205
Views
15
CrossRef citations to date
0
Altmetric
Reviews

Emerging drugs for chronic constipation

, PhD, , MS MD, , MD & , MD
Pages 493-504 | Published online: 04 Aug 2009

Bibliography

  • Higgins PD, Johanson JF. Epidemiology of constipation in North America: a systematic review. Am J Gastroenterol 2004;99:750-9
  • American college of gastroenterology chronic constipation task force. An evidence-based approach tothe management of chronic constipation in North America. Am J Gastroenterol 2005;100(Suppl 1):S1-4
  • Talley NJ, Fleming KC, Evans JM, et al. Constipation in an elderly community: a study of prevalence and potential risk factors. Am J Gastroenterol 1996;91:19-25
  • Johanson JF, Sonnenberg A, Koch TR. Clinical epidemiology of chronic constipation. J Clin Gastroenterol 1989;11:525-36
  • Harari D, Gurwitz JH, Minaker KL. Constipation in the elderly. J Am Geriatr Soc 1993;41:1130-40
  • Harris L, Chang L. The functional bowel spectrum. In: Clauw. DJWDJ, editor, Textbook of Fibromyalgia and Other Non-Neuropathic Pain Syndromes. Philadelphia, Pa: Lippincott, Williams & Wilkins, 2005
  • Crowell MD. Pathogenesis of slow transit and pelvic floor dysfunction: from bench to bedside. Rev Gastroenterol Disord 2004;4(Suppl 2):S17-27
  • Gill KP, Chia YW, Henry MM, et al. Defecography in multiple sclerosis patients with severe constipation. Radiology 1994;191:553-6
  • Bassotti G, Maggio D, Battaglia E, et al. Manometric investigation of anorectal function in early and late stage Parkinson's disease. J Neurol Neurosurg Psychiatry 2000;68:768-70
  • Klauser AG, Voderholzer WA, Heinrich CA, et al. Behavioral modification of colonic function. Can constipation be learned? Dig Dis Sci 1990;35:1271-5
  • Heymen S, Scarlett Y, Jones K, et al. Randomized, controlled trial shows biofeedback to be superior to alternative treatments for patients with pelvic floor dyssynergia-type constipation. Dis Colon Rectum 2007;50:428-41
  • Chiarioni G, Heymen S, Whitehead WE. Biofeedback therapy for dyssynergic defecation. World J Gastroenterol 2006;12:7069-74
  • Choung RS, Locke GR 3rd, Zinsmeister AR, et al. Epidemiology of slow and fast colonic transit using a scale of stool form in a community. Aliment Pharmacol Ther 2007;26:1043-50
  • Yu CS, Kim HC, Hong HK, et al. Evaluation of myenteric ganglion cells and interstitial cells of Cajal in patients with chronic idiopathic constipation. Int J Colorectal Dis 2002;17:253-8
  • Bjornsson ES, Chey WD, Hooper F, et al. Impaired gastrocolonic response and peristaltic reflex in slow-transit constipation: role of 5-HT(3) pathways. Am J Physiol 2002;283:G400-7
  • Mitolo-Chieppa D, Mansi G, Rinaldi R, et al. Cholinergic stimulation and nonadrenergic, noncholinergic relaxation of human colonic circular muscle in idiopathic chronic constipation. Dig Dis Sci 1998;43:2719-26
  • Lyford GL, He CL, Soffer E, et al. Pan-colonic decrease in interstitial cells of Cajal in patients with slow transit constipation. Gut 2002;51:496-501
  • Longstreth GF, Thompson WG, Chey WD, et al. Functional bowel disorders. Gastroenterology 2006;130:1480-91
  • Brandt LJ, Prather CM, Quigley EM, et al. Systematic review on the management of chronic constipation in North America. Am J Gastroenterol 2005;100(Supp l):S5-21
  • Ramkumar D, Rao SS. Efficacy and safety of traditional medical therapies for chronic constipation: systematic review. Am J Gastroenterol 2005;100:936-71
