Abstract
Chronic constipation is a very common functional gastrointestinal disorder which can be associated with significant impairments in quality of life for some people with the condition. Its management has, traditionally, been based on dietary and lifestyle changes and the use of a variety of laxative agents. The evidence base for the efficacy of the latter is, in many cases, slim. Not surprisingly, many patients remain dissatisfied with laxatives thus leading to the development of more pharmacological approaches. Among these approaches is the use of prokinetic agents; while prior molecules have been troubled by lack of selectivity and cardiac side effects, the new agent, prucalopride, appears to be highly selective for the serotonin 5-HT4 receptor and is, therefore, a potent stimulator of gut motility. In three large pivotal randomized controlled trials, prucalopride has been effective in relieving the cardinal symptoms of chronic constipation; these effects have been sustained in open-label follow up for as long as 18 months. The safety profile has been encouraging and, especially so, the absence of arrhythmogenic potential. Studies in men, in constipation-predominant irritable bowel syndrome and in other motor disorders are eagerly awaited.
Keywords: constipation, irritable bowel syndrome, prucalopride, prokinetic, colonic motilty
The clinical context
Chronic constipation
Chronic constipation (CC) is certainly common, with prevalence rates of up to 28% being reported in the USA [Sonnenberg and Koch, 1989]. Constipation is at least twice as common in women as in men and its occurrence increases with advancing age, particularly after age 65 [Drossman et al. 1993; Talley et al. 1992; Sonnenberg and Koch, 1989]. While constipation may be associated with, or caused by, many underlying disease entities and a long list of pharmacological agents, we will confine our discussion to those in whom there is no obvious or detectable primary cause for their constipation: functional constipation. This entity is also known as chronic idiopathic constipation and is often referred to in the literature as simply CC. Nowadays, the selection process for the inclusion or exclusion of people for entry into a clinical trial involving CC is usually dictated by the Rome criteria, the most recent iteration of which, Rome III, defined functional constipation as a symptom complex which must include at least two of the following: straining*, lumpy or hard stools*, sensation of incomplete evacuation*, sensation of anorectal obstruction/blockage*, manual maneuvers to facilitate defecation (e.g. digital evacuation, support of the pelvic floor)*, less than three defecations/week. Furthermore, each of the symptoms denoted by an asterisk must occur in relation to at least 25% of defecations. In addition, loose stool should rarely be present without the use of laxatives and there must be insufficient criteria for the diagnosis of constipation-predominant irritable bowel syndrome (IBS-C) [Longstreth et al. 2006].
Chronic idiopathic constipation is traditionally divided into two broad categories, slow-transit constipation (colonic inertia) and ‘outlet-type’ constipation, also referred to as defecatory dysfunction or anismus [Brandt et al. 2005]. In terms of symptom associations, the former would, in theory, manifest as infrequent stools, the latter as some difficulty associated with the act of defecation (straining, sense of incomplete evacuation, rectal fullness). While this distinction is attractive from the pathophysiological point of view, such a clear separation is often difficult, if not impossible, in clinical practice. Indeed, it is commonplace to find patients complain of symptomatology suggestive of both disorders, that is, infrequent stool frequency combined with straining and/or a sensation of incomplete evacuation, for example. These distinctions have important therapeutic implications given that prokinetic agents, such as the compound that is the focus of this review, would be expected to have their greatest impact among those with delayed transit and that colectomy, the most drastic intervention that one may contemplate in the management of constipation, should be considered only among those with severe refractory colonic inertia. As a corollary, colectomy should not be contemplated in any patient where symptoms and/or evaluation suggest the presence of pelvic floor or other ‘outlet’ problems. Because of the limitations of symptoms in predicting underlying pathology, and the aforementioned overlap, in a given patient, between the two broad categories of constipation, the clinical reality is that many patients, including those entered into clinical trials, will exhibit features of both delayed colon transit and defecatory dysfunction; a factor that must be borne in mind in interpreting the results of any therapeutic intervention. It should come as no surprise, therefore, that constipation represents a significant therapeutic challenge!
