Abstract
Constipation, a condition characterized by heterogeneous symptoms, is common in Western society. It is associated with reduced physical health, mental health, and social functioning. Because constipation is rarely due to a life-threatening disease (for example, colon cancer), current guidelines recommend empiric therapy. Limited surveys suggest that fewer than half of treated individuals are satisfied with treatment, perhaps because the efficacy of drugs is limited, they are associated with undesirable side effects, or they may not target the underlying pathophysiology. For example, although a substantial proportion of constipated patients have a defecatory disorder that is more appropriately treated with pelvic floor biofeedback therapy than with laxatives, virtually no pharmacological trials formally assessed for anorectal dysfunction. Recent advances in investigational tools have improved our understanding of the physiology and pathophysiology of colonic and defecatory functions. In particular, colonic and anorectal high-resolution manometry are now available. High-resolution anorectal manometry, which is increasingly used in clinical practice, at least in the United States, provides a refined assessment of anorectal pressures and may uncover structural abnormalities. Advances in our understanding of colonic molecular physiology have led to the development of new therapeutic agents (such as secretagogues, pro-kinetics, inhibitors of bile acid transporters and ion exchangers). However, because clinical trials compare these newer agents with placebo, their efficacy relative to traditional laxatives is unknown. This article reviews these physiologic, diagnostic, and therapeutic advances and focuses particularly on newer therapeutic agents.
Keywords: Constipation, diagnosis, management, lubiprostone, linaclotide, plecanatide, prucalopride, velusetrag, elobixibat, NGM282, tenapanor
Background
Symptoms of constipation are reported by 10% to 20% of adults worldwide 1. Classically, the term “constipation” refers to infrequent bowel motions or hard feces. However, the disorder is heterogeneous; patients report a variety of symptoms including reduced bowel motion frequency, straining, hard stools, the sensation of incomplete emptying, the sensation of anal blockage, or the use of digitation or positioning to aid defecation ( Box 1) 2. In affected individuals, physical health, mental health, and social functioning are reduced 3, 4. Despite this, only one-fifth of constipated individuals seek health-care advice 5. Yet, given the high prevalence of this condition, this represents over 8 million health-care visits 6 and $230 million in costs 7 annually in the United States.
Box 1. Diagnostic criteria for functional constipation.
Two or more of the following must be present
I. Straining during more than one-fourth (25%) of defecations II. Lumpy or hard stools (Bristol Stool Form Scale 1–2) in more than one-fourth (25%) of defecations III. Sensation of incomplete evacuation in more than one-fourth (25%) of defecations IV. Sensation of anorectal obstruction/blockage in more than one-fourth (25%) of defecations V. Manual maneuvers to facilitate more than one-fourth (25%) of defecations (for example, digital evacuation and support of the pelvic floor) VI. Fewer than three spontaneous bowel movements per week These criteria must be fulfilled for the previous 3 months. The symptom onset must be at least 6 months prior to diagnosis. |
In addition
I. Loose stools are rarely present without the use of laxatives II. There are insufficient criteria for irritable bowel syndrome |
Based upon Mearin et al. 2.
In the absence of alarm features, constipation is seldom due to a life-threatening organic disorder (for example, colon cancer) or another disease (for example, hypothyroidism). Hence, guidelines recommend initial empiric therapy for constipated patients rather than investigating for a cause (for example, with colonoscopy) 8, 9. Likewise, although primary constipation generally results from slow colonic transit, impaired rectal evacuation, or both 8, tests to evaluate these processes are recommended only in patients who do not respond to laxatives.
Slow colonic transit is associated with hard stools 10. Consequently, osmotic and stimulant laxatives are the two most commonly prescribed agents 11, 12. Osmotic laxatives (for example, polyethylene glycol or lactulose) retain water in the intestinal lumen, accelerating colonic transit and reducing the consistency of evacuated stool. Stimulant laxatives (for example, senna, bisacodyl, or glycerin) stimulate colonic contractions and the urge to defecate. Although laxatives increase bowel motion frequency 13, satisfaction is variable 14. In an internet-based survey of 1,355 patients with self-reported constipation in 10 European countries in 2009, 855 patients were taking laxatives. Of these patients, 28% were very satisfied or satisfied with their treatment, 44% were neutral, and 28% were dissatisfied with therapy. In particular, the symptom of bloating persists 15. Conceptually, satisfaction may be suboptimal because the efficacy of drugs is limited, they are associated with undesirable side effects, or they may not target the underlying pathophysiology. In particular, a substantial proportion of constipated patients have a defecatory disorder that may be associated with normal or slow colonic transit. Defecatory disorders are more appropriately treated with pelvic floor biofeedback therapy than with laxatives 16– 18.
Advances in understanding the molecular pathophysiology of constipation
Abnormalities of ion channels within the intestine have been shown to affect secretion, absorption, motility, and sensation, potentially resulting in constipation, diarrhea, and irritable bowel syndrome (IBS) 19. The presence of these dysfunctional channels may be suggested by a family history of a functional bowel disorder 19. Documented “channelopathies” include those affecting the voltage-gated sodium channel Na V1.5, present on smooth muscle cells; the voltage-gated sodium channels Na V1.7 and Na V1.9, present on neurons; and ion exchange channels, present on enterocytes 19. Altered ion channel expression or function occurs because of genetic mutations, post-translational modification, or accessory protein malfunction. In particular, the voltage-gated sodium channel Na V1.5 has been associated with constipation 20. In one study, mutations, predominantly resulting in loss of function, were found in 7% of patients (4 out of 59) with constipation-predominant IBS (IBS-C) 20. Further studies are necessary to determine whether these mutations were responsible for the bowel symptoms.
In selected studies, about one in four patients with diarrhea-predominant IBS has high concentrations of fecal bile acids 21. Conversely, a small fraction (that is, about 6%) of patients with IBS-C have low fecal concentrations of bile acids 22. It is unclear to what extent these findings represent a primary disturbance (that is, due to reduced secretion or increased reabsorption or both) or are secondary to slow colon transit. Nonetheless, because bile acids stimulate colonic secretion of water and high-amplitude-propagated contractions 23, 24, it is hypothesized that a paucity of colonic bile acids may cause constipation. Indeed, the corollary is also true: excess bile acid administered orally 24 or modification of the enterohepatic recycling pathway 25 can exert a laxative effect.
Normal and abnormal colonic and anal structure and function
Defecation is an intricate viscerosomatic process 26, and the pathophysiology of defecatory disorders is heterogeneous. A clinical history is insufficient for differentiating the subtypes of constipation 27. A detailed digital rectal examination 28 may suggest the presence of manometric features of a defecatory disorder—positive predictive value (PPV) of 97% and negative predictive value (NPV) of 37%—but is less useful for predicting an abnormal rectal balloon expulsion test (PPV of 33% and NPV of 65%) 29, 30. Indeed, agreement among anorectal tests is variable, perhaps partly because they assess different aspects of structure or function or both 31, 32. Consequently, the Rome criteria recommend that when standard empiric laxative therapy fails to provide relief for patients with constipation, defecatory disturbances be documented with two tests 32. However, where manometry or defecography is not available, a digital rectal examination and balloon expulsion test are sufficient for screening.