  • Di Palma JA, Cleveland MV, McGowan J, et al. A randomized, multicenter, placebo-controlled trial of polyethylene glycol laxative for chronic treatment of chronic constipation. Am J Gastroenterol 2007;102:1436-41
  • Di Palma JA, Cleveland MV, McGowan J, et al. A randomized, multicenter comparison of polyethylene glycol laxative and tegaserod in treatment of patients with chronic constipation. Am J Gastroenterol 2007;102:1964-71
  • Nurko S, Youssef NN, Sabri M, et al. PEG3350 in the treatment of childhood constipation: a multicenter, double-blinded, placebo-controlled trial. J Pediatr 2008;153:254-61, 61 e1
  • Whitehead WE, Levy RL, Von Korff M, et al. The usual medical care for irritable bowel syndrome. Aliment Pharmacol Ther 2004;20:1305-15
  • Bueno L, Fioramonti J, Delvaux M, et al. Mediators and pharmacology of visceral sensitivity: from basic to clinical investigations. Gastroenterology 1997;112:1714-43
  • Goyal RK, Hirano I. The enteric nervous system. New Engl J Med 1996;334:1106-15
  • Gershon MD. The enteric nervous system: a second brain. Hosp Practice (1995) 1999;34:31-2, 5-8, 41-2 passim
  • Lesurtel M, Soll C, Graf R, et al. Role of serotonin in the hepato-gastroIntestinal tract: an old molecule for new perspectives. Cell Mol Life Sci 2008;65:940-52
  • Talley NJ. Review article: 5-hydroxytryptamine agonists and antagonists in the modulation of gastrointestinal motility and sensation: clinical implications. Aliment Pharmacol Ther 1992;6:273-89
  • Camilleri M. Review article: tegaserod. Aliment Pharmacol Ther 2001;15:277-89
  • Prather CM, Camilleri M, Zinsmeister AR, et al. Tegaserod accelerates orocecal transit in patients with constipation-predominant irritable bowel syndrome. Gastroenterology 2000;118:463-8
  • Degen L, Matzinger D, Merz M, et al. Tegaserod, a 5-HT4 receptor partial agonist, accelerates gastric emptying and gastrointestinal transit in healthy male subjects. Aliment Pharmacol Ther 2001;15:1745-51
  • Kamm MA, Muller-Lissner S, Talley NJ, et al. Tegaserod for the treatment of chronic constipation: a randomized, double-blind, placebo-controlled multinational study. Am J Gastroenterol 2005;100:362-72
  • Johanson JF, Wald A, Tougas G, et al. Effect of tegaserod in chronic constipation: a randomized, double-blind, controlled trial. Clin Gastroenterol Hepatol 2004;2:796-805
  • Physicians' Desk Reference, 58th edition Montvale, NJ: Thomson PDR, 2004
  • Wooltorton E. Tegaserod (Zelnorm) for irritable bowel syndrome: reports of serious diarrhea and intestinal ischemia. CMAJ 2004;170:1908
  • Tegaserod: withdrawal from the world market. A treatment for constipation with cardiovascular adverse effects. Prescrire Int 2008;17:112-3
  • Camilleri M, Kerstens R, Rykx A, et al. A placebo-controlled trial of prucalopride for severe chronic constipation. New Engl J Med 2008;358:2344-54
  • 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;58:357-65
  • 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;29:315-28
  • Movetis. Available from: http:// www.movetis.com/product/intro, [Accessed 11 May 2009]
  • Beattie DT, Armstrong SR, Shaw JP, et al. The in vivo gastrointestinal activity of TD-5108, a selective 5-HT(4) receptor agonist with high intrinsic activity. Naunyn-Schmiedeberg's Arch Pharmacol 2008;378:139-47
  • Theravance. Available from: http:// investor.theravance.com/releasedetail.cfm?ReleaseID=250925