Irritable bowel syndrome
Irritable bowel syndrome (IBS) is one of the most common disorders encountered in modern medicine; community surveys in Western Europe and North America suggesting a prevalence of around 10% in the adult population [Brandt et al. 2009; Quigley et al. 2007]. It should be stressed, in addition, that IBS appears to be common worldwide regardless of geography or socioeconomic status. There is no single specific diagnostic test for IBS; its definition relies, therefore, either on the exclusion of diseases that may share its symptomatology in whole or in part, or on the application of symptom-based criteria the integrity of which has been validated in cross-sectional and longitudinal studies. The cardinal symptoms of IBS are abdominal pain/discomfort and bowel dysfunction; typically, these are interrelated such that, for example, an affected patient may report that his or, more likely, her symptoms worsen when constipated, only to be relieved once a bowel movement has been achieved. In clinical research, most studies apply the definitions enshrined in the Rome criteria, whose third iteration (Rome III) was released in early 2006 and defined IBS as:
Recurrent abdominal pain or discomfort (an uncomfortable sensation not described as pain) at least 3 days per month in the last 3 months associated with 2 or more of the following:
improvement with defecation,
onset associated with a change in frequency of stool,
onset associated with a change in form (appearance) of stool.
These criteria should have been fulfilled for the last 3 months with symptom onset at least 6 months prior to diagnosis [Longstreth et al. 2006].
IBS is further subtyped based on predominant bowel habit at the time of presentation as:
IBS-D, where diarrhea is the predominant bowel habit,
IBS-C, where constipation predominates; hard or lumpy stools (scored as 1 or 2 on the Bristol Stool Form Scale) ≥25% of defecations and, in conjunction, loose or watery stools (scored as 6 or 7 on the Bristol Stool Form Scale <25% of defecations,
IBS-M (mixed), where both diarrhea and constipation occur, and,
IBS-U (unclassified), where the subject does not fit into one of the above categories.
Where does chronic constipation end and irritable bowel syndrome begin?
Currently, IBS lacks an objective test or biomarker to confirm or refute the diagnosis, monitor progress or evaluate response to treatment; this remains a major obstacle to progress. As individual IBS symptoms are very nonspecific and may occur in a host of other clinical conditions, the potential for diagnostic confusion is considerable. Even when taken collectively, as in the Rome criteria, the potential for diagnostic overlap persists unless the criteria become overly restrictive and this is of special relevance to the potential for overlap between IBS-C and CC. Where, indeed, does IBS-C end and CC begin? Right now this is a value judgment and is based on how much pain and discomfort one is prepared to accept in a patient with constipation. It will be the experience of every clinician that many patients with constipation complain of bloating, distension and abdominal discomfort; cardinal symptoms of IBS. It is the opinion of this author that it is only the degree or prominence of these symptoms that differentiates IBS-C from CC, a differentiation that often seems arbitrary, if not impossible, in clinical practice. It should come as no surprise that many of the therapeutic classes that demonstrate efficacy in CC are then evaluated in IBS-C. However, given the primacy of pain in IBS and slow colonic transit in CC, it stands to reason that results with a new therapeutic agent or class in one may not translate into similar efficacy in the other.
Motility in constipation and irritable bowel syndrome
While defecatory dysfunction is thought to be due to difficulties at the level of the pelvic floor and, thus, is not a colonic motor disorder per se, slow-transit constipation is presumed to reflect a primary disorder of colonic motility. In support of this presumption, these patients who, by definition, have slow colonic transit, have been shown to demonstrate a variety of motor abnormalities on manometric studies of the colon. Foremost among these and the colonic function that has been most widely adopted for use in clinical trials, is slow transit through the entire colon or segments of the colon.
In clinical practice, colon transit is most usually measured using the radio-opaque marker technique [Lin et al. 2005]. This approach has been shown to provide an accurate and reproducible assessment of overall colonic transit and by examining the distribution of retained markers may give some hints as to whether the underlying problem is colonic inertia or defaecatory dysfunction. To plan appropriate treatment strategies, however, further testing may be required.
More accurate and dynamic assessments of colon transit, including the determination of transit within various segments of the colon can be obtained from radio-isotopic approaches. Though these methodologies have been largely confined to a few centers, they have been widely used in the initial assessment of potentially colo-kinetic drugs in clinical research protocols [Odunsi and Camilleri, 2009; von der Ohe and Camilleri, 1992].
