Boeckxstaens GE. 434 - Evidence for TRP Channel Sensitization in IBS with Histamine 1 Receptor Antagonism as Effective Treatment. Gastroenterology. 2017;152:S102–S103. [Google Scholar]
- 37.Adam B, Liebregts T, Best J, Bechmann L, Lackner C, Neumann J, Koehler S, Holtmann G. A combination of peppermint oil and caraway oil attenuates the post-inflammatory visceral hyperalgesia in a rat model. Scand J Gastroenterol. 2006;41:155–160. doi: 10.1080/00365520500206442. [DOI] [PubMed] [Google Scholar]
- 38.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;61:560–571. doi: 10.1007/s10620-015-3858-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Augé C, Balz-Hara D, Steinhoff M, Vergnolle N, Cenac N. Protease-activated receptor-4 (PAR 4): a role as inhibitor of visceral pain and hypersensitivity. Neurogastroenterol Motil. 2009;21:1189–e1107. doi: 10.1111/j.1365-2982.2009.01310.x. [DOI] [PubMed] [Google Scholar]
- 40.Cenac N, Andrews CN, Holzhausen M, Chapman K, Cottrell G, Andrade-Gordon P, Steinhoff M, Barbara G, Beck P, Bunnett NW, et al. Role for protease activity in visceral pain in irritable bowel syndrome. J Clin Invest. 2007;117:636–647. doi: 10.1172/JCI29255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rolland-Fourcade C, Denadai-Souza A, Cirillo C, Lopez C, Jaramillo JO, Desormeaux C, Cenac N, Motta JP, Larauche M, Taché Y, et al. Epithelial expression and function of trypsin-3 in irritable bowel syndrome. Gut. 2017 doi: 10.1136/gutjnl-2016-312094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Magen E, Chikovani T. Case report of irritable bowel syndrome responding to omalizumab. Georgian Med News. 2015:42–45. [PubMed] [Google Scholar]
- 43.Magen E, Chikovani T. Possible therapeutic role of IgE blockade in irritable bowel syndrome. World J Gastroenterol. 2016;22:9451–9456. doi: 10.3748/wjg.v22.i43.9451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Pearson JS, Niven RM, Meng J, Atarodi S, Whorwell PJ. Immunoglobulin E in irritable bowel syndrome: another target for treatment? A case report and literature review. Therap Adv Gastroenterol. 2015;8:270–277. doi: 10.1177/1756283X15588875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Vicario M, González-Castro AM, Martínez C, Lobo B, Pigrau M, Guilarte M, de Torres I, Mosquera JL, Fortea M, Sevillano-Aguilera C, et al. Increased humoral immunity in the jejunum of diarrhoea-predominant irritable bowel syndrome associated with clinical manifestations. Gut. 2015;64:1379–1388. doi: 10.1136/gutjnl-2013-306236. [DOI] [PubMed] [Google Scholar]
- 46.Valentin N, Camilleri M, Carlson P, Harrington SC, Eckert D, O’Neill J, Burton D, Chen J, Shaw AL, Acosta A. Potential mechanisms of effects of serum-derived bovine immunoglobulin/protein isolate therapy in patients with diarrhea-predominant irritable bowel syndrome. Physiol Rep. 2017;5 doi: 10.14814/phy2.13170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Detzel CJ, Horgan A, Henderson AL, Petschow BW, Warner CD, Maas KJ, Weaver EM. Bovine immunoglobulin/protein isolate binds pro-inflammatory bacterial compounds and prevents immune activation in an intestinal co-culture model. PLoS One. 2015;10:e0120278. doi: 10.1371/journal.pone.0120278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Cremon C, Stanghellini V, Barbaro MR, Cogliandro RF, Bellacosa L, Santos J, Vicario M, Pigrau M, Alonso Cotoner C, Lobo B, et al. Randomised clinical trial: the analgesic properties of dietary supplementation with palmitoylethanolamide and polydatin in irritable bowel syndrome. Alimentary Pharmacology & Therapeutics. 2017;45:909–922. doi: 10.1111/apt.13958. [DOI] [PubMed] [Google Scholar]