  • ARYx. Available from: http:// www.aryx.com/wt/page/ati7505
  • 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;19:30-8
  • Dennis DPM, Irwin, I Druzgala, P Teichman. ATI-7505 is a novel, selective 5HT(4) receptor agonist that causes gastrointestinal prokinetic activity in dogs. Gastroenterology 2004;126:A641
  • Liu Z, Sakakibara R, Odaka T, et al. Mosapride citrate, a novel 5-HT4 agonist and partial 5-HT3 antagonist, ameliorates constipation in parkinsonian patients. Mov Disord 2005;20:680-6
  • Curran MP, Robinson DM. Mosapride in gastrointestinal disorders. Drugs 2008;68:981-91
  • Camilleri M, McKinzie S, Fox J, et al. Effect of renzapride on transit in constipation-predominant irritable bowel syndrome. Clin Gastroenterol Hepatol 2004;2:895-904
  • George AM, Meyers NL, Hickling RI. Clinical trial: renzapride therapy for constipation-predominant irritable bowel syndrome–multicentre, randomized, placebo-controlled, double-blind study in primary healthcare setting. Aliment Pharmacol Ther 2008;27:830-7
  • Spiller RC, Meyers NL, Hickling RI. Identification of patients with non-d, non-C irritable bowel syndrome and treatment with renzapride: an exploratory, multicenter, randomized, double-blind, placebo-controlled clinical trial. Dig Diseases Sci 2008;53:3191-200
  • Alizyme. Available from: http://ww7.investorrelations.co.uk/alizyme/investors/regnews/item.jsp?ric=AZM.L&ref=24061
  • Evangelista S. Drug evaluation: Pumosetrag for the treatment of irritable bowel syndrome and gastroesophageal reflux disease. Curr Opin Investig Drugs 2007;8:416-22
  • Fujita T, Yokota S, Sawada M, et al. Effect of MKC-733, a 5-HT receptor partial agonist, on bowel motility and symptoms in subjects with constipation: an exploratory study. J Clin Pharm Ther 2005;30:611-22
  • Feighner SD, Tan CP, McKee KK, et al. Receptor for motilin identified in the human gastrointestinal system. Science(NY) 1999;284:2184-8
  • Weber FH Jr, Richards RD, McCallum RW. Erythromycin: a motilin agonist and gastrointestinal prokinetic agent. Am J Gastroenterol 1993;88:485-90
  • Van Assche G, DepoortereI, Thijs T, et al. Contractile effects and intracellular Ca2+ signalling induced by motilin and erythromycin in the circular smooth muscle of human colon. Neurogastroenterol Motil 2001;13:27-35
  • Peeters TL. GM-611 (Chugai Pharmaceutical). Curr Opin Investig Drugs 2001;2:555-7
  • Ozaki K, Sudo H, Muramatsu H, et al. Mitemcinal (GM-611), an orally active motilin receptor agonist, accelerates colonic motility and bowel movement in conscious dogs. Inflammopharmacology 2007;15:36-42
  • Sudo H, Ozaki K, Muramatsu H, et al. Mitemcinal (GM-611), an orally active motilin agonist, facilitates defecation in rabbits and dogs without causing loose stools. Neurogastroenterol Motil 2007;19:318-26
  • Lipecka J, Bali M, Thomas A, et al. Distribution of ClC-2 chloride channel in rat and human epithelial tissues. Am J Physiol Cell Physiol 2002;282:C805-16
  • Vandewalle A. Expression and function of CLC and cystic fibrosis transmembrane conductance regulator chloride channels in renal epithelial tubule cells: pathophysiological implications. Chang Gung Med J 2007;30:17-25
  • Cuppoletti J, Malinowska DH, Tewari KP, et al. SPI-0211 activates T84 cell chloride transport and recombinant human ClC-2 chloride currents. Am J Physiol Cell Physiol 2004;287:C1173-83