More direct assessments of colonic motor function can be obtained by manometry. However, colonic manometry presents formidable challenges, foremost among these being that of positioning the catheter assembly in the first place and ensuring that it retains its position throughout the period of study. Furthermore, patterns of colonic motility are poorly defined and subject to tremendous variation between normal people, not to mention disease states. While the absence of the most recognizable pressure wave pattern, the high amplitude power contraction (HAPC), during a recording period of adequate duration, or following exposure to adequate stimulation, has been proposed as being of diagnostic value among children and both a reduced frequency and amplitude of HAPCs and an impaired colonic motor response to food and exercise have been reported among adults with CC, there is at present no consensus with regard to the utility of colonic manometry in clinical practice in the adult patient [Camilleri et al 2008a, 1998] and the role of this modality in drug development has been limited.
Conditions such as CC and IBS, whose definitions rest exclusively on the interpretation of symptoms, are likely to encompass a heterogeneous population whose constituents may ultimately be found to have different causes. Not surprisingly, the search for a unifying hypothesis to explain all CC or IBS has proven unfruitful. Several phenomena undoubtedly contribute to symptom genesis, including disordered bowel motility (‘spasm’), increased bowel sensitivity (visceral hypersensitivity or hyperalgesia in IBS, hyposensitivity in some with CC), altered cerebral processing of gut events, environmental stressors and intrinsic psychopathology [Quigley, 2003].
While a variety of abnormal electromyographic and motor patterns have been described in the various parts of the gastrointestinal tract in IBS, the specificity of any of these for IBS remains unclear [McKee and Quigley, 1993] and interest has shifted to the role of colorectal sensation as well as dysfunction along the brain–gut axis in the pathogenesis of symptoms in IBS [Bouin et al. 2002].
Prucalopride for constipation
Prucalopride is a highly selective serotonin 5-HT4 receptor agonist which has been shown to stimulate gut motility in vitro and in vivo [Sanger and Quigley, 2010]. In healthy volunteers, scintigraphic studies demonstrated that prucalopride accelerated whole gut and colonic transit but not gastric emptying or small bowel transit [Bouras et al. 1999]. Among patients with constipation, however, the very same authors using the exact same scintigraphic technique found that prucalopride in doses of 2 or 4 mg daily accelerated whole gut, gastric, small bowel and colonic transit in constipated patients [Bouras et al. 2001]. Importantly, prucalopride did not appear to adversely affect a number of parameters of anorectal motor function or impair rectal sensation in either healthy volunteers or patients with constipation [Sloots et al. 2002; Bouras et al. 2001; Poen et al. 1999; Emmanuel et al. 1998]. In studies of colonic motility, prucalopride has been shown to be stimulatory [De Schryer et al. 2002; Emmanule et al. 2002].
Given the profile of pharmacological effects described for prucalopride in vivo, it should come as no surprise that the major clinical focus of prucalopride has been in constipation [Emmanuel et al. 2002; Camilleri et al. 2008b; Quigley et al. 2009; Tack et al. 2009]. Indeed, each of the three major studies of prucalopride in humans has been in CC [Tack et al. 2009; Quigley et al. 2009; Camilleri et al. 2008b] (Table 1). For reasons related to the transfer of the drug from one company to another, these trials have only been published recently, even though they were actually designed and completed some time ago. These three trials, which became pivotal in terms of regulatory submission, featured a randomized, placebo-controlled, parallel group design. The major inclusion criterion was the presence of CC, defined as two or fewer spontaneous complete bowel movements (SCBMs) per week for at least 6 months prior to screening plus any one of the following: hard/very hard stools, a sensation of incomplete evacuation, or straining, during defecation in relation to at least 25% of bowel movements. After a 2-week baseline period, eligible patients were randomized to either a placebo, or 2 mg or 4 mg of prucalopride for 12 weeks. The primary endpoint, in each study, was the proportion of patients passing at least three SCBMs per week during the 12 weeks of the trial, based on an intention-to-treat analysis. All three trials (which assessed 620, 641, and 713 patients, respectively) demonstrated a significant increase in the proportion of patients achieving at least three SCBMs per week compared with placebo. Response rates ranged from 19.5% to 31% with 2 mg prucalopride, 24% to 28% with 4 mg prucalopride, and 9.6% to 12% with placebo. Clinically relevant and statistically significant improvements were also demonstrated in a number of secondary endpoints, including satisfaction with bowel function, perception of constipation severity, and patient-assessed symptom scores. A validated, disease-specific quality of life instrument (Patient Assessment of Constipation-Quality of Life – PAC-QOL) [Dubois et al. 2010], but not a generic quality of life instrument (Short Form 36 – SF-36), showed significant improvements with prucalopride [Tack et al. 2009; Quigley et al. 2009; Camilleri et al. 2008b]. Even though men and women were enrolled in these studies, over 85% of evaluated patients were female. While this led to the approval of the drug being restricted to women, it needs to be stressed that this should not be taken to imply prucalopride is not effective in men but, rather, that it has not been adequately tested in this gender. Though two doses, 2 mg once daily and 4 mg once daily, were tested in these studies, a dose–response effect was not obvious, thus the recommended dose is 2 mg.