A detailed rectoanal manometry and rectal balloon expulsion test can provide information regarding rectoanal neuromuscular functions 33. Relative to water-perfused manometry, high-resolution manometry is simpler to perform 34. However, in clinical practice, the incremental utility of high-resolution manometry above water-perfused manometry is unclear 33.
In contrast to high-resolution anorectal manometry, high-definition anorectal manometry provides a three-dimensional evaluation of pressures within the anal canal. To date, relatively small studies suggest that high-resolution and high-definition anorectal manometry have similar performance characteristics for evaluating anorectal disorders 35, 36. In high-definition anorectal manometry, similar to high-resolution anorectal manometry, 70% of healthy individuals exhibit a dyssynergic manometry pattern during simulated evacuation 36. More recent studies observed that both high-resolution and high-definition anorectal manometry can identify rectoceles, intra-anal intussusception, and rectal prolapse 37– 40. However, whether these techniques will augment or supplant defecography, or fall by the wayside, for diagnosing these conditions has yet to be determined.
Within the colon, high-resolution manometry is better than standard water-perfused manometry for visualizing propagated colonic contractions 41. While high-resolution colonic manometry remains primarily a research tool, attempts are being made to incorporate these data into clinical practice. For example, among 18 pediatric patients with slow transit constipation and six children with normal transit constipation, manometric findings were predictive of neuropathy with a sensitivity of 100% and a specificity of 86% 42.
Radiological tests are also useful for evaluating anorectal and colonic functions. Barium defecography is used to exclude or diagnose rectoceles and pelvic organ prolapse causing obstructed defecation. Magnetic resonance imaging (MRI) defecography offers a radiation-free alternative. Furthermore, it provides a comprehensive evaluation of pelvic organ structure and function during defecation without the need to instill radiopaque material into the small bowel, bladder, or vagina. With the exception of internal intussusception 43 and retentive rectoceles 44, 45, which are less frequently identified during MRI defecography, barium and MRI defecography have similar performance characteristics.
MRI has been used as a research tool to evaluate colonic motor function in constipation and IBS 46– 48. Inoh et al. evaluated the relationship between colonic diameter and gastrointestinal symptoms in 20 patients with self-reported chronic constipation by using abdominal MRI 49. Ascending colon diameter correlated with a sense of incomplete evacuation, and rectal diameter correlated with constipation scores 49. However, although the sum of all the segmental diameters (cecum, ascending, transverse, descending, sigmoid colon, and rectum) correlated with an increasing severity of constipation, the sum of certain segmental diameters also positively correlated with diarrhea. Furthermore, no statistical adjustment appears to have been made for the multiple comparisons performed in this study. Although the hypothesis is interesting, further studies are required to understand the relationship among colonic fecal volume, colonic diameter, and bowel function. Park et al. demonstrated that rectal gas volume was a marker of defecatory disorders; at a specificity of 90%, a rectal gas volume of 30 mL had a PPV of 77.3% for an evacuation disorder 50, 51.
Development of novel therapeutic agents for the treatment of constipation
A few medications that selectively target intestinal secretion or motility are available. Serotonin receptor agonists have been used to accelerate intestinal transit. Activation or inhibition of intestinal ionic transporters can increase luminal fluid content and accelerates the rate of colonic transit. Inhibition of ileal bile acid transporters exposes the colon to a greater concentration of these ionic detergents, resulting in the secretion of water into the colonic lumen and accelerated colonic transit.
Selective 5-HT(4) receptor agonists: prucalopride and velusetrag
Cisapride and tegaserod, the initial 5-HT(4) receptor agonists used to treat functional bowel disorders 52, 53, were withdrawn from the market because of cardiovascular events 54– 56. By targeting differing pharmacophores, with greater receptor selectivity, novel 5-HT(4) receptor agonists avoid this pro-arrhythmic risk 57, 58.
Prucalopride is a selective, high-affinity, 5-HT(4) receptor agonist with prokinetic gastrointestinal activity 59. Prucalopride accelerated colonic transit in healthy individuals 60, 61 and gastric, small bowel, and colonic transit in constipated patients 62. The initial phase 3, double-blind, parallel-group, placebo-controlled trials demonstrated that prucalopride was substantially more efficacious than placebo for increasing the number of spontaneous complete bowel movements by one per week (47% versus 26%, p <0.001) and promoting more than three complete spontaneous bowel motions (CSBMs) per week (31% versus 12%, p <0.001). Patients reported less-severe symptoms and improved satisfaction with their bowel function 63. Subsequent findings include improved constipation-related quality of life 64, satisfaction with prucalopride in patients who were dissatisfied with previous laxative treatments 65, 66, and efficacy for treating constipation in men 67, elderly patients 68, and patients with chronic intestinal pseudo-obstruction 69, opioid-induced constipation 70, or spinal cord injury 71. Moreover, prucalopride remains efficacious after 18 months of therapy 72. Only one study, a double-blind, placebo-controlled trial over 24 weeks, demonstrated no benefit above placebo 73. Even in older patients, the risk of cardiac events, including QT prolongation, is not increased 74, 75. Only 5% of patients discontinue the medication because of adverse effects (for example, abdominal pain, nausea, diarrhea, or headache) 72. Prucalopride is approved by the European Medicines Agency (EMA), but not by the US Food and Drug Administration (FDA), for the treatment of constipation.
Velusetrag (TD-5108), a newer selective 5-HT(4) receptor agonist, accelerates colonic and gastric transit 76, 77. A phase 2 trial of about 400 patients demonstrated a significant increase above placebo in the number of spontaneous bowel motions (about 3.5 versus 1.4, p <0.001) and CSBMs per week (about 2 versus 0.6, p <0.001) for all doses of velusetrag 78. A phase 2 trial of naronapride (ATI-7505) demonstrated beneficial physiological and clinical effects 79. These studies with velusetrag and ATI-7505 were published almost a decade ago. In November 2016, after a considerable delay, the FDA recommended that efficacy and cardiovascular safety of naronapride be evaluated in two additional phase 3 studies with 1,000 patients each 80. However, no phase 3 trials of velusetrag for constipation are currently registered on ClinicalTrials.gov. Another highly selective 5-HT(4) receptor agonist, YH12852, accelerated upper and lower intestinal transit in animal models 81. Human studies are awaited.
Intestinal chloride channel activators: lubiprostone, linaclotide, and plecanatide
The secretion of ions, and thereby fluid, into the intestinal lumen through ion channels can be pharmacologically driven by lubiprostone, linaclotide, and plecanatide. Activation of the cystic fibrosis transmembrane conductance regulator (CFTR) on the apical surface of enterocytes results in chloride secretion into the intestinal lumen, which is followed by a net secretion of sodium and subsequently water 82.