  • Catalan M, Niemeyer MI, Cid LP, et al. Basolateral ClC-2 chloride channels in surface colon epithelium: regulation by a direct effect of intracellular chloride. Gastroenterology 2004;126:1104-14
  • Bijvelds MJ, Bot AG, Escher JC, et al. Activation of intestinal Cl-secretion by lubiprostone requires the cystic fibrosis transmembrane conductance regulator. Gastroenterology 2009 [Epub ahead of print - Pubmed]
  • Lubiprostone: RU 0211, SPI 0211. Drugs R D 2005;6:245-8
  • Lacy BE, Levy LC. Lubiprostone: a chloride channel activator. J Clin Gastroenterol 2007;41:345-51
  • Johanson JF, Morton D, Geenen J, et al. Multicenter, 4-week, double-blind, randomized, placebo-controlled trial of lubiprostone, a locally-acting type-2 chloride channel activator, in patients with chronic constipation. Am J Gastroenterol 2008;103:170-7
  • Drossman DA, Chey WD, Johanson JF, et al. Clinical trial: lubiprostone in patients with constipation-associated irritable bowel syndrome–results of two randomized, placebo-controlled studies. Aliment Pharmacol Ther 2009;29:329-41
  • Kuhn M. Function and dysfunction of mammalian membrane guanylyl cyclase receptors: lessons from genetic mouse models and implications for human diseases. Handb Exp Pharmacol 2009;191:47-69
  • Hasegawa M, Shimonishi Y. Recognition and signal transduction mechanism of Escherichia coli heat-stable enterotoxin and its receptor, guanylate cyclase C. J Pept Res 2005;65:261-71
  • Forte LR, Eber SL, Turner JT, et al. Guanylin stimulation of Cl-secretion in human intestinal T84 cells via cyclic guanosine monophosphate. J Clin Invest 1993;91:2423-8
  • Sellers ZM, Mann E, Smith A, et al. Heat-stable enterotoxin of Escherichia coli (STa) can stimulate duodenal HCO3(-) secretion via a novel GC-C- and CFTR-independent pathway. FASEB J 2008;22:1306-16
  • Forte LR Jr. Uroguanylin and guanylin peptides: pharmacology and experimental therapeutics. Pharmacol Ther 2004;104:137-62
  • 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;133:761-8
  • Johnston JM, Kurtz CB, Drossman DA, et al. Pilot study on the effect of linaclotide in patients with chronic constipation. Am J Gastroenterol 2009;104:125-32
  • Harris LA, Crowell MD. Linaclotide, a new direction in the treatment of irritable bowel syndrome and chronic constipation. Curr Opin Mol Ther 2007;9:403-10
  • Currie MG, Kurtz CB, Mahajan-Miklos S, et al. Effects of a single dose administration of MD-1100 on safety, tolerability, exposure, and stool consistency in healthy subjects. Am J Gastoenterol 2005;100:S328
  • Available from: http://www.synergybio.net/sp304.htm
  • Manchikanti L, Singh A. Therapeutic opioids: a ten-year perspective on the complexities and complications of the escalating use, abuse, and nonmedical use of opioids. Pain Physician 2008;11:S63-88
  • Kurz A, Sessler DI. Opioid-induced bowel dysfunction: pathophysiology and potential new therapies. Drugs 2003;63:649-71
  • Brown DR, Goldberg LI. The use of quaternary narcotic antagonists in opiate research. Neuropharmacology 1985;24:181-91
  • Saad R, Chey WD. Lubiprostone for chronic idiopathic constipation and irritable bowel syndrome with constipation. Expert Rev Gastroenterol Hepatol 2008;2:497-508
  • Peeters TL. New motilin agonists: a long and winding road. Neurogastroenterol Motil 2006;18:1-5
  • Yuan CS, Wei G, Foss JF, et al. Effects of subcutaneous methylnaltrexone on morphine-induced peripherally mediated side effects: a double-blind randomized placebo-controlled trial. J Pharmacol Exp Ther 2002;300:118-23