Table 1.
Reference | Study design (No. of patients) | Outcomes (2 mg and 4 mg prucalopride versus placebo) |
---|---|---|
[Camilleri et al. 2008b] | 12 weeks (620) | No. of patients achieving |
Prucalopride 2 mg or 4 mg dailyversus placebo |
|
|
[Quigley et al. 2009] | 12 weeks (641) | No. of patients achieving: |
Prucalopride 2 mg or 4 mgdaily versus placebo |
|
|
[Tack et al. 2009] | 12 weeks (713) | No. of patients achieving: |
Prucalopride 2 mg or 4 mgdaily versus placebo |
|
SCBM, spontaneous complete bowel movement.
CC is not a short-term problem; long-term results are critically important for such a chronic problem. Experience over a longer term is based on open-label follow up, for up to 24 months, of 86% of those patients who had completed the three randomized, double-blind pivotal studies described above [Camilleri et al. 2010]. Improvement in the PAC-QOL observed in the double-blind phase was maintained during open-label treatment for up to 18 months.
These benefits were supported in smaller studies of CC [Emmanuel et al. 2002; Sloots et al. 2002] as well as in studies of special populations: elderly people [Müller-Lissner et al. 2010], those with severe, refractory constipation [Coremans et al. 2003], opioid-induced constipation [Sloots et al. 2010], and constipation related to spinal cord injury [Krogh et al. 2002].
Prucalopride in other motility disorders
To date, there are very limited data on the impact of prucalopride on motility disorders affecting other parts of the gastrointestinal tract [Boecxstaens et al. 2002] and it is certainly too early to assess its potential usefulness in disorders such as gastroparesis, intestinal pseudo-obstruction, functional dyspepsia and, most importantly, IBS-C.
Safety
Given the cardiac adverse event history that led to the withdrawal of the less selective serotonergic agonists, cisapride [Quigley, 2011] and tegaserod [Chan et al. 2009], considerable attention has been paid to the safety profile of prucalopride and to its cardiac toxicity, in particular. In the pivotal studies, which collectively represent the largest experience with the drug, the most common treatment-associated adverse events were headache (25–30% prucalopride; 12–17% placebo), nausea (12–24%; 8–14%), abdominal pain or cramps (16–23%; 11–19%) and diarrhea (12–19%; 3–5%) [Tack et al. 2009; Quigley et al. 2009; Camilleri et al. 2008b]. A majority of these adverse events occurred within the first 24 h of treatment and were short lived [Quigley et al. 2009]. Where reported, the prevalence of serious adverse events was similar for placebo and prucalopride [Tack et al. 2009; Quigley et al. 2009; Camilleri et al. 2008b].
Of critical importance, given the experience with cisapride, is that prucalopride has not been found to interact with either the hERG potassium channel (relevant to cisapride-induced arrhythmias) or 5HT1B receptors (where tegaserod may have affinity) [Potet et al. 2001] – both postulated to be responsible for the development of adverse cardiovascular effects with other 5HT4 agonists. Furthermore, no significant hemodynamic or clinically relevant electrocardiographic changes were detected in healthy control studies [Boyce et al. 2009], nor were they detected in any of the major clinical trials [Tack et al 2009, 2008; Quigley et al. 2009; Camilleri et al. 2008b]. In a smaller study conducted in a relatively high-risk population, 89 elderly nursing home residents of whom 80% had a prior history of cardiovascular disease, no significant hemodynamic or electrocardiographic changes were detected. Specifically, there was no evidence of an increase in the incidence of prolongation of the QTc interval [Camilleri et al. 2009].