Lubiprostone is a prostaglandin E analog that activates apical type 2 chloride channels, prostaglandin EP receptors, and the apical CFTR 83. In a 4-week randomized parallel-group placebo-controlled phase 3 trial involving 237 patients with chronic constipation, lubiprostone (24 μg daily) was superior to placebo 84. Lubiprostone-treated patients experienced more frequent spontaneous bowel motions than those treated with placebo (5.9 versus 4.0, p <0.001). Lubiprostone reduced bloating 85 but did not affect pain thresholds during colonic distention 86. It is efficacious for treating constipation associated with cystic fibrosis 83, diabetes 87, and opioids 88. In general, lubiprostone is well tolerated. However, nausea (20%), diarrhea (10%), abdominal distension (7%), headache (7%), and abdominal pain (5%) are reported frequently 85.
Linaclotide and plecanatide are uroguanylin analogs that activate cell-surface guanylate cyclase-C receptors on enterocytes, inducing translocation of the CFTR to the apical surface of the cell. Initial studies of linaclotide demonstrated a dose-dependent increase in colonic transit with an associated increase in bowel motion frequency and consistency and reduced straining scores in patients with IBS-C 89 and chronic constipation 90. Larger studies confirmed these findings, and only 4% of patients stopped the medication because of adverse side effects 91– 93. Linaclotide 145 μg and 290 μg increased the mean number of CSBMs per week to about 2.5 versus 0.9 with placebo ( p <0.001) 92. In rodent models of visceral pain, linaclotide reduced visceral sensitivity 94. Perhaps this explains, at least in part, why linaclotide reduced abdominal pain and improved bowel motion frequency and consistency in patients with IBS-C 95. These benefits appear to persist with longer-term administration 96.
In 2017, plecanatide, which works via mechanisms similar to those of linaclotide, was approved by the FDA for treating chronic idiopathic constipation 97 based on a phase 3, multi-center, double-blind, placebo-controlled study of 1,394 patients 98. Plecanatide increased the weekly number of CSBMs (about 2.2 versus 1.2, p <0.001) and spontaneous bowel motions (about 3.1 versus 1.3, p <0.001) per week above those seen with placebo during a 12-week study. Adverse effects (most commonly diarrhea) occurred in about 6% of patients taking plecanatide and 1% of patients receiving placebo. Similar findings were reported elsewhere 99. An open-label follow-up study of 2,370 patients who had been enrolled in phase 2b or phase 3 studies demonstrated that 82% had completed or were still receiving the study drug 100. These patients reported a median satisfaction score for treatment of 4.0 (quite satisfied) and were “quite likely” to continue the medication. Animal models suggest that plecanatide, similar to linaclotide, also reduces visceral sensitivity 101, and phase 3 trials in patients with IBS-C have demonstrated a significant improvement in bowel motion frequency, stool consistency, and abdominal pain above that seen with placebo 102. Sustained response with the 6 mg dose of plecanatide was seen in 30% of patients in the first study (placebo response 18%, p <0.001) and 24% in the second trial (14% placebo response, p <0.001) 102. Additional CFTR activators (CFTRact-J027 and its derivatives) have demonstrated efficacy for treating constipation in mouse models 103, 104.
Modifiers of bile acid recycling and synthesis: elobixibat and NGM282
Elobixibat (A3309) is the first-in-class ileal bile acid transporter inhibitor 105. Inhibiting the absorption of bile acids from the ileum exposes the colonic mucosa to a higher concentration of these ionic detergents. This accelerates colonic transit, increases stool frequency, and potentially relieves the symptoms of constipation 23, 24, 106. In a randomized phase 2b trial, 190 patients with chronic constipation received 5, 10, or 15 mg of elobixibat or placebo once daily for 8 weeks. The times to the first spontaneous bowel motion and CSBM were significantly shortened in the 10 and 15 mg groups. Stool frequency and constipation-related symptoms were significantly improved 107. A phase 3 trial confirmed the efficacy of elobixibat for the treatment of chronic constipation but demonstrated that adverse drug reactions occur in up to half of patients 108. These are usually mild abdominal pain or diarrhea. Though approved for clinical use in Japan, elobixibat is not currently approved for use by the EMA or the FDA.
In patients with functional constipation, the fibroblast growth factor 19 analog NGM282 accelerated gastric and colonic transit, resulting in an increased number of bowel movements, looser stool form, and increased ease of stool passage 25. The rationale for this study was the observation that NGM282 induced diarrhea in phase 2 trials relating to type 2 diabetes, primary biliary cholangitis, and non-alcoholic steatohepatitis. However, NGM282 is a potent inhibitor of bile acid synthesis, and in this study, in contrast to the effect of elobixibat, bile acid concentration in stool was reduced. Further physiological studies are needed to elucidate how NGM282 exerts its effects.
Sodium/hydrogen exchanger inhibitors: tenapanor
Tenapanor (AZD1722) is a first-in-class, minimally absorbed, small-molecule inhibitor of the gastrointestinal sodium/hydrogen exchanger NHE3. It inhibits the absorption of dietary sodium and phosphate, which increases intestinal fluid volume and transit 109. The effect is more pronounced when tenapanor is administered pre-meal 110. A phase 2 randomized placebo-controlled trial of 356 patients—87% women, mean ± standard deviation (SD) of 46 ± 13 years—with IBS-C demonstrated a CSBM responder rate of 61% with tenapanor 50 mg twice a day as compared with 34% in the placebo group ( p <0.001). Additionally, abdominal pain was significantly reduced in the tenapanor group 111. The most common adverse events were diarrhea, headache, nausea, urinary tract infection, and abdominal pain. Diarrhea occurred in 9% of patients and resulted in medication cessation in 3%. Of note, data regarding the effect of tenapanor on hyperphosphatemia in patients with chronic kidney disease, which encompassed an older patient cohort (mean ± SD of 59 ± 14 years), demonstrated a similar side effect profile 112. Thereafter, two phase 3 trials in IBS-C, each with about 600 patients treated for 12 and 26 weeks, have been reported. In the first phase 3 trial, tenapanor met its primary endpoint (combined pain and stool pattern responder rate of 27% versus 19% with placebo, p = 0.02) 113. Significance was met for the secondary endpoint of abdominal pain relief but not for the CSBM endpoint. In the second study, tenapanor met all primary and secondary endpoints 114; the company plans to submit an application to the FDA in the second half of 2018. Tenapanor is not licensed by the EMA.
New but not necessarily better
Although these new agents are efficacious, only two studies have directly compared the clinical efficacy of newer and older laxatives. The first study compared polyethylene glycol with tegaserod, which has since been withdrawn from the market, in a randomized open-label, parallel-group study of 237 patients. Polyethylene glycol was better for improving symptoms of constipation. In the second, a randomized, double-blind, double-dummy study of 240 patients with chronic constipation, polyethylene glycol with electrolytes was compared with prucalopride 115. Polyethylene glycol with electrolytes was non-inferior to prucalopride for promoting more than three CSBMs per week and was better tolerated.