  • Russell J, Bass P, Goldberg LI, et al. Antagonism of gut, but not central effects of morphine with quaternary narcotic antagonists. Eur J Pharmacol 1982;78:255-61
  • Tavani A, Bianchi G, Ferretti P, et al. Morphine is most effective on gastrointestinal propulsion in rats by intraperitoneal route: evidence for local action. Life Sci 1980;27:2211-7
  • Manara L, Bianchi G, Ferretti P, et al. Inhibition of gastrointestinal transit by morphine in rats results primarily from direct drug action on gut opioid sites. J Pharmacol Exp Ther 1986;237:945-9
  • Foss JF. A review of the potential role of methylnaltrexone in opioid bowel dysfunction. Am J Surg 2001;182:19S-26S
  • Camilleri M. Alvimopan, a selective peripherally acting mu-opioid antagonist. Neurogastroenterol Motil 2005;17:157-65
  • Becker G, Blum HE. Novel opioid antagonists for opioid-induced bowel dysfunction and postoperative ileus. Lancet 2009;373:1198-206
  • Neyens R, Jackson KC. Novel opioid antagonists for opioid-induced bowel dysfunction and postoperative ileus. J Pain Palliat Care Pharmacother 2007;21:27-33
  • Yuan CS, Foss JF, O'Connor M, et al. Methylnaltrexone prevents morphine-induced delay in oral-cecal transit time without affecting analgesia: a double-blind randomized placebo-controlled trial. Clin Pharmacol Ther 1996;59:469-75
  • Murphy DB, Sutton JA, Prescott LF, et al. Opioid-induced delay in gastric emptying: a peripheral mechanism in humans. Anesthesiology 1997;87:765-70
  • Yuan CS, Foss JF, Osinski J, et al. The safety and efficacy of oral methylnaltrexone in preventing morphine-induced delay in oral-cecal transit time. Clin Pharmacol Ther 1997;61:467-75
  • Yuan CS, Foss JF. Oral methylnaltrexone for opioid-induced constipation. JAMA 2000;284:1383-4
  • Thomas J, Karver S, Cooney GA, et al. Methylnaltrexone for opioid-induced constipation in advanced illness. New Engl J Med 2008;358:2332-43
  • Viscusi E, Rathmell J, Fichera A, et al. A double-blind, randomized placebo-controlled trial of methylnaltrexone (MNTX) for post-operative bowel dysfunction in segmental colectomy. Proc Ann Soc Anesth 2005;103:A893
  • Paulson DM, Kennedy DT, Donovick RA, et al. Alvimopan: an oral, peripherally acting, mu-opioid receptor antagonist for the treatment of opioid-induced bowel dysfunction–a 21-day treatment-randomized clinical trial. J Pain 2005;6:184-92
  • Webster L, Jansen JP, Peppin J, et al. Alvimopan, a peripherally acting mu-opioid receptor (PAM-OR) antagonist for the treatment of opioid-induced bowel dysfunction: results from a randomized, double-blind, placebo-controlled, dose-finding study in subjects taking opioids for chronic non-cancer pain. Pain 2008;137:428-40
  • Buchler MW, Seiler CM, Monson JR, et al. Clinical trial: alvimopan for the management of post-operative ileus after abdominal surgery: results of an international randomized, double-blind, multicentre, placebo-controlled clinical study. Aliment Pharmacol Ther 2008;28:312-25
  • McNicol E, Boyce DB, Schumann R, et al. Efficacy and safety of mu-opioid antagonists in the treatment of opioid-induced bowel dysfunction: systematic review and meta-analysis of randomized controlled trials. Pain Med (Malden, Mass) 2008;9:634-59
  • Erowele G. Alvimopan (Entereg), a peripherally acting mu-opioid receptor antagonist for postoperative ileus. PT Drug Ther 2008;33:574-83

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.