What is the place of prucalopride in the management of constipation and other gastrointestinal disorders?
To date, high-quality evidence for prucalopride exists only in the area of CC and, then, only for women. Given the aforementioned clinical overlap between CC and IBS, studies in IBS-C are eagerly awaited, as are studies in motor disorders of the foregut and midgut. Efficacy in these latter areas would be of relevance to patients with CC given the frequency with which symptoms such as heartburn, dyspepsia and postprandial fullness occur in these patients.
Currently, prucalopride is approved in a number of countries in Western Europe for use in the management of constipation in women with CC in whom laxatives fail to provide adequate relief; approval is, no doubt, being sought throughout the world. Given the limited number of men included in the pivotal studies, the restriction of approval to women makes sense pending positive data for men. The restriction to laxative failures is also very much in keeping with a number of authoritative guidelines which suggest a step-up approach to the treatment of constipation: beginning with dietary and lifestyle changes, progressing through osmotic laxatives (such as polyethylene glycol derivatives) and reserving pharmacological therapies for people whose condition fails to respond to these steps [Lindberg et al. 2011; Tack et al. 2011; Emmanuel et al. 2009; Brandt et al. 2005]. It must be stressed that there are very few studies comparing the efficacy of a new pharmacological agent with a traditional laxative; for this reason, the real therapeutic gain attributable to prucalopride over and above a laxative remains somewhat conjectural. The one study that compared a new agent, tegaserod, with a laxative, polyethylene glycol, found the latter to be more effective; a salutary lesson [Di Palma et al. 2007].
As a prescription drug for the management of constipation and given the virtual demise of other prokinetic agents for this indication, prucalopride competes primarily with another class of agents: those that stimulate secretion. Two drugs in this class have been extensively studied: the type-2 chloride channel stimulant, lubiprostone and the guanylate cyclase C agonist, linaclotide. Both have demonstrated efficacy in CC and C-IBS [Johnston et al. 2010; Lembo et al. 2010; Drossman et al. 2009; Johanson et al. 2008a, 2008b]. Interestingly, linaclotide has shown an effect against abdominal pain in IBS-C, an effect that seems independent of laxation and is postulated to represent inhibitory activity on sensory afferent nerves [Johnston et al. 2010]. Both drugs are well tolerated, though nausea has been somewhat common with lubiprostone [Drossman et al. 2009; Johanson et al. 2008a, 2008b]. There have been no comparisons with prucalopride.
Conclusions
Prucalopride is a highly selective serotonin 5-HT4 receptor agonist which acts as a prokinetic in the gut. Data from large randomized, controlled clinical trials indicate that it is effective in CC and offers a new therapeutic alternative for those whose condition fails to respond to conventional laxatives. To date, cardiac toxicity, which bedeviled other drugs in this class, has not been a major issue. Long-term studies and postmarketing data will be critical in assessing the real benefits and risks of this promising compound.
Footnotes
Eamonn Quigley has served on the advisory board of Shire and Movetis.
This research received no specific grant from any funding agency in the public, commercial, or not for-profit sectors.