Indeed, a network meta-analysis comparing prucalopride, lubiprostone, linaclotide, tegaserod, velusetrag, elobixibat, bisacodyl, and sodium picosulfate was undertaken, albeit with limited data for some medications, and observed that these drugs were of comparable efficacy for the endpoints of at least three CSBMs per week or an increase over baseline of at least one CSBM per week. Bisacodyl was superior to the other agents for inducing a greater change from baseline in the number of spontaneous bowel motions per week 116.
Furthermore, the newer laxatives are much more expensive than the older, over-the-counter agents. In the United States, a 30-day supply of lubiprostone, linaclotide, or plecanatide costs about $450. By comparison, a 30-day supply of psyllium, polyethylene glycol, bisacodyl, or senna costs less than $10, whereas lactulose costs less than $15.
Optimizing therapy and the potential role for individualized treatment of constipation
Consensus guidelines recommend that pelvic floor biofeedback therapy, not laxatives, is the cornerstone for managing defecatory disorders 8. However, none of the pharmacological studies described above rigorously evaluated anorectal function and excluded patients with defecatory disorders. Therefore, the relative efficacy of these drugs in patients with isolated normal or slow transit constipation is unknown.
Where constipation does not respond to empiric therapy and a defecatory disorder has been excluded, investigation could guide tailored therapy. For example, the 5-HT(4) receptor agonist prucalopride might be a preferential choice for patients with diffusely slow intestinal transit. Ileal bile acid transporter inhibitors could be of benefit for constipated patients with a deficiency of bile acids reaching the colon. A single case report observed that mexiletine normalized bowel functions in a patient with IBS-C and a mutation in the SCN5A gene, which encodes the alpha-subunit of the voltage-gated sodium channel Na V1.5 20.
Future studies should evaluate the efficacy of novel and standard laxatives in patients with defined normal or slow transit constipation and the efficacy of targeted therapy (for example, with ileal bile acid transporter inhibitors in constipated patients with a deficiency of bile acids reaching the colon).
Summary and conclusions
Constipation is common, but the underlying pathophysiology remains unclear in many cases. Many patients can be effectively and inexpensively treated with simple laxatives. Newer intestinal secretagogues and promotility agents are more expensive and should be considered in patients who do not respond to simple laxatives. A few new agents are being evaluated in clinical trials. There is a critical need to compare the efficacy of these newer agents relative to established laxatives and also clarify their efficacy in the subtypes of constipation (that is, normal transit, slow transit, and defecatory disorders).
Editorial Note on the Review Process
F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).
The referees who approved this article are:
Eamonn M M Quigley, Houston Methodist Hospital, Houston, TX, USA
Massimo Bellini, Gastrointestinal Unit, Department of Gastroenterology, University of Pisa, Pisa, Italy
Funding Statement
This work was supported in part by US Public Health Service National Institutes of Health grant P01 DK68055.
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[version 1; referees: 2 approved]
References
- 1. Mugie SM, Benninga MA, Di Lorenzo C: Epidemiology of constipation in children and adults: a systematic review. Best Pract Res Clin Gastroenterol. 2011;25(1):3–18. 10.1016/j.bpg.2010.12.010 [DOI] [PubMed] [Google Scholar]
- 2. Mearin F, Lacy BE, Chang L, et al. : Bowel Disorders. Gastroenterology. 2016; pii: S0016-5085(16)00222-5. 10.1053/j.gastro.2016.02.031 [DOI] [PubMed] [Google Scholar]
- 3. Belsey J, Greenfield S, Candy D, et al. : Systematic review: impact of constipation on quality of life in adults and children. Aliment Pharmacol Ther. 2010;31(9):938–49. 10.1111/j.1365-2036.2010.04273.x [DOI] [PubMed] [Google Scholar]
- 4. Heidelbaugh JJ, Stelwagon M, Miller SA, et al. : The spectrum of constipation-predominant irritable bowel syndrome and chronic idiopathic constipation: US survey assessing symptoms, care seeking, and disease burden. Am J Gastroenterol. 2015;110(4):580–7. 10.1038/ajg.2015.67 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Drossman DA, Li Z, Andruzzi E, Temple RD, et al. : U.S. householder survey of functional gastrointestinal disorders. Prevalence, sociodemography, and health impact. Dig Dis Sci. 1993;38(9):1569–80. 10.1007/BF01303162 [DOI] [PubMed] [Google Scholar]
- 6. Shah ND, Chitkara DK, Locke GR, et al. : Ambulatory care for constipation in the United States, 1993-2004. Am J Gastroenterol. 2008;103(7):1746–53. 10.1111/j.1572-0241.2008.01910.x [DOI] [PubMed] [Google Scholar]
- 7. Martin BC, Barghout V, Cerulli A: Direct medical costs of constipation in the United States. Manag Care Interface. 2006;19(12):43–9. [PubMed] [Google Scholar]
- 8. Bharucha AE, Pemberton JH, Locke GR, 3rd: American Gastroenterological Association technical review on constipation. Gastroenterology. 2013;144(1):218–38. 10.1053/j.gastro.2012.10.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Wald A, Bharucha AE, Cosman BC, et al. : ACG clinical guideline: management of benign anorectal disorders. Am J Gastroenterol. 2014;109(8):1141–57; (Quiz) 1058. 10.1038/ajg.2014.190 [DOI] [PubMed] [Google Scholar]
- 10. Degen LP, Phillips SF: How well does stool form reflect colonic transit? Gut. 1996;39(1):109–13. 10.1136/gut.39.1.109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Shafe AC, Lee S, Dalrymple JS, et al. : The LUCK study: Laxative Usage in patients with GP-diagnosed Constipation in the UK, within the general population and in pregnancy. An epidemiological study using the General Practice Research Database (GPRD). Therap Adv Gastroenterol. 2011;4(6):343–63. 10.1177/1756283X11417483 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Menees SB, Guentner A, Chey SW, et al. : How Do US Gastroenterologists Use Over-the-Counter and Prescription Medications in Patients With Gastroesophageal Reflux and Chronic Constipation? Am J Gastroenterol. 2015;110(11):1516–25. 10.1038/ajg.2015.156 [DOI] [PubMed] [Google Scholar]
- 13. Paré P, Fedorak RN: Systematic review of stimulant and nonstimulant laxatives for the treatment of functional constipation. Can J Gastroenterol Hepatol. 2014;28(10):549–57. 10.1155/2014/631740 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Müller-Lissner S, Tack J, Feng Y, et al. : Levels of satisfaction with current chronic constipation treatment options in Europe - an internet survey. Aliment Pharmacol Ther. 2013;37(1):137–45. 10.1111/apt.12124 [DOI] [PubMed] [Google Scholar]