- 49.Fujikawa Y, Tominaga K, Tanaka F, Tanigawa T, Watanabe T, Fujiwara Y, Arakawa T. Enteric glial cells are associated with stress-induced colonic hyper-contraction in maternally separated rats. Neurogastroenterology & Motility. 2015;27:1010–1023. doi: 10.1111/nmo.12577. [Erratum appears in Neurogastroenterol Motil 2016 Feb;28(2):306; [DOI] [PubMed] [Google Scholar]
- 50.Stoffels B, Hupa KJ, Snoek SA, van Bree S, Stein K, Schwandt T, Vilz TO, Lysson M, Veer CV, Kummer MP, et al. Postoperative ileus involves interleukin-1 receptor signaling in enteric glia. Gastroenterology. 2014;146:176–187.e171. doi: 10.1053/j.gastro.2013.09.030. [DOI] [PubMed] [Google Scholar]
- 51.Esposito G, Capoccia E, Turco F, Palumbo I, Lu J, Steardo A, Cuomo R, Sarnelli G, Steardo L. Palmitoylethanolamide improves colon inflammation through an enteric glia/toll like receptor 4-dependent PPAR-alpha activation. Gut. 2014;63:1300–1312. doi: 10.1136/gutjnl-2013-305005. [DOI] [PubMed] [Google Scholar]
- 52.Abbasnezhad A, Amani R, Hajiani E, Alavinejad P, Cheraghian B, Ghadiri A. Effect of vitamin D on gastrointestinal symptoms and health-related quality of life in irritable bowel syndrome patients: a randomized double-blind clinical trial. Neurogastroenterology & Motility. 2016;28:1533–1544. doi: 10.1111/nmo.12851. [DOI] [PubMed] [Google Scholar]
- 53.Steadman CJ, Talley NJ, Phillips SF, Zinsmeister AR. Selective 5-hydroxytryptamine type 3 receptor antagonism with ondansetron as treatment for diarrhea-predominant irritable bowel syndrome: a pilot study. Mayo Clinic Proceedings. 1992;67:732–738. doi: 10.1016/s0025-6196(12)60797-6. [DOI] [PubMed] [Google Scholar]
- 54.von der Ohe MR, Camilleri M, Kvols LK. A 5HT3 antagonist corrects the postprandial colonic hypertonic response in carcinoid diarrhea. Gastroenterology. 1994;106:1184–1189. doi: 10.1016/0016-5085(94)90008-6. [DOI] [PubMed] [Google Scholar]
- 55.Zheng Y, Yu T, Tang Y, Xiong W, Shen X, Jiang L, Lin L. Efficacy and safety of 5-hydroxytryptamine 3 receptor antagonists in irritable bowel syndrome: A systematic review and meta-analysis of randomized controlled trials. PLoS One. 2017;12:e0172846. doi: 10.1371/journal.pone.0172846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zielińska M, Jarmuż A, Wasilewski A, Cami-Kobeci G, Husbands S, Fichna J. Methyl-orvinol-Dual activity opioid receptor ligand inhibits gastrointestinal transit and alleviates abdominal pain in the mouse models mimicking diarrhea-predominant irritable bowel syndrome. Pharmacol Rep. 2017;69:350–357. doi: 10.1016/j.pharep.2016.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cremon C, Stanghellini V, Barbaro MR, Cogliandro RF, Bellacosa L, Santos J, Vicario M, Pigrau M, Alonso Cotoner C, Lobo B, et al. Randomised clinical trial: the analgesic properties of dietary supplementation with palmitoylethanolamide and polydatin in irritable bowel syndrome. Aliment Pharmacol Ther. 2017;45:909–922. doi: 10.1111/apt.13958. [DOI] [PubMed] [Google Scholar]
- 58.Deiteren A, De Man JG, Ruyssers NE, Moreels TG, Pelckmans PA, De Winter BY. Histamine H4 and H1 receptors contribute to postinflammatory visceral hypersensitivity. Gut. 2014;63:1873–1882. doi: 10.1136/gutjnl-2013-305870. [DOI] [PubMed] [Google Scholar]
- 59.Annaházi A, Gecse K, Dabek M, Ait-Belgnaoui A, Rosztóczy A, Róka R, Molnár T, Theodorou V, Wittmann T, Bueno L, et al. Fecal proteases from diarrheic-IBS and ulcerative colitis patients exert opposite effect on visceral sensitivity in mice. Pain. 2009;144:209–217. doi: 10.1016/j.pain.2009.04.017. [DOI] [PubMed] [Google Scholar]
- 60.Zielińska M, Fichna J, Bashashati M, Habibi S, Sibaev A, Timmermans JP, Storr M. G protein-coupled estrogen receptor and estrogen receptor ligands regulate colonic motility and visceral pain. Neurogastroenterol Motil. 2017 doi: 10.1111/nmo.13025. [DOI] [PubMed] [Google Scholar]