References
- Boeckxstaens G.E., Bartelsman J.F., Lauwers L., Tytgat G.N. (2002) Treatment of GI dysmotility in scleroderma with the new enterokinetic agent prucalopride. Am J Gastroenterol 97: 194–197 [DOI] [PubMed] [Google Scholar]
- Bouin M., Plourde V., Boivin M., Riberdy M., Lupien F., Laganière M., et al. (2002) Rectal distension testing in patients with the irritable bowel syndrome: sensitivity, specificity, and predictive values of pain sensory thresholds. Gastroenterology 122: 1771–1777 [DOI] [PubMed] [Google Scholar]
- Bouras E.P., Camilleri M., Burton D.D., McKinzie S. (1999) Selective stimulation of colonic transit by the benzofuran 5HT4 agonist, prucalopride, in healthy humans. Gut 44: 682–686 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bouras E.P., Camilleri M., Burton D.D., Thomforde G., McKinzie S., Zinsmeister A.R. (2001) Prucalopride accelerates gastrointestinal and colonic transit in patients with constipation without a rectal evacuation disorder. Gastroenterology 120: 354–360 [DOI] [PubMed] [Google Scholar]
- Boyce M., Kerstens R., Beyens G., Ausma J., Vandeplassche L. (2009) Cardiovascular safety of prucalopride in healthy subjects: results from two randomized, double-blind, placebo-controlled, cross-over trials [abstract]. Gastroenterology 136(Suppl. 1): T1265 [Google Scholar]
- Brandt L.J., Chey W.D., Schiller L.R., Schoenfeld P.S., Spiegel B.M., Talley N.J., et al. (2009) An evidence-based systematic review of the management of irritable bowel syndrome. Am J Gastroenterol 104: S1–S35 [DOI] [PubMed] [Google Scholar]
- Brandt L.J., Schoenfeld P., Prather C.M., Quigley E.M.M., Schiller L.R., Talley N.J. (2005) Evidence-based position statement on the management of chronic constipation in North America. Am J Gastroenterol 100(Suppl. 1): S5–S21 [DOI] [PubMed] [Google Scholar]
- Camilleri M., Beyens G., Kerstens R., Robinson P., Vandeplassche L. (2009) Safety assessment of prucalopride in elderly patients with constipation: a double-blind, placebo-controlled study. Neurogastroenterol Motil 21: e1256–e117 [DOI] [PubMed] [Google Scholar]
- Camilleri M., Bharucha A.E., di Lorenzo C., Hasler W.L., Prather C.M., Rao S.S., et al. (2008a) American Neurogastroenterology and Motility Society consensus statement on intraluminal measurement of gastrointestinal and colonic motility in clinical practice. Neurogastroenterol Motil 20: 1269–1282 [DOI] [PubMed] [Google Scholar]
- Camilleri M., Hasler W.L., Parkman H.P., Quigley E.M., Soffer E. (1998) Measurement of gastrointestinal motility in the GI laboratory. Gastroenterology 115: 747–762 [DOI] [PubMed] [Google Scholar]
- Camilleri M., Kerstens R., Rykx A., Vandeplassche L. (2008b) A placebo-controlled trial of prucalopride for severe chronic constipation. N Engl J Med 358: 2344–2354 [DOI] [PubMed] [Google Scholar]
- Camilleri M., Van Outryve M.J., Beyens G., Kerstens R., Robinson P., Vandeplassche L. (2010) 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 32: 1113–1123 [DOI] [PubMed] [Google Scholar]
- Chan K.Y., de Vries R., Leijten F.P., Pfannkuche H.J., van den Bogaerdt A.J., Danser A.H., et al. (2009) Functional characterization of contractions to tegaserod in human isolated proximal and distal coronary arteries. Eur J Pharmacol 619: 61–67 [DOI] [PubMed] [Google Scholar]
- Coremans G., Kerstens R., De Pauw M., Stevens M. (2003) Prucalopride is effective in patients with severe chronic constipation in whom laxatives fail to provide adequate relief. Results of a double-blind, placebo-controlled clinical trial. Digestion 67: 82–89 [DOI] [PubMed] [Google Scholar]
- De Schryver A.M., Andriesse G.I., Samsom M., Smout A.J., Gooszen H.G., Akkermans L.M. (2002) The effects of the specific 5HT(4) receptor agonist, prucalopride, on colonic motility in healthy volunteers. Aliment Pharmacol Ther 16: 603–612 [DOI] [PubMed] [Google Scholar]