- 15. Emmanuel A, Quigley EM, Simrén M, et al. : Factors affecting satisfaction with treatment in European women with chronic constipation: An internet survey. United European Gastroenterol J. 2013;1(5):375–84. 10.1177/2050640613494200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Noelting J, Eaton JE, Choung RS, et al. : The incidence rate and characteristics of clinically diagnosed defecatory disorders in the community. Neurogastroenterol Motil. 2016;28(11):1690–7. 10.1111/nmo.12868 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Rao SS, Mudipalli RS, Stessman M, et al. : Investigation of the utility of colorectal function tests and Rome II criteria in dyssynergic defecation (Anismus). Neurogastroenterol Motil. 2004;16(5):589–96. 10.1111/j.1365-2982.2004.00526.x [DOI] [PubMed] [Google Scholar]
- 18. Chiarioni G, Salandini L, Whitehead WE: Biofeedback benefits only patients with outlet dysfunction, not patients with isolated slow transit constipation. Gastroenterology. 2005;129(1):86–97. 10.1053/j.gastro.2005.05.015 [DOI] [PubMed] [Google Scholar]
- 19. Beyder A, Farrugia G: Ion channelopathies in functional GI disorders. Am J Physiol Gastrointest Liver Physiol. 2016;311(4):G581–G586. 10.1152/ajpgi.00237.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 20. Beyder A, Mazzone A, Strege PR, et al. : Loss-of-function of the voltage-gated sodium channel NaV1.5 (channelopathies) in patients with irritable bowel syndrome. Gastroenterology. 2014;146(7):1659–68. 10.1053/j.gastro.2014.02.054 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 21. Slattery SA, Niaz O, Aziz Q, et al. : Systematic review with meta-analysis: the prevalence of bile acid malabsorption in the irritable bowel syndrome with diarrhoea. Aliment Pharmacol Ther. 2015;42(1):3–11. 10.1111/apt.13227 [DOI] [PubMed] [Google Scholar]
- 22. Vijayvargiya P, Busciglio I, Burton D, et al. : Bile Acid Deficiency in a Subgroup of Patients With Irritable Bowel Syndrome With Constipation Based on Biomarkers in Serum and Fecal Samples. Clin Gastroenterol Hepatol. 2018;16(4):522–7. 10.1016/j.cgh.2017.06.039 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 23. Thaysen EH, Pedersen L: Idiopathic bile acid catharsis. Gut. 1976;17(12):965–70. 10.1136/gut.17.12.965 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Rao AS, Wong BS, Camilleri M, et al. : Chenodeoxycholate in females with irritable bowel syndrome-constipation: a pharmacodynamic and pharmacogenetic analysis. Gastroenterology. 2010;139(5):1549–58, 1558.e1. 10.1053/j.gastro.2010.07.052 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 25. Oduyebo I, Camilleri M, Nelson AD, et al. : Effects of NGM282, an FGF19 variant, on colonic transit and bowel function in functional constipation: a randomized phase 2 trial. Am J Gastroenterol. 2018;113(5):725–34. 10.1038/s41395-018-0042-7 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 26. Bharucha AE: Pelvic floor: anatomy and function. Neurogastroenterol Motil. 2006;18(7):507–19. 10.1111/j.1365-2982.2006.00803.x [DOI] [PubMed] [Google Scholar]
- 27. Ratuapli SK, Bharucha AE, Noelting J, et al. : Phenotypic identification and classification of functional defecatory disorders using high-resolution anorectal manometry. Gastroenterology. 2013;144(2):314–322.e2. 10.1053/j.gastro.2012.10.049 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 28. Rao SSC: Rectal Exam: Yes, it can and should be done in a busy practice! Am J Gastroenterol. 2018;113(5):635–8. 10.1038/s41395-018-0006-y [DOI] [PubMed] [Google Scholar]
- 29. Tantiphlachiva K, Rao P, Attaluri A, et al. : Digital rectal examination is a useful tool for identifying patients with dyssynergia. Clin Gastroenterol Hepatol. 2010;8(11):955–60. 10.1016/j.cgh.2010.06.031 [DOI] [PubMed] [Google Scholar]
- 30. Soh JS, Lee HJ, Jung KW, et al. : The diagnostic value of a digital rectal examination compared with high-resolution anorectal manometry in patients with chronic constipation and fecal incontinence. Am J Gastroenterol. 2015;110(8):1197–204. 10.1038/ajg.2015.153 [DOI] [PubMed] [Google Scholar]
- 31. Carrington EV, Scott SM, Bharucha A, et al. : Expert consensus document: Advances in the evaluation of anorectal function. Nat Rev Gastroenterol Hepatol. 2018;15(5):309–23. 10.1038/nrgastro.2018.27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Rao SS, Bharucha AE, Chiarioni G, et al. : Functional Anorectal Disorders. Gastroenterology. 2016;150(6):1430–1442.e4, pii: S0016-5085(16)00175-X. 10.1053/j.gastro.2016.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Basilisco G, Bharucha AE: High-resolution anorectal manometry: An expensive hobby or worth every penny? Neurogastroenterol Motil. 2017;29(8):e13125. 10.1111/nmo.13125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Kang HR, Lee JE, Lee JS, et al. : Comparison of High-resolution Anorectal Manometry With Water-perfused Anorectal Manometry. J Neurogastroenterol Motil. 2015;21(1):126–32. 10.5056/jnm14025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Chakraborty S, Feuerhak KJ, Zinsmeister AR, et al. : Reproducibility of high-definition (3D) manometry and its agreement with high-resolution (2D) manometry in women with fecal incontinence. Neurogastroenterol Motil. 2017;29(3):e12950. 10.1111/nmo.12950 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Mion F, Garros A, Brochard C, et al. : 3D High-definition anorectal manometry: Values obtained in asymptomatic volunteers, fecal incontinence and chronic constipation. Results of a prospective multicenter study (NOMAD). Neurogastroenterol Motil. 2017;29(8):e13049. 10.1111/nmo.13049 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 37. Heinrich H, Sauter M, Fox M, et al. : Assessment of Obstructive Defecation by High-Resolution Anorectal Manometry Compared With Magnetic Resonance Defecography. Clin Gastroenterol Hepatol. 2015;13(7):1310–1317.e1. 10.1016/j.cgh.2015.01.017 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 38. Prichard DO, Lee T, Parthasarathy G, et al. : High-resolution Anorectal Manometry for Identifying Defecatory Disorders and Rectal Structural Abnormalities in Women. Clin Gastroenterol Hepatol. 2017;15(3):412–20. 10.1016/j.cgh.2016.09.154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Brusciano L, Tolone S, Limongelli P, et al. : Anatomical and Functional Features of the Internal Rectal Prolapse With Outlet Obstruction Determined With 3D Endorectal Ultrasonography and High-Resolution Anorectal Manometry: An Observational Case-Control Study. Am J Gastroenterol. 2018;113(8):1247–50. 10.1038/s41395-018-0141-5 [DOI] [PubMed] [Google Scholar]
- 40. Benezech A, Cappiello M, Baumstarck K, et al. : Rectal intussusception: can high resolution three-dimensional ano-rectal manometry compete with conventional defecography? Neurogastroenterol Motil. 2017;29(4):e12978. 10.1111/nmo.12978 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 41. Corsetti M, Pagliaro G, Demedts I, et al. : Pan-Colonic Pressurizations Associated With Relaxation of the Anal Sphincter in Health and Disease: A New Colonic Motor Pattern Identified Using High-Resolution Manometry. Am J Gastroenterol. 2017;112(3):479–89. 10.1038/ajg.2016.341 [DOI] [PubMed] [Google Scholar]
- 42. Giorgio V, Borrelli O, Smith VV, et al. : High-resolution colonic manometry accurately predicts colonic neuromuscular pathological phenotype in pediatric slow transit constipation. Neurogastroenterol Motil. 2013;25(1):70–8.e8-9. 10.1111/nmo.12016 [DOI] [PubMed] [Google Scholar]
- 43. Pilkington SA, Nugent KP, Brenner J, et al. : Barium proctography vs magnetic resonance proctography for pelvic floor disorders: a comparative study. Colorectal Dis. 2012;14(10):1224–30. 10.1111/j.1463-1318.2012.02945.x [DOI] [PubMed] [Google Scholar]
- 44. van Iersel JJ, Formijne Jonkers HA, Verheijen PM, et al. : Comparison of dynamic magnetic resonance defaecography with rectal contrast and conventional defaecography for posterior pelvic floor compartment prolapse. Colorectal Dis. 2017;19(1):O46–O53. 10.1111/codi.13563 [DOI] [PubMed] [Google Scholar]
- 45. Zafar A, Seretis C, Feretis M, et al. : Comparative study of magnetic resonance defaecography and evacuation proctography in the evaluation of obstructed defaecation. Colorectal Dis. 2017;19(6):O204–O209. 10.1111/codi.13657 [DOI] [PubMed] [Google Scholar]
- 46. Nilsson M, Sandberg TH, Poulsen JL, et al. : Quantification and variability in colonic volume with a novel magnetic resonance imaging method. Neurogastroenterol Motil. 2015;27(12):1755–63. 10.1111/nmo.12673 [DOI] [PubMed] [Google Scholar]
- 47. Sandberg TH, Nilsson M, Poulsen JL, et al. : A novel semi-automatic segmentation method for volumetric assessment of the colon based on magnetic resonance imaging. Abdom Imaging. 2015;40(7):2232–41. 10.1007/s00261-015-0475-z [DOI] [PubMed] [Google Scholar]
- 48. Lam C, Chaddock G, Marciani L, et al. : Colonic response to laxative ingestion as assessed by MRI differs in constipated irritable bowel syndrome compared to functional constipation. Neurogastroenterol Motil. 2016;28(6):861–70. 10.1111/nmo.12784 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Inoh Y, Kanoshima K, Ohkuma K, et al. : Assessment of colonic contents in patients with chronic constipation using MRI. J Clin Biochem Nutr. 2018;62(3):277–80. 10.3164/jcbn.17-104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Park SY, Khemani D, Acosta A, et al. : Rectal gas volume: Defining cut-offs for screening for evacuation disorders in patients with constipation. Neurogastroenterol Motil. 2017;29(7):e13044. 10.1111/nmo.13044 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 51. Park SY, Khemani D, Nelson AD, et al. : Rectal Gas Volume Measured by Computerized Tomography Identifies Evacuation Disorders in Patients With Constipation. Clin Gastroenterol Hepatol. 2017;15(4):543–552.e4. 10.1016/j.cgh.2016.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Quigley EM: Cisapride: what can we learn from the rise and fall of a prokinetic? J Dig Dis. 2011;12(3):147–56. 10.1111/j.1751-2980.2011.00491.x [DOI] [PubMed] [Google Scholar]
- 53. Chey WD: Review article: tegaserod -- the global experience. Aliment Pharmacol Ther. 2004;20 Suppl 7:15–9. 10.1111/j.1365-2036.2004.02181.x [DOI] [PubMed] [Google Scholar]
- 54. Hennessy S, Leonard CE, Newcomb C, et al. : Cisapride and ventricular arrhythmia. Br J Clin Pharmacol. 2008;66(3):375–85. 10.1111/j.1365-2125.2008.03249.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Wysowski DK, Corken A, Gallo-Torres H, et al. : Postmarketing reports of QT prolongation and ventricular arrhythmia in association with cisapride and Food and Drug Administration regulatory actions. Am J Gastroenterol. 2001;96(6):1698–703. 10.1111/j.1572-0241.2001.03927.x [DOI] [PubMed] [Google Scholar]
- 56. Thompson CA: Novartis suspends tegaserod sales at FDA's request. Am J Health Syst Pharm. 2007;64(10):1020. 10.2146/news070044 [DOI] [PubMed] [Google Scholar]
- 57. Potet F, Bouyssou T, Escande D, et al. : Gastrointestinal prokinetic drugs have different affinity for the human cardiac human ether-à-gogo K + channel. J Pharmacol Exp Ther. 2001;299(3):1007–12. [PubMed] [Google Scholar]
- 58. Rampe D, Roy ML, Dennis A, et al. : A mechanism for the proarrhythmic effects of cisapride (Propulsid): high affinity blockade of the human cardiac potassium channel HERG. FEBS Lett. 1997;417(1):28–32. 10.1016/S0014-5793(97)01249-0 [DOI] [PubMed] [Google Scholar]
- 59. Bharucha AE, Wouters MM, Tack J: Existing and emerging therapies for managing constipation and diarrhea. Curr Opin Pharmacol. 2017;37:158–66. 10.1016/j.coph.2017.10.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Bouras EP, Camilleri M, Burton DD, et al. : Selective stimulation of colonic transit by the benzofuran 5HT 4 agonist, prucalopride, in healthy humans. Gut. 1999;44(5):682–6. 10.1136/gut.44.5.682 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Miner PB, Jr, Camilleri M, Burton D, et al. : Prucalopride induces high-amplitude propagating contractions in the colon of patients with chronic constipation: a randomized study. Neurogastroenterol Motil. 2016;28(9):1341–8. 10.1111/nmo.12832 [DOI] [PubMed] [Google Scholar]
- 62. Bouras EP, Camilleri M, Burton DD, et al. : Prucalopride accelerates gastrointestinal and colonic transit in patients with constipation without a rectal evacuation disorder. Gastroenterology. 2001;120(2):354–60. 10.1053/gast.2001.21166 [DOI] [PubMed] [Google Scholar]
- 63. Camilleri M, Kerstens R, Rykx A, et al. : A placebo-controlled trial of prucalopride for severe chronic constipation. N Engl J Med. 2008;358(22):2344–54. 10.1056/NEJMoa0800670 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 64. Quigley EM, Vandeplassche L, Kerstens R, et al. : Clinical trial: the efficacy, impact on quality of life, and safety and tolerability of prucalopride in severe chronic constipation--a 12-week, randomized, double-blind, placebo-controlled study. Aliment Pharmacol Ther. 2009;29(3):315–28. 10.1111/j.1365-2036.2008.03884.x [DOI] [PubMed] [Google Scholar]
- 65. Tack J, van Outryve M, Beyens G, et al. : Prucalopride (Resolor) in the treatment of severe chronic constipation in patients dissatisfied with laxatives. Gut. 2009;58(3):357–65. 10.1136/gut.2008.162404 [DOI] [PubMed] [Google Scholar]