- Di Palma J.A., Cleveland M.V., McGowan J., Herrera J.L. (2007) A randomized, multicenter comparison of polyethylene glycol laxative and tegaserod in treatment of patients with chronic constipation. Am J Gastroenterol 102: 1964–1971 [DOI] [PubMed] [Google Scholar]
- Drossman D.A., Li Z., Andruzzi E., Temple R.D., Talley N.J., Thompson W.G., et al. (1993) US householder survey of functional gastrointestinal disorders. Prevalence, sociodemography and health impact. Dig Dis Sci 38: 1569–1580 [DOI] [PubMed] [Google Scholar]
- Drossman D.A., Chey W.D., Johanson J.F., Fass R., Scott C., Panas R., et al. (2009) Clinical trial: lubiprostone in patients with constipation-associated irritable bowel syndrome – results of two randomized, placebo-controlled studies. Aliment Pharmacol Ther 29: 329–341 [DOI] [PubMed] [Google Scholar]
- Dubois D., Gilet H., Viala-Danten M., Tack J. (2010) Psychometric performance and clinical meaningfulness of the Patient Assessment of Constipation-Quality of Life questionnaire in prucalopride (RESOLOR) trials for chronic constipation. Neurogastroenterol Motil 22: e54–e63 [DOI] [PubMed] [Google Scholar]
- Emmanuel A.V., Kamm M.A., Roy A.J., Antonelli K. (1998) Effect of a novel prokinetic drug, R093877, on gastrointestinal transit in healthy volunteers. Gut 42: 511–516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Emmanuel A.V., Roy A.J., Nicholls T.J., Kamm M.A. (2002) Prucalopride, a systemic enterokinetic, for the treatment of constipation. Aliment Pharmacol Ther 16: 1347–1356 [DOI] [PubMed] [Google Scholar]
- Emmanuel A.V., Tack J., Quigley E.M., Talley N.J. (2009) Pharmacological management of constipation. Neurogastroenterol Motil 21(Suppl. 2): 41–54 [DOI] [PubMed] [Google Scholar]
- Johanson J.F., Drossman D.A., Panas R., Wahle A., Ueno R. (2008a) Clinical trial: phase 2 study of lubiprostone for irritable bowel syndrome with constipation. Aliment Pharmacol Ther 27: 685–696 [DOI] [PubMed] [Google Scholar]
- Johanson J.F., Morton D., Geenen J., Ueno R. (2008b) Multicenter, 4-week, double-blind, randomized, placebo-controlled trial of lubiprostone, a locally-acting type-2 chloride channel activator, in patients with chronic constipation. Am J Gastroenterol 103: 170–177 [DOI] [PubMed] [Google Scholar]
- Johnston J.M., Kurtz C.B., Macdougall J.E., Lavins B.J., Currie M.G., Fitch D.A., et al. (2010) Linaclotide improves abdominal pain and bowel habits in a phase IIb study of patients with irritable bowel syndrome with constipation. Gastroenterology 139: 1877–1886 [DOI] [PubMed] [Google Scholar]
- Krogh K., Jensen M.B., Gandrup P., Laurberg S., Nilsson J., Kerstens R., et al. (2002) Efficacy and tolerability of prucalopride in patients with constipation due to spinal cord injury. Scand J Gastroenterol 37: 431–436 [DOI] [PubMed] [Google Scholar]
- Lembo A.J., Kurtz C.B., Macdougall J.E., Lavins B.J., Currie M.G., Fitch D.A., et al. (2010) Efficacy of linaclotide for patients with chronic constipation. Gastroenterology 138: 886–895 [DOI] [PubMed] [Google Scholar]
- Lin H.C., Prather C., Fisher R.S., Meyer J.H., Summers R.W., Pimentel M., et al. (2005) Measurement of gastrointestinal transit. Dig Dis Sci 50: 989–1004 [DOI] [PubMed] [Google Scholar]
- Lindberg G., Hamid S., Malfertheiner P., Thomsen O., Bustos Fernandez L., Garisch J., et al. (2011) World Gastroenterology Organisation Global Guidelines. Constipation: a global perspective. http://www.worldgastroenterology.org/assets/export/userfiles/05_constipation.pdf (accessed 11 July 2011). [DOI] [PubMed]
- Longstreth G.F., Thompson W.G., Chey W.D., Houghton L.A., Mearin F., Spiller R.C. (2006) Functional bowel disorders. Gastroenterology 130: 1480–1491 [DOI] [PubMed] [Google Scholar]
- McKee D.P., Quigley E.M.M. (1993) Intestinal motility in irritable bowel syndrome: is IBS a motility disorder? Part 1: Definition of IBS and colonic motility. Dig Dis Sci 38: 1761–1772 [DOI] [PubMed] [Google Scholar]
- Müller-Lissner S., Rykx A., Kerstens R., Vandeplassche L. (2010) A double-blind, placebo-controlled study of prucalopride in elderly patients with chronic constipation. Neurogastroenterol Motil 22: 991–998 [DOI] [PubMed] [Google Scholar]
- Odunsi S.T., Camilleri M. (2009) Selected interventions in nuclear medicine: gastrointestinal motor functions. Semin Nucl Med 39: 186–194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poen A.C., Felt-Bersma R.J., Van Dongen P.A., Meuwissen S.G. (1999) Effect of prucalopride, a new enterokinetic agent, on gastrointestinal transit and anorectal function in healthy volunteers. Aliment Pharmacol Ther 13: 1493–1497 [DOI] [PubMed] [Google Scholar]
- Potet F., Bouyssou T., Escande D., Baro I. (2001) Gastrointestinal prokinetic drugs have different affinity for the human cardiac human ether-a-gogo K(+) channel. J Pharmacol Exp Ther 299: 1007–1012 [PubMed] [Google Scholar]
- Quigley E.M. (2003) Current concepts of the irritable bowel syndrome. Scand J Gastroenterol Suppl 237: 1–8 [DOI] [PubMed] [Google Scholar]
- Quigley E.M.M. (2011) Cisapride. What can we learn from the rise and fall of a prokinetic?. J Dig Dis 12: 147–156 [DOI] [PubMed] [Google Scholar]
- Quigley E.M.M., Bergmann J.-F., Bytzer P., Fried M., Hunt R., Hungin A.P.S., et al. (2007) An evidence-based approach to the management of irritable bowel syndrome in Europe. Eur J Gastroenterol Hepatol 19(Suppl 1): S1–S3717325545 [Google Scholar]
- Quigley E.M., Vandeplassche L., Kerstens R., Ausma J. (2009) 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 29: 315–328 [DOI] [PubMed] [Google Scholar]
- Sanger G.J., Quigley E.M.M. (2010) Constipation, IBS and the 5-HT4 receptor: what role for prucalopride?. Clin Med Insights: Gastroenterology 3: 21–33 [Google Scholar]
- Sloots C.E., Poen A.C., Kerstens R., Stevens M., De Pauw M., Van Oene J.C., et al. (2002) Effects of prucalopride on colonic transit, anorectal function and bowel habits in patients with chronic constipation. Aliment Pharmacol Ther 16: 759–767 [DOI] [PubMed] [Google Scholar]
- Sloots C.E., Rykx A., Cools M., Kerstens R., De Pauw M. (2010) Efficacy and safety of prucalopride in patients with chronic noncancer pain suffering from opioid-induced constipation. Dig Dis Sci 55: 2912–2921 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sonnenberg A., Koch T.R. (1989) Physician visits in the United States for constipation: 1958 to 1986. Dig Dis Sci 34: 606–611 [DOI] [PubMed] [Google Scholar]
- Tack J., Ausma J., Kerstens R., Vandeplassche L. (2008) Safety and tolerability of prucalopride (Resolor®) in patients with chronic constipation: pooled data from three pivotal phase III studies [abstract]. Gastroenterology 134: A530–A531 [Google Scholar]
- Tack J., Müller-Lissner S., Stanghellini V., Boeckxstaens G., Kamm M.A., Simren M., et al. (2011) Diagnosis and treatment of chronic constipation – a European perspective. Neurogastroenterol Motil 23: 697–710 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tack J., van Outryve M., Beyens G., Kerstens R., Vandeplassche L. (2009) Prucalopride (Resolor) in the treatment of severe chronic constipation in patients dissatisfied with laxatives. Gut 58: 357–365 [DOI] [PubMed] [Google Scholar]
- Talley N.J., O’Keefe E.A., Zinsmeister A.R., Melton L.J., 3rd (1992) Prevalence of gastrointestinal symptoms in the elderly: a population-based study. Gastroenterology 102: 895–901 [DOI] [PubMed] [Google Scholar]
- von der Ohe M.R., Camilleri M. (1992) Measurement of small bowel and colonic transit: indications and methods. Mayo Clin Proc 67: 1169–1179 [DOI] [PubMed] [Google Scholar]