- 66. Tack J, Quigley E, Camilleri M, et al. : Efficacy and safety of oral prucalopride in women with chronic constipation in whom laxatives have failed: an integrated analysis. United European Gastroenterol J. 2013;1(1):48–59. 10.1177/2050640612474651 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Yiannakou Y, Piessevaux H, Bouchoucha M, et al. : A randomized, double-blind, placebo-controlled, phase 3 trial to evaluate the efficacy, safety, and tolerability of prucalopride in men with chronic constipation. Am J Gastroenterol. 2015;110(5):741–8. 10.1038/ajg.2015.115 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 68. Müller-Lissner S, Rykx A, Kerstens R, et al. : A double-blind, placebo-controlled study of prucalopride in elderly patients with chronic constipation. Neurogastroenterol Motil. 2010;22(9):991–8, e255. 10.1111/j.1365-2982.2010.01533.x [DOI] [PubMed] [Google Scholar]
- 69. Emmanuel AV, Kamm MA, Roy AJ, et al. : Randomised clinical trial: the efficacy of prucalopride in patients with chronic intestinal pseudo-obstruction--a double-blind, placebo-controlled, cross-over, multiple n = 1 study. Aliment Pharmacol Ther. 2012;35(1):48–55. 10.1111/j.1365-2036.2011.04907.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Sloots CE, Rykx A, Cools M, et al. : Efficacy and safety of prucalopride in patients with chronic noncancer pain suffering from opioid-induced constipation. Dig Dis Sci. 2010;55(10):2912–21. 10.1007/s10620-010-1229-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Krogh K, Jensen MB, Gandrup P, et al. : Efficacy and tolerability of prucalopride in patients with constipation due to spinal cord injury. Scand J Gastroenterol. 2002;37(4):431–6. 10.1080/003655202317316060 [DOI] [PubMed] [Google Scholar]
- 72. Camilleri M, Van Outryve MJ, Beyens G, et al. : Clinical trial: the efficacy of open-label prucalopride treatment in patients with chronic constipation - follow-up of patients from the pivotal studies. Aliment Pharmacol Ther. 2010;32(9):1113–23. 10.1111/j.1365-2036.2010.04455.x [DOI] [PubMed] [Google Scholar]
- 73. Piessevaux H, Corazziari E, Rey E, et al. : A randomized, double-blind, placebo-controlled trial to evaluate the efficacy, safety, and tolerability of long-term treatment with prucalopride. Neurogastroenterol Motil. 2015;27(6):805–15. 10.1111/nmo.12553 [DOI] [PubMed] [Google Scholar]
- 74. Mendzelevski B, Ausma J, Chanter DO, et al. : Assessment of the cardiac safety of prucalopride in healthy volunteers: a randomized, double-blind, placebo- and positive-controlled thorough QT study. Br J Clin Pharmacol. 2012;73(2):203–9. 10.1111/j.1365-2125.2011.04088.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Camilleri M, Beyens G, Kerstens R, et al. : Safety assessment of prucalopride in elderly patients with constipation: a double-blind, placebo-controlled study. Neurogastroenterol Motil. 2009;21(12):1256–e117. 10.1111/j.1365-2982.2009.01398.x [DOI] [PubMed] [Google Scholar]
- 76. Manini ML, Camilleri M, Goldberg M, et al. : Effects of Velusetrag (TD-5108) on gastrointestinal transit and bowel function in health and pharmacokinetics in health and constipation. Neurogastroenterol Motil. 2010;22(1):42–9, e7–8. 10.1111/j.1365-2982.2009.01378.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Bassotti G, Gambaccini D, Bellini M: Velusetrag for the treatment of chronic constipation. Expert Opin Investig Drugs. 2016;25(8):985–90. 10.1080/13543784.2016.1195369 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 78. Goldberg M, Li YP, Johanson JF, et al. : Clinical trial: the efficacy and tolerability of velusetrag, a selective 5-HT 4 agonist with high intrinsic activity, in chronic idiopathic constipation - a 4-week, randomized, double-blind, placebo-controlled, dose-response study. Aliment Pharmacol Ther. 2010;32(9):1102–12. 10.1111/j.1365-2036.2010.04456.x [DOI] [PubMed] [Google Scholar]
- 79. Camilleri M, Vazquez-Roque MI, Burton D, et al. : Pharmacodynamic effects of a novel prokinetic 5-HT receptor agonist, ATI-7505, in humans. Neurogastroenterol Motil. 2007;19(1):30–8. 10.1111/j.1365-2982.2006.00865.x [DOI] [PubMed] [Google Scholar]
- 80. Renexxion Achieves Positive FDA Guidance for Phase 3-ready GI Drug Naronapride.2016; [cited 2018 7th August]. Reference Source [Google Scholar]
- 81. Jeong EJ, Chung SY, Hong HN, et al. : The novel, potent and highly selective 5-HT 4 receptor agonist YH12852 significantly improves both upper and lower gastrointestinal motility. Br J Pharmacol. 2018;175(3):485–500. 10.1111/bph.14096 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Ahsan MK, Tchernychev B, Kessler MM, et al. : Linaclotide activates guanylate cyclase-C/cGMP/protein kinase-II-dependent trafficking of CFTR in the intestine. Physiol Rep. 2017;5(11): pii: e13299. 10.14814/phy2.13299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Norimatsu Y, Moran AR, MacDonald KD: Lubiprostone activates CFTR, but not ClC-2, via the prostaglandin receptor (EP 4). Biochem Biophys Res Commun. 2012;426(3):374–9. 10.1016/j.bbrc.2012.08.097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Barish CF, Drossman D, Johanson JF, et al. : Efficacy and safety of lubiprostone in patients with chronic constipation. Dig Dis Sci. 2010;55(4):1090–7. 10.1007/s10620-009-1068-x [DOI] [PubMed] [Google Scholar]
- 85. Lembo AJ, Johanson JF, Parkman HP, et al. : Long-term safety and effectiveness of lubiprostone, a chloride channel (ClC-2) activator, in patients with chronic idiopathic constipation. Dig Dis Sci. 2011;56(9):2639–45. 10.1007/s10620-011-1801-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Whitehead WE, Palsson OS, Gangarosa L, et al. : Lubiprostone does not influence visceral pain thresholds in patients with irritable bowel syndrome. Neurogastroenterol Motil. 2011;23(10):944–e400. 10.1111/j.1365-2982.2011.01776.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Christie J, Shroff S, Shahnavaz N, et al. : A Randomized, Double-Blind, Placebo-Controlled Trial to Examine the Effectiveness of Lubiprostone on Constipation Symptoms and Colon Transit Time in Diabetic Patients. Am J Gastroenterol. 2017;112(2):356–64. 10.1038/ajg.2016.531 [DOI] [PubMed] [Google Scholar]
- 88. Webster LR, Brewer RP, Lichtlen P, et al. : Efficacy of Lubiprostone for the Treatment of Opioid-Induced Constipation, Analyzed by Opioid Class. Pain Med. 2018;19(6):1195–205. 10.1093/pm/pnx212 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 89. Andresen V, Camilleri M, Busciglio IA, et al. : Effect of 5 days linaclotide on transit and bowel function in females with constipation-predominant irritable bowel syndrome. Gastroenterology. 2007;133(3):761–8. 10.1053/j.gastro.2007.06.067 [DOI] [PubMed] [Google Scholar]
- 90. Johnston JM, Kurtz CB, Drossman DA, et al. : Pilot study on the effect of linaclotide in patients with chronic constipation. Am J Gastroenterol. 2009;104(1):125–32. 10.1038/ajg.2008.59 [DOI] [PubMed] [Google Scholar]
- 91. Lembo AJ, Kurtz CB, MacDougall JE, et al. : Efficacy of linaclotide for patients with chronic constipation. Gastroenterology. 2010;138(3):886–95.e1. 10.1053/j.gastro.2009.12.050 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 92. Lembo AJ, Schneier HA, Shiff SJ, et al. : Two randomized trials of linaclotide for chronic constipation. N Engl J Med. 2011;365(6):527–36. 10.1056/NEJMoa1010863 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 93. Bassotti G, Usai-Satta P, Bellini M: Linaclotide for the treatment of chronic constipation. Expert Opin Pharmacother. 2018;19(11):1261–6. 10.1080/14656566.2018.1494728 [DOI] [PubMed] [Google Scholar]
- 94. Eutamene H, Bradesi S, Larauche M, et al. : Guanylate cyclase C-mediated antinociceptive effects of linaclotide in rodent models of visceral pain. Neurogastroenterol Motil. 2010;22(3):312–e84. 10.1111/j.1365-2982.2009.01385.x [DOI] [PubMed] [Google Scholar]
- 95. Johnston JM, Kurtz CB, MacDougall JE, et al. : Linaclotide improves abdominal pain and bowel habits in a phase IIb study of patients with irritable bowel syndrome with constipation. Gastroenterology. 2010;139(6):1877–1886.e2. 10.1053/j.gastro.2010.08.041 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 96. 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):1702–12. 10.1038/ajg.2012.254 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 97. Al-Salama ZT, Syed YY: Plecanatide: First Global Approval. Drugs. 2017;77(5):593–8. 10.1007/s40265-017-0718-0 [DOI] [PubMed] [Google Scholar]
- 98. 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):613–21. 10.1038/ajg.2016.611 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 99. DeMicco M, Barrow L, Hickey B, et al. : Randomized clinical trial: efficacy and safety of plecanatide in the treatment of chronic idiopathic constipation. Therap Adv Gastroenterol. 2017;10(11):837–51. 10.1177/1756283X17734697 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Barish CF, Griffin P: Safety and tolerability of plecanatide in patients with chronic idiopathic constipation: long-term evidence from an open-label study. Curr Med Res Opin. 2018;34(4):751–5. 10.1080/03007995.2018.1430024 [DOI] [PubMed] [Google Scholar]
- 101. Boulete IM, Thadi A, Beaufrand C, et al. : Oral treatment with plecanatide or dolcanatide attenuates visceral hypersensitivity via activation of guanylate cyclase-C in rat models. World J Gastroenterol. 2018;24(17):1888–900. 10.3748/wjg.v24.i17.1888 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Brenner DM, Fogel R, Dorn SD, et al. : Efficacy, safety, and tolerability of plecanatide in patients with irritable bowel syndrome with constipation: results of two phase 3 randomized clinical trials. Am J Gastroenterol. 2018;113(5):735–45. 10.1038/s41395-018-0026-7 [DOI] [PubMed] [Google Scholar]
- 103. Cil O, Phuan PW, Lee S, et al. : CFTR activator increases intestinal fluid secretion and normalizes stool output in a mouse model of constipation. Cell Mol Gastroenterol Hepatol. 2016;2(3):317–27. 10.1016/j.jcmgh.2015.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Son JH, Zhu JS, Phuan PW, et al. : High-Potency Phenylquinoxalinone Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Activators. J Med Chem. 2017;60(6):2401–10. 10.1021/acs.jmedchem.6b01759 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Wong BS, Camilleri M: Elobixibat for the treatment of constipation. Expert Opin Investig Drugs. 2013;22(2):277–84. 10.1517/13543784.2013.753056 [DOI] [PubMed] [Google Scholar]
- 106. Rudling M, Camilleri M, Graffner H, et al. : Specific inhibition of bile acid transport alters plasma lipids and GLP-1. BMC Cardiovasc Disord. 2015;15:75. 10.1186/s12872-015-0070-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Chey WD, Camilleri M, Chang L, et al. : A randomized placebo-controlled phase IIb trial of a3309, a bile acid transporter inhibitor, for chronic idiopathic constipation. Am J Gastroenterol. 2011;106(10):1803–12. 10.1038/ajg.2011.162 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 108. Nakajima A, Seki M, Taniguchi S, et al. : Safety and efficacy of elobixibat for chronic constipation: results from a randomised, double-blind, placebo-controlled, phase 3 trial and an open-label, single-arm, phase 3 trial. Lancet Gastroenterol Hepatol. 2018;3(8):537–47. 10.1016/S2468-1253(18)30123-7 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 109. Rosenbaum DP, Yan A, Jacobs JW: Pharmacodynamics, Safety, and Tolerability of the NHE3 Inhibitor Tenapanor: Two Trials in Healthy Volunteers. Clin Drug Investig. 2018;38(4):341–51. 10.1007/s40261-017-0614-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Johansson SA, Knutsson M, Leonsson-Zachrisson M, et al. : Effect of Food Intake on the Pharmacodynamics of Tenapanor: A Phase 1 Study. Clin Pharmacol Drug Dev. 2017;6(5):457–65. 10.1002/cpdd.341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. 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. Am J Gastroenterol. 2017;112(5):763–74. 10.1038/ajg.2017.41 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 112. Block GA, Rosenbaum DP, Leonsson-Zachrisson M, et al. : Effect of Tenapanor on Serum Phosphate in Patients Receiving Hemodialysis. J Am Soc Nephrol. 2017;28(6):1933–42. 10.1681/ASN.2016080855 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Ardelyx Reports Successful Phase 3 T3MPO-1 Trial of Tenapanor in Patients with IBS-C.2017; [cited 2018 18th July]. Reference Source [Google Scholar]
- 114. Ardelyx's Pivotal Phase 3 Study of Tenapanor for IBS-C Hits Primary and All Secondary Endpoints to Support NDA Submission in 2018.2017; [cited 2018 18th July]. Reference Source [Google Scholar]
- 115. Cinca R, Chera D, Gruss HJ, et al. : Randomised clinical trial: macrogol/PEG 3350+electrolytes versus prucalopride in the treatment of chronic constipation -- a comparison in a controlled environment. Aliment Pharmacol Ther. 2013;37(9):876–86. 10.1111/apt.12278 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 116. Nelson AD, Camilleri M, Chirapongsathorn S, et al. : Comparison of efficacy of pharmacological treatments for chronic idiopathic constipation: a systematic review and network meta-analysis. Gut. 2017;66(9):1611–22. 10.1136/gutjnl-2016-311835 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation